1
|
Seiglie MP, Umemori H. Adolescent alcohol exposure disrupts episodic-like memory by impairing dopamine synapses in the mouse prelimbic cortex. Neuropharmacology 2025; 265:110255. [PMID: 39643240 DOI: 10.1016/j.neuropharm.2024.110255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/21/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Heavy alcohol use during adolescence has a significant impact on cognitive functions, such as episodic memory, even after detoxification. However, in animal models, defects in episodic memory by adolescent alcohol exposure have not been consistently replicated, and thus, the brain regions and systems that are involved remain to be elucidated. Here, we show that adolescent alcohol exposure impairs episodic memory through the impairment of the dopamine system in the prelimbic region (PrL) of the medial prefrontal cortex in both females and males. Using mice as a model, we first show that adolescent alcohol exposure disrupts episodic-like memory in female and male adult mice. We then show that adolescent alcohol exposure decreases dopaminergic presynaptic terminals in the PrL in female and male mice. This decrease persists into adulthood. Finally, we show that the adult application of a D1 dopamine receptor agonist into the PrL of adolescent alcohol-exposed mice rescues episodic-like memory in female and male mice. Together, our results identify that dopaminergic synapses in the PrL play critical roles in the effects of adolescent alcohol use on episodic memory and provide a potential strategy to reverse memory deficits caused by adolescent alcohol use in both sexes.
Collapse
Affiliation(s)
- Mariel P Seiglie
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
2
|
Zwierzyńska E, Klimczak M, Nasiadek M, Stragierowicz J, Pietrzak B. Impact of levetiracetam and ethanol on memory, selected neurotransmitter levels, oxidative stress parameters, and essential elements in rats. Pharmacol Rep 2024; 76:1363-1376. [PMID: 39352642 PMCID: PMC11582331 DOI: 10.1007/s43440-024-00659-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Ethanol disrupts brain activity and memory. There is evidence supporting the beneficial effect of levetiracetam on alcohol consumption. Therefore, the aim of the study was to examine whether levetiracetam has a protective activity against ethanol-induced memory impairment, alterations in selected neurotransmission activities, oxidative stress, and selected essential elements in rats. METHODS The rats were given levetiracetam (300 mg/kg b.w. po) with ethanol for three weeks prior to behavioral tests. Spatial memory was tested using the Morris water maze, while recognition memory was evaluated using the Novel object recognition test. The GABA and glutamate concentration was determined in three rat brain regions (cerebellum, hippocampus, and cerebral cortex). Serum oxidative stress parameters and selected essential elements concentration (Cu, Mn, Zn, Fe, Mg) in the rat brain were analyzed. RESULTS Levetiracetam administered with ethanol improved spatial memory, but did not affect abstinence-induced impairment. The drug also decreased ethanol-induced long-term recognition memory impairment. No alterations in glutamate levels were observed. GABA levels were elevated by levetiracetam in the cerebral cortex and by ethanol in the cerebellum. Ethanol increased catalase activity (CAT) and decreased superoxide dismutase activity (SOD) in the serum. Levetiracetam significantly increased the activity of SOD. Alcohol disrupted the levels of trace elements (Mn, Zn, Mg) in the rat brain. Additionally, levetiracetam alone increased Mg, Fe, and Cu concentrations while all animals receiving the drug also had significantly lower concentrations of Zn. CONCLUSIONS Levetiracetam had differential effects against ethanol-induced impairments. These findings could have important implications for future levetiracetam treatment in patients.
Collapse
Affiliation(s)
- Ewa Zwierzyńska
- Department of Pharmacodynamics, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland.
| | - Michał Klimczak
- Department of Toxicology, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland
| | - Marzenna Nasiadek
- Department of Toxicology, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland
| | - Joanna Stragierowicz
- Department of Toxicology, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland
| | - Bogusława Pietrzak
- Department of Pharmacodynamics, Medical University of Lodz, Muszyńskiego 1, Łódź, 90-151, Poland
| |
Collapse
|
3
|
Saenno R, Suwannakot K, Prajit R, Sirichoat A, Aranarochana A, Sritawan N, Pannangrong W, Wigmore P, Welbat JU. Caffeic Acid Attenuates Neuronal Apoptosis, Oxidative Stress, and Memory Deficits via Antioxidant Properties in Aging Rats Induced by D-Galactose. Mol Neurobiol 2024:10.1007/s12035-024-04610-w. [PMID: 39516391 DOI: 10.1007/s12035-024-04610-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Aging is a main factor related to cognitive deficits. D-Galactose (D-gal), a monosaccharide, increases oxidative stress leading to cellular senescence, memory deficits, and neuronal apoptosis. Caffeic acid (CA) is an antioxidant that can interrupt free radicals and reduce oxidative stress. The present study purposely evaluated the benefits of CA in attenuating loss of neuronal apoptosis, oxidative stress, and memory in D-gal-activated rat brain aging. Male Sprague-Dawley rats were arbitrarily allocated into 6 groups (9 rats per group). The D-gal group was intraperitoneal (i.p.) injected with D-gal (50 mg/kg). The CA groups were orally given 20 or 40 mg/kg CA for 8 weeks. During that time, the co-treatment groups were given 50 mg/kg of D-gal and 20 or 40 mg/kg of CA. The results reveal that animals receiving only D-gal showed memory deficit in both the novel object location (NOL) and novel object recognition (NOR) tests. Reduction in scavenging enzyme activities and levels of B-cell lymphoma 2 (Bcl-2) protein expression were detected in the D-gal group. Furthermore, D-gal treatment significantly enhanced in the number of p21 positive cells in the subgranular zone (SGZ) of the hippocampal dentate gyrus, Bcl-2 associated X protein (Bax) and caspase3 protein expression, and malondialdehyde (MDA) levels. By contrast, both 20 and 40 mg/kg CA treatment alleviated these effects. These consequences confirmed that D-gal-activated brain aging led to enhancing apoptotic protein expression including Bcl-2, Bax, and caspase3 and memory impairments. Nevertheless, CA attenuated these effects in brain aging induced by D-gal via antioxidant properties.
Collapse
Affiliation(s)
- Rasa Saenno
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kornrawee Suwannakot
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Basic Medical Science, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, 10300, Thailand
| | - Ram Prajit
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Apiwat Sirichoat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Anusara Aranarochana
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nattaya Sritawan
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Wanassanun Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Peter Wigmore
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2RD, UK
| | - Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
4
|
Mei F, Zhao C, Li S, Xue Z, Zhao Y, Xu Y, Ye R, You H, Yu P, Han X, Carr GV, Weinberger DR, Yang F, Lu B. Ngfr + cholinergic projection from SI/nBM to mPFC selectively regulates temporal order recognition memory. Nat Commun 2024; 15:7342. [PMID: 39187496 PMCID: PMC11347598 DOI: 10.1038/s41467-024-51707-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 08/13/2024] [Indexed: 08/28/2024] Open
Abstract
Acetylcholine regulates various cognitive functions through broad cholinergic innervation. However, specific cholinergic subpopulations, circuits and molecular mechanisms underlying recognition memory remain largely unknown. Here we show that Ngfr+ cholinergic neurons in the substantia innominate (SI)/nucleus basalis of Meynert (nBM)-medial prefrontal cortex (mPFC) circuit selectively underlies recency judgements. Loss of nerve growth factor receptor (Ngfr-/- mice) reduced the excitability of cholinergic neurons in the SI/nBM-mPFC circuit but not in the medial septum (MS)-hippocampus pathway, and impaired temporal order memory but not novel object and object location recognition. Expression of Ngfr in Ngfr-/- SI/nBM restored defected temporal order memory. Fiber photometry revealed that acetylcholine release in mPFC not only predicted object encounters but also mediated recency judgments of objects, and such acetylcholine release was absent in Ngfr-/- mPFC. Chemogenetic and optogenetic inhibition of SI/nBM projection to mPFC in ChAT-Cre mice diminished mPFC acetylcholine release and deteriorated temporal order recognition. Impaired cholinergic activity led to a depolarizing shift of GABAergic inputs to mPFC pyramidal neurons, due to disturbed KCC2-mediated chloride gradients. Finally, potentiation of acetylcholine signaling upregulated KCC2 levels, restored GABAergic driving force and rescued temporal order recognition deficits in Ngfr-/- mice. Thus, NGFR-dependent SI/nBM-mPFC cholinergic circuit underlies temporal order recognition memory.
Collapse
Affiliation(s)
- Fan Mei
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Chen Zhao
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Shangjin Li
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Zeping Xue
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- School of Basic Medicine, Capital Medical University, Beijing, China
- Laboratory of Cognitive and Behavioral Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yueyang Zhao
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yihua Xu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Rongrong Ye
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - He You
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Peng Yu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Xinyu Han
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Gregory V Carr
- Department of Pharmacology and Molecular Sciences, Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Weinberger
- Department of Pharmacology and Molecular Sciences, Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Feng Yang
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
- Laboratory of Cognitive and Behavioral Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Abdelaal SM, Abdel Rahman MM, Mahmoud LM, Rashed LA, Abd El-Galil TI, Mahmoud MM. Combined swimming with melatonin protects against behavioural deficit in cerebral ischemia-reperfusion injury induced rats associated with modulation of Mst1- MAPK -ERK signalling pathway. Arch Physiol Biochem 2024:1-16. [PMID: 39152720 DOI: 10.1080/13813455.2024.2392186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND The inconvenience of social and behavioural deficits after cerebral ischaemia reperfusion (I/R) injury is still not well documented. AIM We aimed to study the protective effect of preconditioning swimming exercise combined with melatonin against cerebral I/R induced injury. METHODOLOGY Sixty rats were allocated into 6 groups; groups I and II served as control. Groups 3,4,5,6 subjected to bilateral carotid ligation for 30 minutes (min.) followed by reperfusion. Group 3 left untreated while groups 4 and 6; underwent swimming exercise 30 min/day, five days a week for three weeks before the surgery. Groups 5 and 6 treated with melatonin 30 minutes before the operation, then, all rats in groups 4, 5,6 were subjected to I/R. After that, groups 5 and 6 treated with 2nd dose of melatonin 30 minutes after reperfusion. RESULTS Combined strategy exhibited the most neuroprotective effect through prevention of cerebral I/R induced inflammation, oxidative stress and apoptosis with subsequent improvement in socio behaviour deficits and enhanced Glial cell proliferative capacity. CONCLUSION The protective contribution of combined strategy is associated with modulation in Macrophage-stimulating 1/mitogen-activated protein kinase/extracellular signal-regulated kinase (MST1/MAPK/ERK) pathway which may explain, at least in part, its protective potential.
Collapse
Affiliation(s)
| | | | | | - Laila Ahmed Rashed
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | |
Collapse
|
6
|
Busceti CL, Di Menna L, Castaldi S, D'Errico G, Taddeucci A, Bruno V, Fornai F, Pittaluga A, Battaglia G, Nicoletti F. Adaptive Changes in Group 2 Metabotropic Glutamate Receptors Underlie the Deficit in Recognition Memory Induced by Methamphetamine in Mice. eNeuro 2024; 11:ENEURO.0523-23.2024. [PMID: 38969501 PMCID: PMC11298959 DOI: 10.1523/eneuro.0523-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 07/07/2024] Open
Abstract
Cognitive dysfunction is associated with methamphetamine use disorder (MUD). Here, we used genetic and pharmacological approaches to examine the involvement of either Group 2 metabotropic glutamate (mGlu2) or mGlu3 receptors in memory deficit induced by methamphetamine in mice. Methamphetamine treatment (1 mg/kg, i.p., once a day for 5 d followed by 7 d of withdrawal) caused an impaired performance in the novel object recognition test in wild-type mice, but not in mGlu2-/- or mGlu3-/- mice. Memory deficit in wild-type mice challenged with methamphetamine was corrected by systemic treatment with selectively negative allosteric modulators of mGlu2 or mGlu3 receptors (compounds VU6001966 and VU0650786, respectively). Methamphetamine treatment in wild-type mice caused large increases in levels of mGlu2/3 receptors, the Type 3 activator of G-protein signaling (AGS3), Rab3A, and the vesicular glutamate transporter, vGlut1, in the prefrontal cortex (PFC). Methamphetamine did not alter mGlu2/3-mediated inhibition of cAMP formation but abolished the ability of postsynaptic mGlu3 receptors to boost mGlu5 receptor-mediated inositol phospholipid hydrolysis in PFC slices. Remarkably, activation of presynaptic mGlu2/3 receptors did not inhibit but rather amplified depolarization-induced [3H]-D-aspartate release in synaptosomes prepared from the PFC of methamphetamine-treated mice. These findings demonstrate that exposure to methamphetamine causes changes in the expression and function of mGlu2 and mGlu3 receptors, which might alter excitatory synaptic transmission in the PFC and raise the attractive possibility that selective inhibitors of mGlu2 or mGlu3 receptors (or both) may be used to improve cognitive dysfunction in individuals affected by MUD.
