1
|
Souza TP, Rodríguez-Vega A, Dutra-Tavares AC, Semeão KA, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Nucleus Accumbens Proteome Disbalance in an Adolescent Mouse Model of Schizophrenia and Nicotine Misuse Comorbidity. Biomedicines 2025; 13:901. [PMID: 40299488 DOI: 10.3390/biomedicines13040901] [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: 02/21/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Schizophrenia and nicotine misuse are a comorbid condition that frequently develops during adolescence. Considering the role of the nucleus accumbens (NAcc) as a common neurobiological substrate for these psychiatric disorders, label-free proteomics was employed to identify NAcc deregulated proteins in male and female mouse models of schizophrenia with a history of adolescent nicotine exposure. Methods: Phencyclidine was used to model schizophrenia, and minipump infusions were used to model nicotine misuse. Results: Enrichment Reactome pathway and protein-protein interaction analyses showed that the cytoskeleton and associated synaptic plasticity mechanisms, energy metabolism, and nervous system development were affected in both sexes. In particular, Ncam1 (Neural cell adhesion molecule 1) could be of interest as a candidate marker of synaptic plasticity disbalance. Its deregulation in the NAcc of both sexes suggests that it lies at the core of the comorbidity pathophysiology. When considering sex-selective effects, Cs (Citrate synthase) and Mapk3 (Mitogen-activated protein kinase 3) were identified as exclusively deregulated in female and male mice, respectively. Since both proteins were previously shown to be exclusively deregulated in the medial prefrontal cortex of co-modeled mice, a common mesocortical and mesolimbic system effect can be inferred, supporting the role of aberrant energy metabolism and synaptic plasticity in the comorbidity model. Conclusions: The current data provide insights into the NAcc proteome disbalance in an adolescent preclinical model of combined schizophrenia and nicotine misuse, pointing to relevant pathways and early markers of the comorbidity.
Collapse
Affiliation(s)
- Thainá Pereira Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 Andar-Vila Isabel, Rio de Janeiro 20550-170, RJ, Brazil
| | - Andrés Rodríguez-Vega
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 Andar-Vila Isabel, Rio de Janeiro 20550-170, RJ, Brazil
| | - Ana Carolina Dutra-Tavares
- Departamento de Ciências Biomédicas e Saúde, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Cabo Frio 28905-320, RJ, Brazil
| | - Keila A Semeão
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 Andar-Vila Isabel, Rio de Janeiro 20550-170, RJ, Brazil
| | - Claudio Carneiro Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 Andar-Vila Isabel, Rio de Janeiro 20550-170, RJ, Brazil
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores, Universidade do Estado do Rio de Janeiro (UERJ), São Gonçalo 24435-005, RJ, Brazil
| | - Alex Christian Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 Andar-Vila Isabel, Rio de Janeiro 20550-170, RJ, Brazil
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 Andar-Vila Isabel, Rio de Janeiro 20550-170, RJ, Brazil
| |
Collapse
|
2
|
Romero-Muñoz L, Sanz-Martos AB, Olmo ND, Merino B, Ruiz-Gayo M, Cano V. Impact of fatty acids on glutamate-related gene expression in the hippocampus: Focus on lauric acid. Neurosci Lett 2025; 850:138152. [PMID: 39923978 DOI: 10.1016/j.neulet.2025.138152] [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: 11/25/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Excessive dietary fat consumption has been linked to impairments in synaptic plasticity in the hippocampus (HIP), a brain region crucial for learning and memory that relies on balanced glutamatergic neurotransmission. This study investigates the acute effects of three fatty acids (FAs)-lauric acid (LA), palmitic acid (PA), and oleic acid (OA)-on glutamate (GLU)-related gene expression in the HIP of male and female young mice. Hippocampal slices were treated with FAs, and mRNA levels of genes involved in GLU transport, GLU-glutamine (GLN) cycling, and GLU receptor subunit encoding were quantified using RT-PCR. FA treatment reduced mRNA levels of enzymes involved in the conversion of GLU to GLN (glutamine synthetase; GS), GABA (glutamate decarboxylase 1; GAD67), and α-ketoglutarate (glutamate pyruvate transaminase 2; AAT2). Additionally, the expression of glutamine transporters (SNAT1, SNAT2, SNAT3), the astrocytic GLU transporter GLT-1, and the NMDA receptor subunit NMDA2a was also reduced. These effects were most pronounced with LA. Notably, while the HIP showed similar sensitivity to fatty acids across sexes, overall gene expression levels were lower in females. These findings highlight the acute susceptibility of hippocampal GLU-related pathways to FA exposure, particularly LA, suggesting potential risks of high-LA diets on cognitive function. Further research is needed to explore the long-term consequences of dietary fat on hippocampal health and its sex-specific effects.