Collapse
Affiliation(s)
| | - Luisa Di Menna
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
| | - Sonia Castaldi
- Department of Physiology and Pharmacology, University Sapienza, Roma 00185, Italy
| | - Giovanna D'Errico
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
| | - Alice Taddeucci
- Department of Pharmacy, University of Genova, Genova 16148, Italy
| | - Valeria Bruno
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
- Department of Physiology and Pharmacology, University Sapienza, Roma 00185, Italy
| | - Francesco Fornai
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy
| | - Anna Pittaluga
- Department of Pharmacy, University of Genova, Genova 16148, Italy
- IRCCS Ospedale Policlinico San Martino, Genova 16145, Italy
| | - Giuseppe Battaglia
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
- Department of Physiology and Pharmacology, University Sapienza, Roma 00185, Italy
| | - Ferdinando Nicoletti
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
- Department of Physiology and Pharmacology, University Sapienza, Roma 00185, Italy
| |
Collapse
|
7
|
Ng THJ, Sarikahya MH, Hudson R, Szkudlarek HJ, Pérez-Valenzuela E, Uzuneser TC, Proud E, Gummerson D, Youssef M, Machado M, Zhaksylyk K, DeVuono MV, Chen C, Yeung KKC, Rushlow WJ, Laviolette SR. Adolescent nicotine exposure induces long-term, sex-specific disturbances in mood and anxiety-related behavioral, neuronal and molecular phenotypes in the mesocorticolimbic system. Neuropsychopharmacology 2024; 49:1171-1182. [PMID: 38521861 PMCID: PMC11109238 DOI: 10.1038/s41386-024-01853-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
The majority of lifetime smokers begin using nicotine during adolescence, a critical period of brain development wherein neural circuits critical for mood, affect and cognition are vulnerable to drug-related insults. Specifically, brain regions such as the medial prefrontal cortex (mPFC), the ventral tegmental area (VTA), nucleus accumbens (NAc) and hippocampus, are implicated in both nicotine dependence and pathological phenotypes linked to mood and anxiety disorders. Clinical studies report that females experience higher rates of mood/anxiety disorders and are more resistant to smoking cessation therapies, suggesting potential sex-specific responses to nicotine exposure and later-life neuropsychiatric risk. However, the potential neural and molecular mechanisms underlying such sex differences are not clear. In the present study, we compared the impacts of adolescent nicotine exposure in male vs. female rat cohorts. We performed a combination of behavioral, electrophysiological and targeted protein expression analyses along with matrix assisted laser deionization imaging (MALDI) immediately post-adolescent exposure and later in early adulthood. We report that adolescent nicotine exposure induced long-lasting anxiety/depressive-like behaviors, disrupted neuronal activity patterns in the mPFC-VTA network and molecular alterations in various neural regions linked to affect, anxiety and cognition. Remarkably, these phenotypes were only observed in males and/or were expressed in the opposite direction in females. These findings identify a series of novel, sex-selective biomarkers for adolescent nicotine-induced neuropsychiatric risk, persisting into adulthood.
Collapse
Affiliation(s)
- Tsun Hay Jason Ng
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Roger Hudson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Hanna J Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Enzo Pérez-Valenzuela
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Taygun C Uzuneser
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Emma Proud
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Dana Gummerson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Miray Youssef
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Madeline Machado
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Kuralay Zhaksylyk
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Marieka V DeVuono
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Chaochao Chen
- Department of Chemistry, University of Western Ontario, London, ON, N6A 3K7, Canada
- Department of Biochemistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Ken K-C Yeung
- Department of Chemistry, University of Western Ontario, London, ON, N6A 3K7, Canada
- Department of Biochemistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Walter J Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada
- Department of Psychiatry, Western University, London, ON, N6A 3K7, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 3K7, Canada.
- Department of Psychiatry, Western University, London, ON, N6A 3K7, Canada.
- Lawson Health Research Institute, London, ON, N6C 2R5, Canada.
- Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), London, ON, Canada.
| |
Collapse
|
8
|
Schafer RM, Giancotti LA, Davis DJ, Larrea IG, Farr SA, Salvemini D. Behavioral characterization of G-protein-coupled receptor 160 knockout mice. Pain 2024; 165:1361-1371. [PMID: 38198232 PMCID: PMC11090760 DOI: 10.1097/j.pain.0000000000003136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/23/2023] [Indexed: 01/12/2024]
Abstract
ABSTRACT Neuropathic pain is a devastating condition where current therapeutics offer little to no pain relief. Novel nonnarcotic therapeutic targets are needed to address this growing medical problem. Our work identified the G-protein-coupled receptor 160 (GPR160) as a potential target for therapeutic intervention. However, the lack of small-molecule ligands for GPR160 hampers our understanding of its role in health and disease. To address this void, we generated a global Gpr160 knockout (KO) mouse using CRISPR-Cas9 genome editing technology to validate the contributions of GPR160 in nociceptive behaviors in mice. Gpr160 KO mice are healthy and fertile, with no observable physical abnormalities. Gpr160 KO mice fail to develop behavioral hypersensitivities in a model of neuropathic pain caused by constriction of the sciatic nerve. On the other hand, responses of Gpr160 KO mice in the hot-plate and tail-flick assays are not affected. We recently deorphanized GPR160 and identified cocaine- and amphetamine-regulated transcript peptide (CARTp) as a potential ligand. Using Gpr160 KO mice, we now report that the development of behavioral hypersensitivities after intrathecal or intraplantar injections of CARTp are dependent on GPR160. Cocaine- and amphetamine-regulated transcript peptide plays a role in various affective behaviors, such as anxiety, depression, and cognition. There are no differences in learning, memory, and anxiety between Gpr160 KO mice and their age-matched and sex-matched control floxed mice. Results from these studies support the pronociceptive roles of CARTp/GPR160 and GPR160 as a potential therapeutic target for treatment of neuropathic pain.
Collapse
Affiliation(s)
- Rachel M Schafer
- Department of Pharmacology and Physiology School of Medicine and Institute for Translational Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd. 63104, St. Louis, Missouri, USA
| | - Luigino A Giancotti
- Department of Pharmacology and Physiology School of Medicine and Institute for Translational Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd. 63104, St. Louis, Missouri, USA
| | - Daniel J Davis
- Animal Modeling Core, University of Missouri, Columbia, Missouri, USA
| | - Ivonne G Larrea
- Department of Pharmacology and Physiology School of Medicine and Institute for Translational Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd. 63104, St. Louis, Missouri, USA
| | - Susan A Farr
- Department of Pharmacology and Physiology School of Medicine and Institute for Translational Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd. 63104, St. Louis, Missouri, USA
- Department of Internal Medicine-Geriatrics, Saint Louis School of Medicine, St. Louis, MO, USA
- VA Medical Center, St Louis. MO 63106, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology School of Medicine and Institute for Translational Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Blvd. 63104, St. Louis, Missouri, USA
| |
Collapse
|
9
|
Shang Q, Dong YB, Xu L, Yang JH, Li JW, Yu WY, Sun J, Gao X, Huang Y, Zhang XQ. Environmental Enrichment Improves the Recognition Memory in Adult Mice Following Social Isolation via Downregulation of Kv4.2 Potassium Channels. Mol Neurobiol 2024; 61:3742-3752. [PMID: 38010561 DOI: 10.1007/s12035-023-03750-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/28/2023] [Indexed: 11/29/2023]
Abstract
Recognition memory is a cognitive process that enables us to distinguish familiar objects and situations from new items, which is essential for mammalian survival and adaptation to a changing environment. Social isolation (SI) has been implicated as a detrimental factor for recognition memory. The medial prefrontal cortex (mPFC) has been shown to carry information concerning the relative familiarity of individual stimuli, and modulating neuronal function in this region may contribute to recognition memory. The present study aimed to investigate the neuronal mechanisms in the mPFC of environmental enrichment (EE) on recognition memory in adult mice following SI. Mice were assigned into three groups: control, SI, and SI + EE groups. Novel location recognition (NLR) and novel object recognition (NOR) tests were performed to evaluate the recognition memory. The levels of Kv4 channels were assessed by qRT-PCR and western blotting. The effects of SI and SI + EE on the excitability of pyramidal neurons in the mPFC were measured using whole-cell recording. We found that SI led to a reduction in the excitability of pyramidal neurons. Specifically, we have identified that the reduction in the firing activity of pyramidal neurons resulted from alterations in the function and expression of Kv4.2 channels. Furthermore, EE regulated Kv4.2 channels, normalized the activity of pyramidal neurons, and restored the behavioral deficits following SI. Thus, the roles of Kv4.2 channels in excitability of pyramidal neurons suggest that the Kv4.2 channels present a promising therapeutic target for recognition memory impairment.
Collapse
Affiliation(s)
- Qing Shang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China
| | - Yi-Bei Dong
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Le Xu
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jian-Hong Yang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China
| | - Jia-Wen Li
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Wei-Yi Yu
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jie Sun
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China
| | - Xiang Gao
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China
| | - Yi Huang
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China
| | - Xiao-Qin Zhang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China.
- Department of Pharmacology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
10
|
Facal CL, Fernández Bessone I, Muñiz JA, Pereyra AE, Pedroncini O, Páez-Paz I, Clerici-Delville R, Arnaiz C, Urrutia L, Falasco G, Argañaraz CV, Saez T, Marin-Burgin A, Soiza-Reilly M, Falzone T, Avale ME. Tau reduction with artificial microRNAs modulates neuronal physiology and improves tauopathy phenotypes in mice. Mol Ther 2024; 32:1080-1095. [PMID: 38310353 PMCID: PMC11163272 DOI: 10.1016/j.ymthe.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/19/2023] [Accepted: 01/30/2024] [Indexed: 02/05/2024] Open
Abstract
Abnormal tau accumulation is the hallmark of several neurodegenerative diseases, named tauopathies. Strategies aimed at reducing tau in the brain are promising therapeutic interventions, yet more precise therapies would require targeting specific nuclei and neuronal subpopulations affected by disease while avoiding global reduction of physiological tau. Here, we developed artificial microRNAs directed against the human MAPT mRNA to dwindle tau protein by engaging the endogenous RNA interference pathway. In human differentiated neurons in culture, microRNA-mediated tau reduction diminished neuronal firing without affecting neuronal morphology or impairing axonal transport. In the htau mouse model of tauopathy, we locally expressed artificial microRNAs in the prefrontal cortex (PFC), an area particularly vulnerable to initiating tau pathology in this model. Tau knockdown prevented the accumulation of insoluble and hyperphosphorylated tau, modulated firing activity of putative pyramidal neurons, and improved glucose uptake in the PFC. Moreover, such tau reduction prevented cognitive decline in aged htau mice. Our results suggest target engagement of designed tau-microRNAs to effectively reduce tau pathology, providing a proof of concept for a potential therapeutic approach based on local tau knockdown to rescue tauopathy-related phenotypes.
Collapse
Affiliation(s)
- Carolina Lucía Facal
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - Iván Fernández Bessone
- Instituto de Biología Celular y Neurociencias (IBCN), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina
| | - Javier Andrés Muñiz
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - A Ezequiel Pereyra
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - Olivia Pedroncini
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Partner Institute of the Max Planck Society, CONICET-MPSP, Buenos Aires, Argentina
| | - Indiana Páez-Paz
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - Ramiro Clerici-Delville
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina
| | - Cayetana Arnaiz
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Partner Institute of the Max Planck Society, CONICET-MPSP, Buenos Aires, Argentina
| | - Leandro Urrutia
- Centro de imágenes Moleculares, FLENI, Buenos Aires, Argentina
| | - Germán Falasco
- Centro de imágenes Moleculares, FLENI, Buenos Aires, Argentina
| | - Carla Verónica Argañaraz
- Instituto de Fisiología Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina
| | - Trinidad Saez
- Instituto de Biología Celular y Neurociencias (IBCN), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina
| | - Antonia Marin-Burgin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Partner Institute of the Max Planck Society, CONICET-MPSP, Buenos Aires, Argentina
| | - Mariano Soiza-Reilly
- Instituto de Fisiología Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina
| | - Tomás Falzone
- Instituto de Biología Celular y Neurociencias (IBCN), Universidad de Buenos Aires, CONICET-UBA, Buenos Aires, Argentina; Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), Partner Institute of the Max Planck Society, CONICET-MPSP, Buenos Aires, Argentina
| | - María Elena Avale
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), CONICET, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Masmudi-Martín M, López-Aranda MF, Navarro-Lobato I, Khan ZU. A role of frontal association cortex in long-term object recognition memory of objects with complex features in rats. Eur J Neurosci 2024; 59:1743-1752. [PMID: 38238909 DOI: 10.1111/ejn.16243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 11/21/2023] [Accepted: 12/13/2023] [Indexed: 04/04/2024]
Abstract
Perirhinal cortex is a brain area that has been considered crucial for the object recognition memory (ORM). However, with the use of an ORM enhancer named RGS14414 as gain-in-function tool, we show here that frontal association cortex and not the Perirhinal cortex is essential for the ORM of objects with complex features that consisted of detailed drawing on the object surface (complex ORM). An expression of RGS14414, in rat brain frontal association cortex, induced the formation of long-term complex ORM, whereas the expression of the same memory enhancer in Perirhinal cortex failed to produce this effect. Instead, RGS14414 expression in Perirhinal cortex caused the formation of ORM of objects with simple features that consisted of the shape of object (simple ORM). Further, a selective elimination of frontal association cortex neurons by treatment with an immunotoxin Ox7-SAP completely abrogated the formation of complex ORM. Thus, our results suggest that frontal association cortex plays a key role in processing of a high-order recognition memory information in brain.