Collapse
Affiliation(s)
- Laura Romero-Muñoz
- Department of Health and Pharmaceutical Sciences Facultad de Farmacia Universidad CEU-San Pablo CEU Universities Madrid Spain
| | - Ana Belén Sanz-Martos
- Department of Psychobiology School of Psychology National University for Distance Education (UNED) Madrid Spain
| | - Nuria Del Olmo
- Department of Psychobiology School of Psychology National University for Distance Education (UNED) Madrid Spain
| | - Beatriz Merino
- Department of Health and Pharmaceutical Sciences Facultad de Farmacia Universidad CEU-San Pablo CEU Universities Madrid Spain
| | - Mariano Ruiz-Gayo
- Department of Health and Pharmaceutical Sciences Facultad de Farmacia Universidad CEU-San Pablo CEU Universities Madrid Spain
| | - Victoria Cano
- Department of Health and Pharmaceutical Sciences Facultad de Farmacia Universidad CEU-San Pablo CEU Universities Madrid Spain.
| |
Collapse
|
3
|
Cuozzo AM, Peeters LD, Ahmed CD, Wills LJ, Gass JT, Brown RW. Investigation of the Roles of the Adenosine A(2A) and Metabotropic Glutamate Receptor Type 5 (mGlu5) Receptors in Prepulse Inhibition and CREB Signaling in a Heritable Rodent Model of Psychosis. Cells 2025; 14:182. [PMID: 39936973 PMCID: PMC11817787 DOI: 10.3390/cells14030182] [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: 12/26/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
The metabotropic glutamate receptor type 5 (mGlu5) and adenosine A(2A) receptor form a mutually inhibitory heteromer with the dopamine D2 receptor, where the activation of either mGlu5 or A(2A) leads to reduced D2 signaling. This study investigated whether a mGlu5-positive allosteric modulator (PAM) or an A(2A) agonist treatment could mitigate sensorimotor gating deficits and alter cyclic AMP response element-binding protein (CREB) levels in a rodent neonatal quinpirole (NQ) model of psychosis. F0 Sprague-Dawley rats were treated with neonatal saline or quinpirole (1 mg/kg) from postnatal day 1 to 21 and bred to produce an F1 generation. F1 offspring underwent prepulse inhibition (PPI) testing from postnatal day 44 to 48 to assess sensorimotor gating. The rats were treated with mGlu5 PAM 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB) or A(2A) agonist CGS21680. Rats with at least one NQ-treated parent showed PPI deficits, which were alleviated by both CDPPB and CGS21680. Sex differences were noted across groups, with CGS21680 showing greater efficacy than CDPPB. Additionally, CREB levels were elevated in the nucleus accumbens (NAc), and both CDPPB and CGS21680 reduced CREB expression to control levels. These findings suggest that targeting the adenosinergic and glutamatergic systems alleviates sensorimotor gating deficits and abnormal CREB signaling, both of which are associated with psychosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Russell W. Brown
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (A.M.C.); (C.D.A.)
| |
Collapse
|
4
|
Antos Z, Żukow X, Bursztynowicz L, Jakubów P. Beyond NMDA Receptors: A Narrative Review of Ketamine's Rapid and Multifaceted Mechanisms in Depression Treatment. Int J Mol Sci 2024; 25:13658. [PMID: 39769420 PMCID: PMC11728282 DOI: 10.3390/ijms252413658] [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: 11/17/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/14/2025] Open
Abstract
The rising prevalence of depression, with its associated suicide risk, demands effective fast-acting treatments. Ketamine has emerged as promising, demonstrating rapid antidepressant effects. While early studies show swift mood improvements, its precise mechanisms remain unclear. This article aims to compile and synthesize the literature on ketamine's molecular actions. Ketamine primarily works by antagonizing NMDA receptors, reducing GABAergic inhibition, and increasing glutamate release. This enhanced glutamate activates AMPA receptors, triggering crucial downstream cascades, including BDNF-TrkB and mTOR pathways, promoting synaptic proliferation and regeneration. Moreover, neuroimaging studies have demonstrated alterations in brain networks involved in emotional regulation, including the Default Mode Network (DMN), Central Executive Network (CEN), and Salience Network (SN), which are frequently disrupted in depression. Despite the promising findings, the literature reveals significant inaccuracies and gaps in understanding the full scope of ketamine's therapeutic potential. For instance, ketamine engages with opioid receptors, insinuating a permissive role of the opioid system in amplifying ketamine's antidepressant effects, albeit ketamine does not operate as a direct opioid agonist. Further exploration is requisite to comprehensively ascertain its safety profile, long-term efficacy, and the impact of genetic determinants, such as BDNF polymorphisms, on treatment responsiveness.