Collapse
Affiliation(s)
- Mariam Masmudi-Martín
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
- Brain Metastasis Group, National Cancer Research Centre (CNIO), Madrid, Spain
| | - Manuel F López-Aranda
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
- Department of Neurobiology, University of California-Los Angeles, Los Angeles, California, USA
| | - Irene Navarro-Lobato
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
- Donders Institute for Brain Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Zafar U Khan
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
- CIBERNED, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
12
|
Hossein Geranmayeh M, Farokhi-Sisakht F, Sadigh-Eteghad S, Rahbarghazi R, Mahmoudi J, Farhoudi M. Simultaneous Pericytes and M2 Microglia Transplantation Improve Cognitive Function in Mice Model of mPFC Ischemia. Neuroscience 2023; 529:62-72. [PMID: 37591334 DOI: 10.1016/j.neuroscience.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/27/2023] [Accepted: 08/04/2023] [Indexed: 08/19/2023]
Abstract
Cerebral ischemia is one of the major problems threatening global health. Many of the cerebral ischemia survivors would suffer from the physical and cognitive disabilities for their whole lifetime. Cell based-therapies have been introduced as a therapeutic approach for alleviating ischemia-enforced limitations. Photothrombotic stroke model was applied on the left medial prefrontal cortex (mPFC) of adult male BALB/c mice. Then, pericytes isolated from brain microvessels of adult male BALB/c mice, microglia isolated from brain cortices of the neonatal male BALB/c mice, and M2 phenotype shifted microglia by IL-4 treatment were used for transplantation into the injured area after 24 h of ischemia induction. The behavioural outcomes evaluated by social interaction and Barnes tests and the levels of growth associated protein (GAP)-43 and inflammatory cytokine interleukin (IL)-1 protein were assessed by western blotting 7 days after cell transplantation. Animals in both of the microglia + pericytes and microglia M2 + pericytes transplanted groups showed better performance in social memory as well as enhanced spatial learning and memory compared to ischemic controls. Also, improved escape latency was only observed in microglia M2 + pericytes (p < 0.01) group compared to ischemic controls. GAP-43 showed significant protein expression in microglia + pericytes and microglia M2 + pericytes groups compared to the control group. Conversely, IL-1 levels diminished in all of the pericytes microglia + pericytes, and microglia M2 + pericytes groups compared to the ischemic controls. Current study highlights efficiency of M2 microglia and pericytes combinatory transplantation therapeutic role on relieving ischemic stroke outcomes.
Collapse
Affiliation(s)
- Mohammad Hossein Geranmayeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Martin M, Pusceddu MM, Teichenné J, Negra T, Connolly A, Escoté X, Torrell Galceran H, Cereto Massagué A, Samarra Mestre I, Del Pino Rius A, Romero-Gimenez J, Egea C, Alcaide-Hidalgo JM, Del Bas JM. Preventive Treatment with Astaxanthin Microencapsulated with Spirulina Powder, Administered in a Dose Range Equivalent to Human Consumption, Prevents LPS-Induced Cognitive Impairment in Rats. Nutrients 2023; 15:2854. [PMID: 37447181 DOI: 10.3390/nu15132854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Cognitive alterations are a common feature associated with many neurodegenerative diseases and are considered a major health concern worldwide. Cognitive alterations are triggered by microglia activation and oxidative/inflammatory processes in specific areas of the central nervous system. Consumption of bioactive compounds with antioxidative and anti-inflammatory effects, such as astaxanthin and spirulina, can help in preventing the development of these pathologies. In this study, we have investigated the potential beneficial neuroprotective effects of a low dose of astaxanthin (ASX) microencapsulated within spirulina (ASXSP) in female rats to prevent the cognitive deficits associated with the administration of LPS. Alterations in memory processing were evaluated in the Y-Maze and Morris Water Maze (MWM) paradigms. Changes in microglia activation and in gut microbiota content were also investigated. Our results demonstrate that LPS modified long-term memory in the MWM and increased microglia activation in the hippocampus and prefrontal cortex. Preventive treatment with ASXSP ameliorated LPS-cognitive alterations and microglia activation in both brain regions. Moreover, ASXSP was able to partially revert LPS-induced gut dysbiosis. Our results demonstrate the neuroprotective benefits of ASX when microencapsulated with spirulina acting through different mechanisms, including antioxidant, anti-inflammatory and, probably, prebiotic actions.
Collapse
Affiliation(s)
- Miquel Martin
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Matteo M Pusceddu
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Joan Teichenné
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | | | | | - Xavier Escoté
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Helena Torrell Galceran
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Adrià Cereto Massagué
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Iris Samarra Mestre
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Antoni Del Pino Rius
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Jordi Romero-Gimenez
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Cristina Egea
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | | | - Josep Maria Del Bas
- Eurecat-Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
| |
Collapse
|
14
|
Delgado-Sallent C, Gener T, Nebot P, López-Cabezón C, Puig MV. Neural substrates of cognitive impairment in a NMDAR hypofunction mouse model of schizophrenia and partial rescue by risperidone. Front Cell Neurosci 2023; 17:1152248. [PMID: 37066076 PMCID: PMC10104169 DOI: 10.3389/fncel.2023.1152248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/10/2023] [Indexed: 04/03/2023] Open
Abstract
N-methyl D-aspartate receptor (NMDAR) hypofunction is a pathophysiological mechanism relevant for schizophrenia. Acute administration of the NMDAR antagonist phencyclidine (PCP) induces psychosis in patients and animals while subchronic PCP (sPCP) produces cognitive dysfunction for weeks. We investigated the neural correlates of memory and auditory impairments in mice treated with sPCP and the rescuing abilities of the atypical antipsychotic drug risperidone administered daily for two weeks. We recorded neural activities in the medial prefrontal cortex (mPFC) and the dorsal hippocampus (dHPC) during memory acquisition, short-term, and long-term memory in the novel object recognition test and during auditory processing and mismatch negativity (MMN) and examined the effects of sPCP and sPCP followed by risperidone. We found that the information about the familiar object and its short-term storage were associated with mPFC→dHPC high gamma connectivity (phase slope index) whereas long-term memory retrieval depended on dHPC→mPFC theta connectivity. sPCP impaired short-term and long-term memories, which were associated with increased theta power in the mPFC, decreased gamma power and theta-gamma coupling in the dHPC, and disrupted mPFC-dHPC connectivity. Risperidone rescued the memory deficits and partly restored hippocampal desynchronization but did not ameliorate mPFC and circuit connectivity alterations. sPCP also impaired auditory processing and its neural correlates (evoked potentials and MMN) in the mPFC, which were also partly rescued by risperidone. Our study suggests that the mPFC and the dHPC disconnect during NMDAR hypofunction, possibly underlying cognitive impairment in schizophrenia, and that risperidone targets this circuit to ameliorate cognitive abilities in patients.
Collapse
Affiliation(s)
- Cristina Delgado-Sallent
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, Barcelona, Spain
| | - Thomas Gener
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, Barcelona, Spain
- Catalan Institute of Nanoscience and Nanotechnology, CSIC and BIST, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pau Nebot
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, Barcelona, Spain
| | - Cristina López-Cabezón
- Catalan Institute of Nanoscience and Nanotechnology, CSIC and BIST, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M. Victoria Puig
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, Barcelona, Spain
- Catalan Institute of Nanoscience and Nanotechnology, CSIC and BIST, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- *Correspondence: M. Victoria Puig,
| |
Collapse
|
15
|
Osorio-Gómez D, Miranda MI, Guzmán-Ramos K, Bermúdez-Rattoni F. Transforming experiences: Neurobiology of memory updating/editing. Front Syst Neurosci 2023; 17:1103770. [PMID: 36896148 PMCID: PMC9989287 DOI: 10.3389/fnsys.2023.1103770] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Long-term memory is achieved through a consolidation process where structural and molecular changes integrate information into a stable memory. However, environmental conditions constantly change, and organisms must adapt their behavior by updating their memories, providing dynamic flexibility for adaptive responses. Consequently, novel stimulation/experiences can be integrated during memory retrieval; where consolidated memories are updated by a dynamic process after the appearance of a prediction error or by the exposure to new information, generating edited memories. This review will discuss the neurobiological systems involved in memory updating including recognition memory and emotional memories. In this regard, we will review the salient and emotional experiences that promote the gradual shifting from displeasure to pleasure (or vice versa), leading to hedonic or aversive responses, throughout memory updating. Finally, we will discuss evidence regarding memory updating and its potential clinical implication in drug addiction, phobias, and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Daniel Osorio-Gómez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Maria Isabel Miranda
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | - Kioko Guzmán-Ramos
- División de Ciencias Biológicas y de la Salud, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Lerma de Villada, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
16
|
Luessen DJ, Gallinger IM, Ferranti AS, Foster DJ, Melancon BJ, Lindsley CW, Niswender CM, Conn PJ. mGlu 1-mediated restoration of prefrontal cortex inhibitory signaling reverses social and cognitive deficits in an NMDA hypofunction model in mice. Neuropsychopharmacology 2022; 47:1826-1835. [PMID: 35643819 PMCID: PMC9372079 DOI: 10.1038/s41386-022-01350-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/08/2022]
Abstract
Extensive evidence supports the hypothesis that deficits in inhibitory GABA transmission in the prefrontal cortex (PFC) may drive pathophysiological changes underlying symptoms of schizophrenia that are not currently treated by available medications, including cognitive and social impairments. Recently, the mGlu1 subtype of metabotropic glutamate (mGlu) receptor has been implicated as a novel target to restore GABAergic transmission in the PFC. A recent study reported that activation of mGlu1 increases inhibitory transmission in the PFC through excitation of somatostatin-expressing GABAergic interneurons, implicating mGlu1 PAMs as a potential treatment strategy for schizophrenia. Here, we leveraged positive allosteric modulators (PAMs) of mGlu1 to examine whether mGlu1 activation might reverse physiological effects and behavioral deficits induced by MK-801, an NMDA receptor antagonist commonly used to model cortical deficits observed in schizophrenia patients. Using ex vivo whole-cell patch-clamp electrophysiology, we found that MK-801 decreased the frequency of spontaneous inhibitory postsynaptic currents onto layer V pyramidal cells of the PFC and this cortical disinhibition was reversed by mGlu1 activation. Furthermore, acute MK-801 treatment selectively induced inhibitory deficits onto layer V pyramidal cells that project to the basolateral amygdala, but not to the nucleus accumbens, and these deficits were restored by selective mGlu1 activation. Importantly, the mGlu1 PAM VU6004909 effectively reversed deficits in sociability and social novelty preference in a three-chamber assay and improved novel objection recognition following MK-801 treatment. Together, these findings provide compelling evidence that mGlu1 PAMs could serve as a novel approach to reduce social and cognitive deficits associated with schizophrenia by enhancing inhibitory transmission in the PFC, thus providing an exciting improvement over current antipsychotic medication.
Collapse
Affiliation(s)
- Deborah J Luessen
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA.
| | - Isabel M Gallinger
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Anthony S Ferranti
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Daniel J Foster
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Bruce J Melancon
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
- Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA.
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
17
|
Alemany-González M, Vilademunt M, Gener T, Puig MV. Postnatal environmental enrichment enhances memory through distinct neural mechanisms in healthy and trisomic female mice. Neurobiol Dis 2022; 173:105841. [PMID: 35988873 DOI: 10.1016/j.nbd.2022.105841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022] Open
Abstract
Stimulating lifestyles have powerful effects on cognitive abilities, especially when they are experienced early in life. Cognitive therapies are widely used to improve cognitive impairment due to intellectual disability, aging, and neurodegeneration, however the underlying neural mechanisms are poorly understood. We investigated the neural correlates of memory amelioration produced by postnatal environmental enrichment (EE) in diploid mice and the Ts65Dn mouse model of Down syndrome (trisomy 21). We recorded neural activities in brain structures key for memory processing, the hippocampus and the prefrontal cortex, during rest, sleep and memory performance in mice reared in non-enriched or enriched environments. Enriched wild-type animals exhibited enhanced neural synchrony in the hippocampus across different brain states (increased gamma oscillations, theta-gamma coupling, sleep ripples). Trisomic females showed increased theta and gamma rhythms in the hippocampus and prefrontal cortex across different brain states along with enlarged ripples and disrupted circuit gamma signals that were associated with memory deficits. These pathological activities were attenuated in their trisomic EE-reared peers. Our results suggest distinct neural mechanisms for the generation and rescue of healthy and pathological brain synchrony, respectively, by EE and put forward hippocampal-prefrontal hypersynchrony and miscommunication as major targets underlying the beneficial effects of EE in intellectual disability.
Collapse
Affiliation(s)
- Maria Alemany-González
- Integrative Pharmacology and Systems Neuroscience, Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Marta Vilademunt
- Integrative Pharmacology and Systems Neuroscience, Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Thomas Gener
- Integrative Pharmacology and Systems Neuroscience, Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain; Catalan Institute of Nanoscience and Nanotechnology (ICN2), the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193 Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - M Victoria Puig
- Integrative Pharmacology and Systems Neuroscience, Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain; Catalan Institute of Nanoscience and Nanotechnology (ICN2), the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193 Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
18
|
Osorio-Gómez D, Guzmán-Ramos K, Bermúdez-Rattoni F. Dopamine activity on the perceptual salience for recognition memory. Front Behav Neurosci 2022; 16:963739. [PMID: 36275849 PMCID: PMC9583835 DOI: 10.3389/fnbeh.2022.963739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
To survive, animals must recognize relevant stimuli and distinguish them from inconspicuous information. Usually, the properties of the stimuli, such as intensity, duration, frequency, and novelty, among others, determine the salience of the stimulus. However, previously learned experiences also facilitate the perception and processing of information to establish their salience. Here, we propose “perceptual salience” to define how memory mediates the integration of inconspicuous stimuli into a relevant memory trace without apparently altering the recognition of the physical attributes or valence, enabling the detection of stimuli changes in future encounters. The sense of familiarity is essential for successful recognition memory; in general, familiarization allows the transition of labeling a stimulus from the novel (salient) to the familiar (non-salient). The novel object recognition (NOR) and object location recognition (OLRM) memory paradigms represent experimental models of recognition memory that allow us to study the neurobiological mechanisms involved in episodic memory. The catecholaminergic system has been of vital interest due to its role in several aspects of recognition memory. This review will discuss the evidence that indicates changes in dopaminergic activity during exposure to novel objects or places, promoting the consolidation and persistence of memory. We will discuss the relationship between dopaminergic activity and perceptual salience of stimuli enabling learning and consolidation processes necessary for the novel-familiar transition. Finally, we will describe the effect of dopaminergic deregulation observed in some pathologies and its impact on recognition memory.
Collapse
Affiliation(s)
- Daniel Osorio-Gómez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Mexico, Mexico
| | - Kioko Guzmán-Ramos
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Lerma, Estado de México, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Mexico, Mexico
- *Correspondence: Federico Bermúdez-Rattoni
| |
Collapse
|
19
|
Florido A, Moreno E, Canela EI, Andero R. Nk3R blockade has sex-divergent effects on memory in mice. Biol Sex Differ 2022; 13:28. [PMID: 35690790 PMCID: PMC9188709 DOI: 10.1186/s13293-022-00437-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/24/2022] [Indexed: 12/25/2022] Open
Abstract
Background Memory consolidation is a process required for the formation of long-term memories. The G-protein-coupled receptor (GPCR) neurokinin-3-receptor (Nk3R) and its interactions with sex hormones seem important for the modulation of fear memory consolidation: Nk3R antagonism in male mice impairs fear memory, but enhances it in females. However, the involvement of the Nk3R as a modulator of other memories in both sexes remains unexplored. Methods We use the novel object recognition paradigm to test the effect of a systemic blockade of Nk3R during memory consolidation. Further, we assess the expression of estrogen receptor α, estrogen receptor β, and androgen receptor and heterodimerization with Nk3R in the medial prefrontal cortex (mPFC) and dorsal hippocampus (DH) of mice. Results Nk3R systemic antagonism elicited decreased memory consolidation in males while it enhanced it in females during proestrus. Nk3R analysis in the different subregions of the mPFC and the DH showed a higher expression in males than females. Moreover, females presented upregulation of the androgen receptor in the CA1 and the estrogen receptor beta in the cingulate cortex, CA1, and dentate gyrus. Overall, males presented an upregulation of the estrogen receptor alpha. We also explored the heterodimerization of GCPR membrane sex hormone receptors with the Nk3R. We found a higher percentage of Nk3R-membrane G-protein estrogen receptors heterodimers in the prelimbic cortex of the mPFC in females, suggesting an interaction of estradiol with Nk3R in memory consolidation. However, males presented a higher percentage of Nk3R-membrane G-protein androgen receptors heterodimers compared to females, pointing to an interaction of testosterone with Nk3R in memory consolidation. Conclusion These data propose novel ideas on functional interactions between Nk3R, sex hormones, estrogen receptors, and androgen receptors in memory consolidation. Nk3R antagonism reduces recognition memory consolidation in male mice and increases it in proestrus females. Androgen receptor expression is higher in the CA1 compared to DG, CA3, and the mPFC. Estrogen repcetor α expression is higher in males than in females in the DH and mPFC. Estrogen receptor β expression is greater in females than in males in the DG, CA1, and CG. Over 60% of Nk3R in the DH and mPFC is heterodimerized with membrane estrogen receptor and androgen receptor. Nk3R–GPAR is more abundant in males than in proestrus females, whereas Nk3R–GPER is greater in proestrus females compared to males.
Collapse
Affiliation(s)
- Antonio Florido
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Estefanía Moreno
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona i Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - Enric I Canela
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona i Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028, Barcelona, Spain.,Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Raül Andero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain. .,Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain. .,Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029, Madrid, Spain. .,Unitat de Neurociència Translacional, Parc Taulí Hospital Universitari, Institut d'Investigació I Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain. .,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
20
|
McGovern HT, Leptourgos P, Hutchinson BT, Corlett PR. Do psychedelics change beliefs? Psychopharmacology (Berl) 2022; 239:1809-1821. [PMID: 35507071 DOI: 10.1007/s00213-022-06153-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/19/2022] [Indexed: 01/29/2023]
Abstract
Renewed interest in psychedelics has reignited the debate about whether and how they change human beliefs. In both the clinical and social-cognitive domains, psychedelic consumption may be accompanied by profound, and sometimes lasting, belief changes. We review these changes and their possible underlying mechanisms. Rather than inducing de novo beliefs, we argue psychedelics may instead change the impact of affect and of others' suggestions on how beliefs are imputed. Critically, we find that baseline beliefs (in the possible effects of psychedelics, for example) might color the acute effects of psychedelics as well as longer-term changes. If we are to harness the apparent potential of psychedelics in the clinic and for human flourishing more generally, these possibilities must be addressed empirically.
Collapse
Affiliation(s)
- H T McGovern
- School of Psychology, The University of Queensland, Brisbane, QLD, Australia
| | - P Leptourgos
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - B T Hutchinson
- Research School of Psychology, The Australian National University, Canberra, ACT, Australia
| | - P R Corlett
- Department of Psychiatry, Yale University, New Haven, CT, USA.
| |
Collapse
|
21
|
Saenno R, Dornlakorn O, Anosri T, Kaewngam S, Sirichoat A, Aranarochana A, Pannangrong W, Wigmore P, Welbat JU. Caffeic Acid Alleviates Memory and Hippocampal Neurogenesis Deficits in Aging Rats Induced by D-Galactose. Nutrients 2022; 14:nu14102169. [PMID: 35631310 PMCID: PMC9145046 DOI: 10.3390/nu14102169] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/21/2022] [Accepted: 05/22/2022] [Indexed: 02/04/2023] Open
Abstract
Hippocampal neurogenesis occurs throughout life, but it declines with age. D-galactose (D-gal) enhances cellular senescence through oxidative stress leading to neurodegeneration and memory impairment. Caffeic acid (CA) acts as an antioxidant via decreasing brain oxidative stress. This study aims to investigate the advantages of CA in alleviating the loss of memory and neurogenesis production in the hippocampus in aged rats activated by D-gal. Fifty-four male Sprague-Dawley rats were unpredictably arranged into six groups. In the D-gal group, rats were administered D-gal (50 mg/kg) by intraperitoneal (i.p.) injection. For the CA groups, rats received 20 or 40 mg/kg CA by oral gavage. In the co-treated groups, rats received D-gal (50 mg/kg) and CA (20 or 40 mg/kg) for eight weeks. The results of novel object location (NOL) and novel object recognition (NOR) tests showed memory deficits. Moreover, a decline of neurogenesis in the hippocampus was detected in rats that received D-gal by detecting rat endothelial cell antigen-1 (RECA-1)/Ki-67, 5-bromo-2′-deoxyuridine (BrdU)/neuronal nuclear protein (NeuN), doublecortin (DCX) by means of staining to evaluate blood vessel associated proliferating cells, neuronal cell survival and premature neurons, respectively. By contrast, CA attenuated these effects. Our results postulate that CA attenuated the impairment of memory in D-gal-stimulated aging by up-regulating levels of hippocampal neurogenesis.
Collapse
Affiliation(s)
- Rasa Saenno
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (O.D.); (T.A.); (S.K.); (A.S.); (A.A.); (W.P.)
| | - Oabnithi Dornlakorn
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (O.D.); (T.A.); (S.K.); (A.S.); (A.A.); (W.P.)
| | - Tanaporn Anosri
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (O.D.); (T.A.); (S.K.); (A.S.); (A.A.); (W.P.)
| | - Soraya Kaewngam
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (O.D.); (T.A.); (S.K.); (A.S.); (A.A.); (W.P.)
| | - Apiwat Sirichoat
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (O.D.); (T.A.); (S.K.); (A.S.); (A.A.); (W.P.)
| | - Anusara Aranarochana
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (O.D.); (T.A.); (S.K.); (A.S.); (A.A.); (W.P.)
| | - Wanassanun Pannangrong
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (O.D.); (T.A.); (S.K.); (A.S.); (A.A.); (W.P.)
| | - Peter Wigmore
- School of Life Sciences, Medical School, Queen’s Medical Centre, The University of Nottingham, Nottingham NG7 2RD, UK;
| | - Jariya Umka Welbat
- Neurogenesis Research Group, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (O.D.); (T.A.); (S.K.); (A.S.); (A.A.); (W.P.)
- Correspondence: ; Tel.: +66-84-667-6105
| |
Collapse
|
22
|
Morici JF, Cicuttin G, Silva A, Gallo FT, Miranda M, Beluscio M, Zold C, Bekinschtein P, Weisstaub NV. Serotonin Type 2a Receptor in the Prefrontal Cortex Controls Perirhinal Cortex Excitability During Object Recognition Memory Recall. Neuroscience 2022; 497:196-205. [PMID: 35597334 DOI: 10.1016/j.neuroscience.2022.05.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 10/18/2022]
Abstract
Previous experiences can drive adaptive behavior based on different characteristics, including contextual ones. Indeed, contextual information can be used as a criterion to guide the recall of the most relevant memory trace and the inhibition of others. The medial Prefontal Cortex (mPFC) has been proposed as an area that plays a pivotal role in regulating the retrieval of memory traces in downstream regions. Also, we have shown that mPFC Serotonin 2a Receptors (5-HT2aR) modulates the retrieval of a contextually guided recognition memory task and modulates the retrieval and reconsolidation of memories in the Perirhinal Cortex (PRH). However, how the mPFC output mediated by the 5-HT2aR activity is modulating memory retrieval in the PRH is a question that remains unclear. To tackle this question, we analyzed neuronal activity in the PRH and mPFC, by measuring expression of the immediate early gene c-Fos. We combined behavioral, pharmacological and immunohistochemical techniques to examine how mPFC 5-HT2aR controls mPFC and the PRH activity. We found that blockade of mPFC 5-HT2aR increase the level of c-Fos expression in the PHR and that this increase correlates with animals' performance in the task. We also found an increase in c-Fos expression in the mPFC after mPFC 5-HT2aR blockade that does not correlate with the animals' behavioral response. However, these changes showed a significant correlation with those observed in the PRH. These results suggest that mPFC 5-HT2aR signaling may modulate the behavioral response during memory recall by controlling the neuronal activation in the PRH.
Collapse
Affiliation(s)
- J F Morici
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - G Cicuttin
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - A Silva
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
| | - F T Gallo
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - M Miranda
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - M Beluscio
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
| | - C Zold
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
| | - P Bekinschtein
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - N V Weisstaub
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina.
| |
Collapse
|
23
|
Hippocampal-medial prefrontal cortex network dynamics predict performance during retrieval in a context-guided object memory task. Proc Natl Acad Sci U S A 2022; 119:e2203024119. [PMID: 35561217 DOI: 10.1073/pnas.2203024119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
SignificanceRecovering relevant information, while ignoring the irrelevant, is crucial for episodic memory (remembering a particular event at a specific temporal and spatial context). Information presented at any time could drive the retrieval of more than one memory trace; thus, there should be a mechanism to select the retrieval of the most relevant trace. However, how the brain controls memory interference is not well understood. Here, we analyzed the communication between ventral hippocampus (vHPC) and medial prefrontal cortex (mPFC) during the resolution of an episodic memory task in rats. We found an increased synchronization between the vHPC and mPFC and identified specific mPFC neural subpopulations that selectively respond to object-context associations, and their firing preference correlates with the animals' behavioral responses.
Collapse
|
24
|
Seyedaghamiri F, Hosseini L, Kazmi S, Mahmoudi J, Shanehbandi D, Ebrahimi-Kalan A, Rahbarghazi R, Sadigh-Eteghad S, Farhoudi M. Varenicline improves cognitive impairment in a mouse model of mPFC ischemia: The possible roles of inflammation, apoptosis, and synaptic factors. Brain Res Bull 2022; 181:36-45. [DOI: 10.1016/j.brainresbull.2022.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
|
25
|
Cnops V, Iyer VR, Parathy N, Wong P, Dawe GS. Test, Rinse, Repeat: A Review of Carryover Effects in Rodent Behavioral Assays. Neurosci Biobehav Rev 2022; 135:104560. [DOI: 10.1016/j.neubiorev.2022.104560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 01/21/2023]
|
26
|
Moya M, López-Valencia L, García-Bueno B, Orio L. Disinhibition-Like Behavior Correlates with Frontal Cortex Damage in an Animal Model of Chronic Alcohol Consumption and Thiamine Deficiency. Biomedicines 2022; 10:biomedicines10020260. [PMID: 35203470 PMCID: PMC8869694 DOI: 10.3390/biomedicines10020260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/08/2022] [Accepted: 01/18/2022] [Indexed: 12/23/2022] Open
Abstract
Wernicke-Korsakoff syndrome (WKS) is induced by thiamine deficiency (TD) and mainly related to alcohol consumption. Frontal cortex dysfunction has been associated with impulsivity and disinhibition in WKS patients. The pathophysiology involves oxidative stress, excitotoxicity and inflammatory responses leading to neuronal death, but the relative contributions of each factor (alcohol and TD, either isolated or in interaction) to these phenomena are still poorly understood. A rat model was used by forced consumption of 20% (w/v) alcohol for 9 months (CA), TD hit (TD diet + pyrithiamine 0.25 mg/kg, i.p. daily injections the last 12 days of experimentation (TDD)), and both combined treatments (CA+TDD). Motor and cognitive performance and cortical damage were examined. CA caused hyperlocomotion as a possible sensitization of ethanol-induced excitatory effects and recognition memory deficits. In addition, CA+TDD animals showed a disinhibited-like behavior which appeared to be dependent on TDD. Additionally, combined treatment led to more pronounced alterations in nitrosative stress, lipid peroxidation, apoptosis and cell damage markers. Correlations between injury signals and disinhibition suggest that CA+TDD disrupts behaviors dependent on the frontal cortex. Our study sheds light on the potential disease-specific mechanisms, reinforcing the need for neuroprotective therapeutic approaches along with preventive treatments for the nutritional deficiency in WKS.
Collapse
Affiliation(s)
- Marta Moya
- Department of Psychobiology and Methods in Behavioral Sciences, Faculty of Psychology, Complutense University of Madrid (UCM), 28223 Madrid, Spain; (M.M.); (L.L.-V.)
- Red de Trastornos Adictivos (RTA), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Leticia López-Valencia
- Department of Psychobiology and Methods in Behavioral Sciences, Faculty of Psychology, Complutense University of Madrid (UCM), 28223 Madrid, Spain; (M.M.); (L.L.-V.)
| | - Borja García-Bueno
- Departament of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain;
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación en Neuroquímica IUIN-UCM, Avda. Complutense s/n, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Laura Orio
- Department of Psychobiology and Methods in Behavioral Sciences, Faculty of Psychology, Complutense University of Madrid (UCM), 28223 Madrid, Spain; (M.M.); (L.L.-V.)
- Red de Trastornos Adictivos (RTA), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
27
|
Patel AV, Codeluppi SA, Ervin KSJ, St-Denis MB, Choleris E, Bailey CDC. Developmental Age and Biological Sex Influence Muscarinic Receptor Function and Neuron Morphology within Layer VI of the Medial Prefrontal Cortex. Cereb Cortex 2021; 32:3137-3158. [PMID: 34864929 DOI: 10.1093/cercor/bhab406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 01/15/2023] Open
Abstract
Acetylcholine (ACh) neurotransmission within the medial prefrontal cortex (mPFC) plays an important modulatory role to support mPFC-dependent cognitive functions. This role is mediated by ACh activation of its nicotinic (nAChR) and muscarinic (mAChR) classes of receptors, which are both present on mPFC layer VI pyramidal neurons. While the expression and function of nAChRs have been characterized thoroughly for rodent mPFC layer VI neurons during postnatal development, mAChRs have not been characterized in detail. We employed whole-cell electrophysiology with biocytin filling to demonstrate that mAChR function is greater during the juvenile period of development than in adulthood for both sexes. Pharmacological experiments suggest that each of the M1, M2, and M3 mAChR subtypes contributes to ACh responses in these neurons in a sex-dependent manner. Analysis of dendrite morphology identified effects of age more often in males, as the amount of dendrite matter was greatest during the juvenile period. Interestingly, a number of positive correlations were identified between the magnitude of ACh/mAChR responses and dendrite morphology in juvenile mice that were not present in adulthood. To our knowledge, this work describes the first detailed characterization of mAChR function and its correlation with neuron morphology within layer VI of the mPFC.
Collapse
Affiliation(s)
- Ashutosh V Patel
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Sierra A Codeluppi
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kelsy S J Ervin
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Myles B St-Denis
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Elena Choleris
- Department of Psychology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Craig D C Bailey
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
28
|
Mossad O, Nent E, Woltemate S, Folschweiller S, Buescher JM, Schnepf D, Erny D, Staeheli P, Bartos M, Szalay A, Stecher B, Vital M, Sauer JF, Lämmermann T, Prinz M, Blank T. Microbiota-dependent increase in δ-valerobetaine alters neuronal function and is responsible for age-related cognitive decline. NATURE AGING 2021; 1:1127-1136. [PMID: 37117525 DOI: 10.1038/s43587-021-00141-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 10/25/2021] [Indexed: 04/30/2023]
Abstract
Understanding the physiological origins of age-related cognitive decline is of critical importance given the rising age of the world's population1. Previous work in animal models has established a strong link between cognitive performance and the microbiota2-5, and it is known that the microbiome undergoes profound remodeling in older adults6. Despite growing evidence for the association between age-related cognitive decline and changes in the gut microbiome, the mechanisms underlying such interactions between the brain and the gut are poorly understood. Here, using fecal microbiota transplantation (FMT), we demonstrate that age-related remodeling of the gut microbiota leads to decline in cognitive function in mice and that this impairment can be rescued by transplantation of microbiota from young animals. Moreover, using a metabolomic approach, we found elevated concentrations of δ-valerobetaine, a gut microbiota-derived metabolite, in the blood and brain of aged mice and older adults. We then demonstrated that δ-valerobetaine is deleterious to learning and memory processes in mice. At the neuronal level, we showed that δ-valerobetaine modulates inhibitory synaptic transmission and neuronal network activity. Finally, we identified specific bacterial taxa that significantly correlate with δ-valerobetaine levels in the brain. Based on our findings, we propose that δ-valerobetaine contributes to microbiota-driven brain aging and that the associated mechanisms represent a promising target for countering age-related cognitive decline.
Collapse
Affiliation(s)
- Omar Mossad
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Elisa Nent
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Sabrina Woltemate
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Shani Folschweiller
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Institute of Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Daniel Schnepf
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs University Freiburg, Freiburg, Germany
| | - Daniel Erny
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Staeheli
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marlene Bartos
- Institute of Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Bärbel Stecher
- Max-von-Pettenkofer Institute, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU Munich, Munich, Germany
| | - Marius Vital
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Jonas F Sauer
- Institute of Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center for NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Blank
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
29
|
Hippocampus-Prefrontal Coupling Regulates Recognition Memory for Novelty Discrimination. J Neurosci 2021; 41:9617-9632. [PMID: 34642213 DOI: 10.1523/jneurosci.1202-21.2021] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/05/2021] [Accepted: 09/27/2021] [Indexed: 11/21/2022] Open
Abstract
Recognition memory provides the ability to distinguish familiar from novel objects and places, and is important for recording and updating events to guide appropriate behavior. The hippocampus (HPC) and medial prefrontal cortex (mPFC) have both been implicated in recognition memory, but the nature of HPC-mPFC interactions, and its impact on local circuits in mediating this process is not known. Here we show that novelty discrimination is accompanied with higher theta activity (4-10 Hz) and increased c-Fos expression in both these regions. Moreover, theta oscillations were highly coupled between the HPC and mPFC during recognition memory retrieval for novelty discrimination, with the HPC leading the mPFC, but not during initial learning. Principal neurons and interneurons in the mPFC responded more strongly during recognition memory retrieval compared with learning. Optogenetic silencing of HPC input to the mPFC disrupted coupled theta activity between these two structures, as well as the animals' (male Sprague Dawley rats) ability to differentiate novel from familiar objects. These results reveal a key role of monosynaptic connections between the HPC and mPFC in novelty discrimination via theta coupling and identify neural populations that underlie this recognition memory-guided behavior.SIGNIFICANCE STATEMENT Many memory processes are highly dependent on the interregional communication between the HPC and mPFC via neural oscillations. However, how these two brain regions coordinate their oscillatory activity to engage local neural populations to mediate recognition memory for novelty discrimination is poorly understood. This study revealed that the HPC and mPFC theta oscillations and their temporal coupling is correlated with recognition memory-guided behavior. During novel object recognition, the HPC drives mPFC interneurons to effectively reduce the activity of principal neurons. This study provides the first evidence for the requirement of the HPC-mPFC pathway to mediate recognition memory for novelty discrimination and describes a mechanism for how this memory is regulated.
Collapse
|
30
|
Sawahata M, Asano H, Nagai T, Ito N, Kohno T, Nabeshima T, Hattori M, Yamada K. Microinjection of Reelin into the mPFC prevents MK-801-induced recognition memory impairment in mice. Pharmacol Res 2021; 173:105832. [PMID: 34450306 DOI: 10.1016/j.phrs.2021.105832] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022]
Abstract
Reelin, a large extracellular matrix protein, helps to regulate neuronal plasticity and cognitive function. Several studies have shown that Reelin dysfunction, resulting from factors such as mutations in gene RELN or low Reelin expression, is associated with schizophrenia (SCZ). We previously reported that microinjection of Reelin into cerebral ventricle prevents phencyclidine-induced cognitive and sensory-motor gating deficits. However, it remains unclear whether and how Reelin ameliorates behavioral abnormalities in the animal model of SCZ. In the present study, we evaluated the effect of recombinant Reelin microinjection into the medial prefrontal cortex (mPFC) on abnormal behaviors induced by MK-801, an N-methyl-D-aspartate receptor antagonist. Microinjection of Reelin into the mPFC prevented impairment of recognition memory of MK-801-treated mice in the novel object recognition test (NORT). On the other hand, the same treatment had no effect on deficits in sensory-motor gating and short-term memory in the pre-pulse inhibition and Y-maze tests, respectively. To establish the neural substrates that respond to Reelin, the number of c-Fos-positive cells in the mPFC was determined. A significant increase in c-Fos-positive cells in the mPFC of MK-801-treated mice was observed when compared with saline-treated mice, and this change was suppressed by microinjection of Reelin into the mPFC. A K2360/2467A Reelin that cannot bind to its receptor failed to ameliorate MK-801-induced cognitive deficits in NORT. These results suggest that Reelin prevents MK-801-induced recognition memory impairment by acting on its receptors to suppress neural activity in the mPFC of mice.
Collapse
Affiliation(s)
- Masahito Sawahata
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan; Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Hiroki Asano
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan; Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake 470-1192, Japan
| | - Norimichi Ito
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, 467-8603, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Sciences, Fujita Health University, Toyoake 470-1192, Japan
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, 467-8603, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan.
| |
Collapse
|
31
|
García-Baos A, Puig-Reyne X, García-Algar Ó, Valverde O. Cannabidiol attenuates cognitive deficits and neuroinflammation induced by early alcohol exposure in a mice model. Biomed Pharmacother 2021; 141:111813. [PMID: 34126352 DOI: 10.1016/j.biopha.2021.111813] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/17/2021] [Accepted: 06/07/2021] [Indexed: 12/16/2022] Open
Abstract
Foetal alcohol spectrum disorder (FASD) is the umbrella term used to describe the physical and mental disabilities induced by alcohol exposure during development. Early alcohol exposure induces cognitive impairments resulting from damage to the central nervous system (CNS). The neuroinflammatory response accompanied by neurodegenerative mechanisms contribute to those detrimental alterations. Cannabidiol (CBD) has recently emerged as an anti-inflammatory drug that might be useful to treat several neuropsychiatric disorders. In our study, we assessed the effects of CBD on long-lasting cognitive deficits induced by early alcohol exposure. Furthermore, we analysed long-term pro-inflammatory and apoptotic markers within the prefrontal cortex and hippocampus. To model alcohol binge drinking during gestational and lactation periods, we used pregnant C57BL/6 female mice with time-limited access to 20% v/v alcohol solution. Following the prenatal and lactation alcohol exposure (PLAE), we treated the male and female offspring with CBD from post-natal day (PD) 25 until PD34, and we evaluated their cognitive performance at PD60. Our results showed that CBD treatment during peri-adolescence period ameliorates cognitive deficits observed in our FASD-like mouse model, without sex differences. Moreover, CBD restores the PLAE-induced increased levels of TNFα and IL-6 in the hippocampus. Thus, our study provides new insights for CBD as a therapeutic agent to counteract cognitive impairments and neuroinflammation caused by early alcohol exposure.
Collapse
Affiliation(s)
- Alba García-Baos
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Xavier Puig-Reyne
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Óscar García-Algar
- Neonatology Unit, ICGON, IDIBAPS, Hospital Clínic-Maternitat, BCNatal, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Programme, IMIM-Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
32
|
Kayyal H, Chandran SK, Yiannakas A, Gould N, Khamaisy M, Rosenblum K. Insula to mPFC reciprocal connectivity differentially underlies novel taste neophobic response and learning in mice. eLife 2021; 10:66686. [PMID: 34219650 PMCID: PMC8282338 DOI: 10.7554/elife.66686] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
To survive in an ever-changing environment, animals must detect and learn salient information. The anterior insular cortex (aIC) and medial prefrontal cortex (mPFC) are heavily implicated in salience and novelty processing, and specifically, the processing of taste sensory information. Here, we examined the role of aIC-mPFC reciprocal connectivity in novel taste neophobia and memory formation, in mice. Using pERK and neuronal intrinsic properties as markers for neuronal activation, and retrograde AAV (rAAV) constructs for connectivity, we demonstrate a correlation between aIC-mPFC activity and novel taste experience. Furthermore, by expressing inhibitory chemogenetic receptors in these projections, we show that aIC-to-mPFC activity is necessary for both taste neophobia and its attenuation. However, activity within mPFC-to-aIC projections is essential only for the neophobic reaction but not for the learning process. These results provide an insight into the cortical circuitry needed to detect, react to- and learn salient stimuli, a process critically involved in psychiatric disorders.
Collapse
Affiliation(s)
- Haneen Kayyal
- Sagol Department of Neuroscience, University of Haifa, Mount Carmel, Israel
| | | | - Adonis Yiannakas
- Sagol Department of Neuroscience, University of Haifa, Mount Carmel, Israel
| | - Nathaniel Gould
- Sagol Department of Neuroscience, University of Haifa, Mount Carmel, Israel
| | - Mohammad Khamaisy
- Sagol Department of Neuroscience, University of Haifa, Mount Carmel, Israel
| | - Kobi Rosenblum
- Sagol Department of Neuroscience, University of Haifa, Mount Carmel, Israel.,Center for Gene Manipulation in the Brain, University of Haifa, Mount Carmel, Israel
| |
Collapse
|
33
|
Cuervo-Zanatta D, Garcia-Mena J, Perez-Cruz C. Gut Microbiota Alterations and Cognitive Impairment Are Sexually Dissociated in a Transgenic Mice Model of Alzheimer's Disease. J Alzheimers Dis 2021; 82:S195-S214. [PMID: 33492296 DOI: 10.3233/jad-201367] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Normal aging is accompanied by cognitive deficiencies, affecting women and men equally. Aging is the main risk factor for Alzheimer's disease (AD), with women having a higher risk. The higher prevalence of AD in women is associated with the abrupt hormonal decline seen after menopause. However, other factors may be involved in this sex-related cognitive decline. Alterations in gut microbiota (GM) and its bioproducts have been reported in AD subjects and transgenic (Tg) mice, having a direct impact on brain amyloid-β pathology in male (M), but not in female (F) mice. OBJECTIVE The aim of this work was to determine GM composition and cognitive dysfunction in M and F wildtype (WT) and Tg mice, in a sex/genotype segregation design. METHODS Anxiety, short term working-memory, spatial learning, and long-term spatial memory were evaluated in 6-month-old WT and Tg male mice. Fecal short chain fatty acids were determined by chromatography, and DNA sequencing and bioinformatic analyses were used to determine GM differences. RESULTS We observed sex-dependent differences in cognitive skills in WT mice, favoring F mice. However, the cognitive advantage of females was lost in Tg mice. GM composition showed few sex-related differences in WT mice. Contrary, Tg-M mice presented a more severe dysbiosis than Tg-F mice. A decreased abundance of Ruminococcaceae was associated with cognitive deficits in Tg-F mice, while butyrate levels were positively associated with better working- and object recognition-memory in WT-F mice. CONCLUSION This report describes a sex-dependent association between GM alterations and cognitive impairment in a mice model of AD.
Collapse
Affiliation(s)
- Daniel Cuervo-Zanatta
- Pharmacology Department, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), Laboratory of Neuroplasticity and Neurodegeneration, Mexico City, Mexico.,Genetics and Molecular Biology Department, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), Laboratory of reference and support for the characterization of genomes, transcriptomes and microbiomes, Mexico City, Mexico
| | - Jaime Garcia-Mena
- Genetics and Molecular Biology Department, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), Laboratory of reference and support for the characterization of genomes, transcriptomes and microbiomes, Mexico City, Mexico
| | - Claudia Perez-Cruz
- Pharmacology Department, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), Laboratory of Neuroplasticity and Neurodegeneration, Mexico City, Mexico
| |
Collapse
|
34
|
Izumi S, Domoto M, Esaki H, Sasase H, Nishitani N, Deyama S, Kaneda K. Nicotine Enhances Firing Activity of Layer 5 Pyramidal Neurons in the Medial Prefrontal Cortex through Inhibition of Kv7 Channels. Biol Pharm Bull 2021; 44:724-731. [PMID: 33952828 DOI: 10.1248/bpb.b21-00137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nicotine enhances attention, working memory and recognition. One of the brain regions associated with these effects of nicotine is the medial prefrontal cortex (mPFC). However, cellular mechanisms that induce the enhancing effects of nicotine remain unclear. To address this issue, we performed whole-cell patch-clamp recordings from mPFC layer 5 pyramidal neurons in slices of C57BL/6J mice. Shortly (approx. 2 min) after bath application of nicotine, the number of action potentials, which were elicited by depolarizing current injection, was increased, and this increase persisted for over 5 min. The effect of nicotine was blocked by the α4β2 nicotinic acetylcholine receptor (nAChR) antagonist dihydro-β-erythroidine, α7 nAChR antagonist methyllycaconitine, or intracellular perfusion with the Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA). Additionally, the voltage-dependent potassium 7 (Kv7) channel blocker, 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE-991), as well as nicotine, shortened the spike threshold latency and increased the spike numbers. By contrast, the Kv7 channel opener, retigabine reduced the number of firings, and the addition of nicotine did not increase the spike numbers. These results indicate that nicotine induces long-lasting enhancement of firing activity in mPFC layer 5 pyramidal neurons, which is mediated by the stimulation of the α4β2 and α7 nAChRs and subsequent increase in intracellular Ca2+ levels followed by the suppression of the Kv7 channels. The novel effect of nicotine might underlie the nicotine-induced enhancement of attention, working memory and recognition.
Collapse
Affiliation(s)
- Shoma Izumi
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Masaki Domoto
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Hirohito Esaki
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Hitoki Sasase
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| |
Collapse
|
35
|
Martínez-Drudis L, Amorós-Aguilar L, Torras-Garcia M, Serra-Elias B, Costa-Miserachs D, Portell-Cortés I, Coll-Andreu M. Delayed voluntary physical exercise restores "when" and "where" object recognition memory after traumatic brain injury. Behav Brain Res 2021; 400:113048. [PMID: 33279639 DOI: 10.1016/j.bbr.2020.113048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/03/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Physical exercise has been associated with improved cognition and may even reduce memory deficits after brain injuries. The aims of this work were to: 1) assess whether voluntary physical exercise can reduce the deficits associated with traumatic brain injury (TBI) in two different components of episodic-like memory based on object recognition, temporal order memory ("when"), and object location memory ("where"); and 2) determine whether changes in levels of brain-derived neurotrophic factor (BDNF) in the hippocampus and prefrontal cortex, as well as alterations in hippocampal cytokines, insulin-like growth factor-1 (IGF-1) and vascular endothelial growth factor (VEGF), may influence the effects exercise has on either or both tasks. The rats were distributed into a sham group, a TBI group that remained sedentary (TBI-sed), and a TBI group that had access to a running wheel for a 25-day period from post-injury day 11 (TBI-exe). The rats were sacrificed after the "where" memory task, at post-injury day 37. Physical exercise restored the "when" and "where" memories, which had been impaired by the TBI, and increased the concentration of BDNF in the hippocampus, but not the prefrontal cortex. Neither TBI nor exercise were found to significantly affect hippocampal cytokines, IGF-1 or VEGF at this time post-injury. BDNF levels showed significant positive correlations with exercise, and with "when" (but not "where") memory. These results indicate that post-injury physical exercise restores "when" and "where" object recognition memory tasks after TBI, and that increased BDNF seems to be involved in this effect, particularly with regard to "when" memory.
Collapse
Affiliation(s)
- Laura Martínez-Drudis
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; CHU de Quebec Research Center, Axe Neurosciences, Department of Molecular Medicine, Faculty of medicine, Université Laval, Quebec City, Canada
| | - Laura Amorós-Aguilar
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Meritxell Torras-Garcia
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Bruna Serra-Elias
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - David Costa-Miserachs
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Isabel Portell-Cortés
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Margalida Coll-Andreu
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Spain.
| |
Collapse
|
36
|
Alghamdi BS. The Effect of Short-Term Feeding of a High-Coconut Oil or High-Fat Diet on Neuroinflammation and the Performance of an Object-Place Task in Rats. Neurochem Res 2021; 46:287-298. [PMID: 33221998 DOI: 10.1007/s11064-020-03163-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/30/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
The consumption of high-fat and high-sugar diets, in the form of junk food, and binge eating are now common. Increasing evidence suggests that a high-fat diet (HFD) can induce neuroinflammation and alter behavior. I aimed to study the effects of diets of differing fat content on neuroinflammation and spatial memory using an object-place (OP) task. Thirty-two adult male rats were allocated to four groups and fed a regular diet (Regular diet), a control diet (Control diet), an HFD (60% of calories from lard), or a high-coconut oil diet (HCOD; 60% of calories from coconut oil) for 3 days. Their water intake, food consumption, body mass, and metabolic variables were measured. HFD-fed rats showed significantly poorer performance on the OP task, as assessed using the discrimination index (- 0.208 ± 0.094), than the Regular (0.462 ± 0.078; P < 0.0001) and Control (0.379 ± 0.081; P = 0.0003) groups. However, no significant difference was observed in spatial memory between the HCOD and Regular groups. The concentrations of neuroinflammatory markers (interleukin [IL]-1β, IL-6, tumor necrosis factor α, and nuclear factor κB) were also measured in the hippocampus and prefrontal cortex. HFD-fed rats showed significantly higher levels of neuroinflammatory markers than the Regular and Control diet-fed groups. HCOD feeding did not induce neuroinflammation in the hippocampus and prefrontal cortex compared with the Regular and Control groups.
Collapse
Affiliation(s)
- Badrah Saeed Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
37
|
Papp M, Gruca P, Lason M, Litwa E, Solecki W, Willner P. AMPA receptors mediate the pro-cognitive effects of electrical and optogenetic stimulation of the medial prefrontal cortex in antidepressant non-responsive Wistar-Kyoto rats. J Psychopharmacol 2020; 34:1418-1430. [PMID: 33200659 PMCID: PMC7708672 DOI: 10.1177/0269881120967857] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The chronic mild stress (CMS) procedure is a widely used animal model of depression, and its application in Wistar-Kyoto (WKY) rats has been validated as a model of antidepressant-refractory depression. While not responding to chronic treatment with antidepressant drugs, WKY rats do respond to acute deep brain stimulation (DBS) of the medial prefrontal cortex (mPFC). In antidepressant-responsive strains there is evidence suggesting a role for AMPA subtype of glutamate receptor in the action mechanism of both antidepressants and DBS. METHODS Animals were subjected to CMS for 6 to 8 weeks; sucrose intake was monitored weekly and novel object recognition (NOR) test was conducted following recovery from CMS. Wistars were treated chronically with venlafaxine (VEN), while WKY were treated acutely with either DBS, optogenetic stimulation (OGS) of virally-transduced (AAV5-hSyn-ChR2-EYFP) mPFC or ventral hippocampus, or acute intra-mPFC injection of the AMPA receptor positive allosteric modulator CX-516. The AMPA receptor antagonist NBQX was administered, at identical sites in mPFC, immediately following the exposure trial in the NOR. RESULTS Sucrose intake and NOR were suppressed by CMS, and restored by VEN in Wistars and by DBS, OGS, or CX-516 in WKY. However, OGS of the ventral hippocampal afferents to mPFC was ineffective. A low dose of NBQX selectively blocked the procognitive effect of VEN, DBS and OGS. CONCLUSIONS These results suggest that activation of AMPA receptors in the mPFC represents a common pathway for the antidepressant effects of both conventional (VEN) and novel (DBS, OGS) antidepressant modalities, in both antidepressant responsive (Wistar) and antidepressant-resistant (WKY) rats.
Collapse
Affiliation(s)
- Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland,Mariusz Papp, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, Krakow, 31-343, Poland.
| | - Piotr Gruca
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Magdalena Lason
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewa Litwa
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wojciech Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| | - Paul Willner
- Department of Psychology, Swansea University, Swansea, UK
| |
Collapse
|
38
|
Wang Z, Alderman MH, Asgari C, Taylor HS. Fetal Bisphenol-A Induced Changes in Murine Behavior and Brain Gene Expression Persisted in Adult-aged Offspring. Endocrinology 2020; 161:5905560. [PMID: 32926169 PMCID: PMC7609133 DOI: 10.1210/endocr/bqaa164] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/10/2020] [Indexed: 12/17/2022]
Abstract
In utero Bisphenol A (BPA) exposure has been linked to many deficits during brain development, including sexual differentiation, behavior, and motor coordination. Yet, how BPA induces these disorders and whether its effects are long lasting are largely unknown. In this study, using a mouse model, we demonstrated that in utero exposure to an environmentally relevant dose of BPA induced locomotor deficits, anxiety-like behavior, and declarative memory impairments that persisted into old age (18 months). Compared to the control animals, the BPA-exposed mice had a significant decrease in locomotor activity, exploratory tendencies, and long-term memory, and an increase in anxiety. The global brain gene expression profile was altered permanently by BPA treatment and showed regional and sexual differences. The BPA-treated male mice had more changes in the hippocampus, while female mice experienced more changes in the cortex. Overall, we demonstrate that in utero exposure to BPA induces permanent changes in brain gene expression in a region-specific and sex-specific manner, including a significant decrease in locomotor activity, learning ability, long-term memory, and an increase in anxiety. Fetal/early life exposures permanently affect neurobehavioral functions that deteriorate with age; BPA exposure may compound the effects of aging.
Collapse
Affiliation(s)
- Zhihao Wang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Myles H Alderman
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Cyrus Asgari
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
- Correspondence: Hugh S. Taylor, MD, Yale University School of Medicine, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, P.O. Box 208063, New Haven, CT 06520-8063, USA. E-mail:
| |
Collapse
|
39
|
Intrinsic Brain Abnormalities in Patients with Hepatitis C Virus Infection with Cognitive Impairment: A Preliminary Resting-State fMRI Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1693043. [PMID: 33204682 PMCID: PMC7655249 DOI: 10.1155/2020/1693043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 11/17/2022]
Abstract
Purpose Patients with a hepatitis C virus (HCV) infection frequently exhibit various neuropsychiatric complications such as cognitive decline. This study is aimed at investigating alterations in regional and network-level neural function in patients with HCV infection and examining the association between these alterations and patients' cognition dysfunction. Methods The study included 17 patients with HCV infection and 17 healthy controls. These individuals had undergone resting-state functional magnetic resonance imaging as well as cognitive assessment using a battery of tests that were collectively called the "psychometric hepatic encephalopathy score (PHES)" examination. Analyses of amplitude of low-frequency fluctuation (ALFF) and seed-based functional connectivity (FC) were conducted to assess, respectively, regional neural function and functional integration. Results HCV-infected patients performed significantly worse in cognitive tests. In the HCV group, ALFF decreased in Region 1 (left medial frontal gyrus and bilateral anterior cingulate gyrus) and Region 2 (right middle and superior frontal gyrus). The HCV group showed lower FC between Region 1 and right middle frontal gyrus, whereas they presented an increase in FC between Region 2 and the left supramarginal gyrus/superior temporal gyrus and right supramarginal gyrus. No significant correlation was observed between ALFF/FC measurements and PHES result. Conclusion This preliminary study presents additional evidence that HCV infection affects brain function, including local intrinsic neural activity and global functional integration.
Collapse
|
40
|
Renczés E, Borbélyová V, Steinhardt M, Höpfner T, Stehle T, Ostatníková D, Celec P. The Role of Estrogen in Anxiety-Like Behavior and Memory of Middle-Aged Female Rats. Front Endocrinol (Lausanne) 2020; 11:570560. [PMID: 33117285 PMCID: PMC7575693 DOI: 10.3389/fendo.2020.570560] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022] Open
Abstract
Aging in women is associated with low estrogen, but also with cognitive decline and affective disorders. Whether low estrogen is causally responsible for these behavioral symptoms is not clear. Thus, we aimed to examine the role of estradiol in anxiety-like behavior and memory in rats at middle age. Twelve-month old female rats underwent ovariectomy (OVX) or were treated with 1 mg/kg of letrozole-an aromatase inhibitor. In half of the OVX females, 10 μg/kg of 17β-estradiol was supplemented daily for 4 weeks. Vehicle-treated sham-operated and OVX females served as controls. For behavioral assessment open field, elevated plus maze and novel object recognition tests were performed. Interaction between ovarian condition and additional treatment had the main effect on anxiety-like behavior of rats in the open field test. In comparison to control females, OVX females entered less frequently into the center zone of the open field (p < 0.01) and showed lower novel object discrimination (p = 0.05). However, estradiol-supplemented OVX rats had higher number of center-zone entries (p < 0.01), spent more time in the center zone (p < 0.05), and showed lower thigmotaxis (p < 0.01) when compared to OVX group. None of the hormonal manipulations affected anxiety-like behavior in the elevated plus maze test significantly, but a mild effect of interaction between ovarian condition and treatment was shown (p = 0.05). In conclusion, ovariectomy had slight negative effect on open-field ambulation and short-term recognition memory in middle-aged rats. In addition, a test-specific anxiolytic effect of estradiol supplementation was found. In contrast, letrozole treatment neither affected anxiety-like behavior nor memory.
Collapse
Affiliation(s)
- Emese Renczés
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Veronika Borbélyová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Manuel Steinhardt
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Tim Höpfner
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Thomas Stehle
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Daniela Ostatníková
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
- Institute of Pathophysiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
41
|
Kuiper LB, Lucas KA, Mai V, Coolen LM. Enhancement of Drug Seeking Following Drug Taking in a Sexual Context Requires Anterior Cingulate Cortex Activity in Male Rats. Front Behav Neurosci 2020; 14:87. [PMID: 32670029 PMCID: PMC7330085 DOI: 10.3389/fnbeh.2020.00087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/08/2020] [Indexed: 11/13/2022] Open
Abstract
Individual variance in vulnerability to develop addictions is influenced by social factors. Specifically, drug-taking in a sexual context appears to enhance further drug-seeking behavior in human users, as these users identify the effects of drugs to enhance sexual pleasure as a primary reason for continued drug use. Methamphetamine (Meth) is commonly used in this context. Similarly, male rats that self-administered Meth immediately followed by sexual behavior display enhanced drug-seeking behavior, including attenuation of extinction and increased reinstatement to seeking of Meth-associated cues. Hence, drug-taking in a sexual context enhances vulnerability for addiction. However, the neural mechanisms by which this occurs are unknown. Here the hypothesis was tested that medial prefrontal cortex is essential for this effect of Meth and sex when experienced concurrently. First it was shown that CaMKII neurons in the anterior cingulate area (ACA) were co-activated by both Meth and sex. Next, chemogenetic inactivation of ACA CaMKII cells using AAV5-CaMKIIa-hM4Di-mCherry was shown not to affect Meth-induced locomotor activity or sexual behavior. Subsequently, chemogenetic inactivation of ACA CaMKII neurons during Meth self-administration followed by sexual behavior was shown to prevent the effects of Meth and sex on enhanced reinstatement of Meth-seeking but did not affect enhanced drug-seeking during extinction tests. These results indicate that ACA CaMKII cell activation during exposure to Meth in a sexual context plays an essential role in the subsequent enhancement of drug-seeking during reinstatement tests.
Collapse
Affiliation(s)
- Lindsey B Kuiper
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Kathryn A Lucas
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Vy Mai
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lique M Coolen
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States.,Brain Health Research Institute, Kent State University, Kent, OH, United States.,Department of Biological Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
42
|
Teixeira AE, Rocha-Gomes A, Pereira dos Santos T, Amaral BLS, da Silva AA, Malagutti AR, Leite FRF, Stuckert-Seixas SR, Riul TR. Cafeteria diet administered from lactation to adulthood promotes a change in risperidone sensitivity on anxiety, locomotion, memory, and social interaction of Wistar rats. Physiol Behav 2020; 220:112874. [DOI: 10.1016/j.physbeh.2020.112874] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 01/24/2023]
|
43
|
Prajit R, Sritawan N, Suwannakot K, Naewla S, Aranarochana A, Sirichoat A, Pannangrong W, Wigmore P, Welbat JU. Chrysin Protects against Memory and Hippocampal Neurogenesis Depletion in D-Galactose-Induced Aging in Rats. Nutrients 2020; 12:nu12041100. [PMID: 32316121 PMCID: PMC7230764 DOI: 10.3390/nu12041100] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/06/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
The interruption of hippocampal neurogenesis due to aging impairs memory. The accumulation of D-galactose (D-gal), a monosaccharide, induces brain aging by causing oxidative stress and inflammation, resulting in neuronal cell damage and memory loss. Chrysin, an extracted flavonoid, has neuroprotective effects on memory. The present study aimed to investigate the effect of chrysin on memory and hippocampal neurogenesis in brains aged using D-gal. Male Sprague-Dawley rats received either D-gal (50 mg/kg) by i.p. injection, chrysin (10 or 30 mg/kg) by oral gavage, or D-gal (50 mg/kg) and chrysin (10 or 30 mg/kg) for 8 weeks. Memory was evaluated using novel object location (NOL) and novel object recognition (NOR) tests. Hippocampal neurogenesis was evaluated using Ki-67, 5-bromo-2′-deoxyuridine (BrdU), and doublecortin (DCX) immunofluorescence staining to determine cell proliferation, cell survival, and number of immature neurons, respectively. We found that D-gal administration resulted in memory impairment as measured by NOL and NOR tests and in depletions in cell proliferation, cell survival, and immature neurons. However, co-treatment with chrysin (10 or 30 mg/kg) attenuated these impairments. These results suggest that chrysin could potentially minimize memory and hippocampal neurogenesis depletions brought on by aging.
Collapse
Affiliation(s)
- Ram Prajit
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.P.); (N.S.); (K.S.); (S.N.); (A.A.); (A.S.); (W.P.)
| | - Nataya Sritawan
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.P.); (N.S.); (K.S.); (S.N.); (A.A.); (A.S.); (W.P.)
| | - Kornrawee Suwannakot
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.P.); (N.S.); (K.S.); (S.N.); (A.A.); (A.S.); (W.P.)
| | - Salinee Naewla
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.P.); (N.S.); (K.S.); (S.N.); (A.A.); (A.S.); (W.P.)
| | - Anusara Aranarochana
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.P.); (N.S.); (K.S.); (S.N.); (A.A.); (A.S.); (W.P.)
| | - Apiwat Sirichoat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.P.); (N.S.); (K.S.); (S.N.); (A.A.); (A.S.); (W.P.)
| | - Wanassanan Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.P.); (N.S.); (K.S.); (S.N.); (A.A.); (A.S.); (W.P.)
| | - Peter Wigmore
- School of Life Sciences, Medical School, Queen’s Medical Centre, The University of Nottingham, Nottingham NG7 2RD, UK;
| | - Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (R.P.); (N.S.); (K.S.); (S.N.); (A.A.); (A.S.); (W.P.)
- Neuroscience Research and Development Group, Khon Kaen University, Khon Kaen 40002, Thailand
- Correspondence: ; Tel.: +66-84-667-6105
| |
Collapse
|
44
|
Anterior retrosplenial cortex is required for long-term object recognition memory. Sci Rep 2020; 10:4002. [PMID: 32152383 PMCID: PMC7062718 DOI: 10.1038/s41598-020-60937-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 02/19/2020] [Indexed: 01/06/2023] Open
Abstract
The retrosplenial cortex (RSC) is implicated on navigation and contextual memory. Lesions studies showed that the RSC shares functional similarities with the hippocampus (HP). Here we evaluated the role of the anterior RSC (aRSC) in the “what” and “where” components of recognition memory and contrasted it with that of the dorsal HP (dHP). Our behavioral and molecular findings show functional differences between the aRSC and the dHP in recognition memory. The inactivation of the aRSC, but not the dHP, impairs the consolidation and expression of the “what” memory component. In addition, object recognition task is accompanied by c-Fos levels increase in the aRSC. Interestingly, we found that the aRSC is recruited to process the “what” memory component only if it is active during acquisition. In contrast, both the aRSC and dHP are required for encoding the “where” component, which correlates with c-Fos levels increase. Our findings introduce a novel role of the aRSC in recognition memory, processing not only the “where”, but also the “what” memory component.
Collapse
|
45
|
Xu Z, Shen B, Taji W, Sun P, Naya Y. Convergence of distinct functional networks supporting naming and semantic recognition in the left inferior frontal gyrus. Hum Brain Mapp 2020; 41:2389-2405. [PMID: 32065445 PMCID: PMC7268040 DOI: 10.1002/hbm.24953] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/21/2019] [Accepted: 02/09/2020] [Indexed: 12/11/2022] Open
Abstract
Naming individual objects is accompanied with semantic recognition. Previous studies examined brain‐networks responsible for these operations individually. However, it remains unclear how these brain‐networks are related. To address this problem, we examined the brain‐networks during a novel object‐naming task, requiring participants to name animals in photographs at a specific‐level (e.g., “pigeon”). When the participants could not remember specific names, they answered basic names (e.g., “bird”). After fMRI scanning during the object‐naming task, the participants rated familiarity of the animals based on their sense of knowing. Since participants tend to remember specific names for familiar objects compared with unfamiliar objects, a typical issue in an object‐naming task is an internal covariance between the naming and familiarity levels. We removed this confounding factor by adjusting the familiarity/naming level of stimuli, and demonstrated distinct brain regions related to the two operations. Among them, the left inferior frontal gyrus triangularis (IFGtri) contained object‐naming and semantic‐recognition related areas in its anterior‐ventral and posterior‐dorsal parts, respectively. Psychophysiological interaction analyses suggested that both parts show connectivity with the brain regions related to object‐naming. By examining the connectivity under control tasks requiring nonlexical semantic retrieval (e.g., animal's body color), we found that both IFGtri parts altered their targeting brain areas according to the required memory attributes, while only the posterior‐dorsal part connected the brain regions related to semantic recognition. Together, the semantic recognition may be processed by distinct brain network from those for voluntary semantic retrievals including object‐naming although all these networks are mediated by the posterior‐dorsal IFGtri.
Collapse
Affiliation(s)
- Zhansheng Xu
- School of Psychological and Cognitive Sciences, Peking University, Beijing, China
| | - Bo Shen
- School of Psychological and Cognitive Sciences, Peking University, Beijing, China.,Department of Psychology, Zhejiang Normal University, Jinhua, China
| | - Wael Taji
- School of Psychological and Cognitive Sciences, Peking University, Beijing, China.,Yenching Academy, Peking University, Beijing, China
| | - Pei Sun
- Department of Psychology, School of Social Sciences, Tsinghua University, Beijing, China.,Tsinghua Laboratory for Brain and Intelligence, Tsinghua University, Beijing, China
| | - Yuji Naya
- School of Psychological and Cognitive Sciences, Peking University, Beijing, China.,Center for Life Sciences, Peking University, Beijing, China.,IDG/McGovern Institute for Brain Research at Peking University, Beijing, China.,Beijing Key Laboratory of Behavior and Mental Health, Peking University, Beijing, China
| |
Collapse
|
46
|
Ayabe T, Ano Y, Ohya R, Kitaoka S, Furuyashiki T. The Lacto-Tetrapeptide Gly-Thr-Trp-Tyr, β-Lactolin, Improves Spatial Memory Functions via Dopamine Release and D1 Receptor Activation in the Hippocampus. Nutrients 2019; 11:nu11102469. [PMID: 31618902 PMCID: PMC6835598 DOI: 10.3390/nu11102469] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 01/06/2023] Open
Abstract
Scope: Peptides containing tryptophan–tyrosine sequences, including the lacto-tetrapeptide glycine–threonine–tryptophan–tyrosine (GTWY) and β-lactolin, from β-lactoglobulin in whey enzymatic digestion, enhance hippocampus-dependent memory functions, which are blocked by the systemic administration of dopamine D1-like antagonist. In this study, we investigated the role of the hippocampal dopaminergic system in the memory-enhancing effect of β-lactolin. Methods and Results: The results of in vivo microdialysis revealed that oral administration of β-lactolin increased the extracellular concentration of dopamine in the hippocampus and enhanced both spatial working memory, as measured in the Y-maze test, and spatial reference memory, as measured in the novel object location test. These memory-enhancing effects of β-lactolin, but not the baseline memory functions, were impaired by the knockdown of the dopamine D1 receptor subtype in the hippocampus. β-Lactolin also enhanced object memory, as measured by the novel object recognition test. However, D1 knockdown in the hippocampus spared this memory function either with or without the administration of β-lactolin. Conclusions: The present results indicate that oral administration of β-lactolin increases dopamine release and D1 receptor signaling in the hippocampus, thereby enhancing spatial memory, but it may improve object memory via a separate mechanism.
Collapse
Affiliation(s)
- Tatsuhiro Ayabe
- Research Laboratories for Health Science & Food Technologies, Kirin Holdings Company Ltd., 1-13-5 Fukuura Kanazawa-ku, Yokohama-shi, Kanagawa 236-0004, Japan.
| | - Yasuhisa Ano
- Research Laboratories for Health Science & Food Technologies, Kirin Holdings Company Ltd., 1-13-5 Fukuura Kanazawa-ku, Yokohama-shi, Kanagawa 236-0004, Japan.
| | - Rena Ohya
- Research Laboratories for Health Science & Food Technologies, Kirin Holdings Company Ltd., 1-13-5 Fukuura Kanazawa-ku, Yokohama-shi, Kanagawa 236-0004, Japan.
| | - Shiho Kitaoka
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
- AMED-CREST, Chiyoda-ku, Tokyo 100-0004, Japan.
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
- AMED-CREST, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
47
|
Flores-Burgess A, Millón C, Gago B, García-Durán L, Cantero-García N, Coveñas R, Narváez JA, Fuxe K, Santín L, Díaz-Cabiale Z. Galanin (1–15)-fluoxetine interaction in the novel object recognition test. Involvement of 5-HT1A receptors in the prefrontal cortex of the rats. Neuropharmacology 2019; 155:104-112. [DOI: 10.1016/j.neuropharm.2019.05.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 04/07/2019] [Accepted: 05/18/2019] [Indexed: 10/26/2022]
|
48
|
Chiquita S, Ribeiro M, Castelhano J, Oliveira F, Sereno J, Batista M, Abrunhosa A, Rodrigues-Neves AC, Carecho R, Baptista F, Gomes C, Moreira PI, Ambrósio AF, Castelo-Branco M. A longitudinal multimodal in vivo molecular imaging study of the 3xTg-AD mouse model shows progressive early hippocampal and taurine loss. Hum Mol Genet 2019; 28:2174-2188. [PMID: 30816415 PMCID: PMC6586150 DOI: 10.1093/hmg/ddz045] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/15/2019] [Accepted: 02/18/2019] [Indexed: 01/09/2023] Open
Abstract
The understanding of the natural history of Alzheimer's disease (AD) and temporal trajectories of in vivo molecular mechanisms requires longitudinal approaches. A behavioral and multimodal imaging study was performed at 4/8/12 and 16 months of age in a triple transgenic mouse model of AD (3xTg-AD). Behavioral assessment included the open field and novel object recognition tests. Molecular characterization evaluated hippocampal levels of amyloid β (Aβ) and hyperphosphorylated tau. Magnetic resonance imaging (MRI) included assessment of hippocampal structural integrity, blood-brain barrier (BBB) permeability and neurospectroscopy to determine levels of the endogenous neuroprotector taurine. Longitudinal brain amyloid accumulation was assessed using 11C Pittsburgh compound B positron emission tomography (PET), and neuroinflammation/microglia activation was investigated using 11C-PK1195. We found altered locomotor activity at months 4/8 and 16 months and recognition memory impairment at all time points. Substantial early reduction of hippocampal volume started at month 4 and progressed over 8/12 and 16 months. Hippocampal taurine levels were significantly decreased in the hippocampus at months 4/8 and 16. No differences were found for amyloid and neuroinflammation with PET, and BBB was disrupted only at month 16. In summary, 3xTg-AD mice showed exploratory and recognition memory impairments, early hippocampal structural loss, increased Aβ and hyperphosphorylated tau and decreased levels of taurine. In sum, the 3xTg-AD animal model mimics pathological and neurobehavioral features of AD, with early-onset recognition memory loss and MRI-documented hippocampal damage. The early-onset profile suggests temporal windows and opportunities for therapeutic intervention, targeting endogenous neuroprotectors such as taurine.
Collapse
Affiliation(s)
- Samuel Chiquita
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Mário Ribeiro
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| | - João Castelhano
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| | - Francisco Oliveira
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| | - José Sereno
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| | - Marta Batista
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Antero Abrunhosa
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| | - Ana C Rodrigues-Neves
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Rafael Carecho
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Filipa Baptista
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Catarina Gomes
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - António F Ambrósio
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
49
|
Effects of the synthetic cannabinoid 5F-AMB on anxiety and recognition memory in mice. Psychopharmacology (Berl) 2019; 236:2235-2242. [PMID: 30868181 DOI: 10.1007/s00213-019-05222-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
Abstract
RATIONALE N-[[1-(5-fluoropentyl)-1H-indazol-3-yl]carbonyl]-L-valine methyl ester (5F-AMB) is a synthetic cannabinoid that has been distributed recently. Although inhalation of 5F-AMB produces adverse effects, such as impaired memory and disturbed consciousness, in humans, the psychopharmacological effects of 5F-AMB in rodents have not been investigated. OBJECTIVES We first examined the effects of intraperitoneal and intracerebroventricular injections of 5F-AMB on anxiety-like behavior and locomotor activity in the open field (OF) test and recognition memory in the novel object recognition test (NOR) in C57BL/6J mice. We also examined whether a cannabinoid 1 (CB1) receptor antagonist AM251 blocks the effects of 5F-AMB. We next examined the effects of 5F-AMB infusion into the medial prefrontal cortex (mPFC), a brain region associated with anxiety and memory, on these tests. RESULTS Intraperitoneal injection of 5F-AMB (0.3 mg/kg) dramatically decreased locomotor activity in the OF, and this effect was partially reversed by AM251 (3 mg/kg). Intracerebroventricular infusion of 5F-AMB (10 nmol) produced an anxiolytic effect in the OF and impaired acquisition, but not retrieval, of recognition memory in the NOR, and these effects were blocked by co-infusion of AM251 (1.8 nmol). Bilateral intra-mPFC infusion of 5F-AMB (10 pmol/side) similarly produced impaired recognition memory acquisition, but no anxiolytic effect. CONCLUSIONS The results demonstrate that centrally administered 5F-AMB produces anxiolytic effect and impaired recognition memory acquisition via activation of CB1 receptors, while systemic 5F-AMB severely impaired locomotor activity. The mPFC is involved in 5F-AMB-induced impairment of recognition memory acquisition. However, other brain region(s) may contribute to the 5F-AMB-induced anxiolytic effect.
Collapse
|
50
|
Moreton E, Baron P, Tiplady S, McCall S, Clifford B, Langley-Evans S, Fone K, Voigt J. Impact of early exposure to a cafeteria diet on prefrontal cortex monoamines and novel object recognition in adolescent rats. Behav Brain Res 2019; 363:191-198. [DOI: 10.1016/j.bbr.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/24/2019] [Accepted: 02/02/2019] [Indexed: 12/19/2022]
|