Collapse
Affiliation(s)
| | | | | | - Piotr Jakubów
- Department of Paediatric Anaesthesiology and Intensive Therapy with Pain Division, Faculty of Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland; (Z.A.); (X.Ż.); (L.B.)
| |
Collapse
|
5
|
Le AA, Lauterborn JC, Jia Y, Cox CD, Lynch G, Gall CM. Metabotropic NMDAR Signaling Contributes to Sex Differences in Synaptic Plasticity and Episodic Memory. J Neurosci 2024; 44:e0438242024. [PMID: 39424366 PMCID: PMC11638816 DOI: 10.1523/jneurosci.0438-24.2024] [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: 02/21/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
NMDA receptor (NMDAR)-mediated calcium influx triggers the induction and initial expression of long-term potentiation (LTP). Here we report that in male rodents, ion flux-independent (metabotropic) NMDAR signaling is critical for a third step in the production of enduring LTP, i.e., cytoskeletal changes that stabilize the activity-induced synaptic modifications. Surprisingly, females rely upon estrogen receptor alpha (ERα) for the metabotropic NMDAR operations used by males. Blocking NMDAR channels with MK-801 eliminated LTP expression in hippocampal field CA1 of both sexes but left intact theta burst stimulation (TBS)-induced actin polymerization within dendritic spines. A selective antagonist (Ro25-6981) of the NMDAR GluN2B subunit had minimal effects on synaptic responses but blocked actin polymerization and LTP consolidation in males only. Conversely, an ERα antagonist thoroughly disrupted TBS-induced actin polymerization and LTP in females while having no evident effect in males. In an episodic memory paradigm, Ro25-6981 prevented acquisition of spatial locations by males but not females, whereas an ERα antagonist blocked acquisition in females but not males. Sex differences in LTP consolidation were accompanied by pronounced differences in episodic memory in tasks involving minimal (for learning) cue sampling. Males did better on acquisition of spatial information whereas females had much higher scores than males on tests for acquisition of the identity of cues (episodic "what") and the order in which the cues were sampled (episodic "when"). We propose that sex differences in synaptic processes used to stabilize LTP result in differential encoding of the basic elements of episodic memory.
Collapse
Affiliation(s)
- Aliza A Le
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Julie C Lauterborn
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Yousheng Jia
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Conor D Cox
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Gary Lynch
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
- Psychiatry and Human Behavior, University of California, Irvine, California 92697
| | - Christine M Gall
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
- Neurobiology and Behavior, University of California, Irvine, California 92697
| |
Collapse
|
6
|
Kniffin AR, Briand LA. Sex differences in glutamate transmission and plasticity in reward related regions. Front Behav Neurosci 2024; 18:1455478. [PMID: 39359325 PMCID: PMC11445661 DOI: 10.3389/fnbeh.2024.1455478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Disruptions in glutamate homeostasis within the mesolimbic reward circuitry may play a role in the pathophysiology of various reward related disorders such as major depressive disorders, anxiety, and substance use disorders. Clear sex differences have emerged in the rates and symptom severity of these disorders which may result from differing underlying mechanisms of glutamatergic signaling. Indeed, preclinical models have begun to uncover baseline sex differences throughout the brain in glutamate transmission and synaptic plasticity. Glutamatergic synaptic strength can be assessed by looking at morphological features of glutamatergic neurons including spine size, spine density, and dendritic branching. Likewise, electrophysiology studies evaluate properties of glutamatergic neurons to provide information of their functional capacity. In combination with measures of glutamatergic transmission, synaptic plasticity can be evaluated using protocols that induce long-term potentiation or long-term depression. This review will consider preclinical rodent literature directly comparing glutamatergic transmission and plasticity in reward related regions of males and females. Additionally, we will suggest which regions are exhibiting evidence for sexually dimorphic mechanisms, convergent mechanisms, or no sex differences in glutamatergic transmission and plasticity and highlight gaps in the literature for future investigation.
Collapse
Affiliation(s)
- Alyssa R. Kniffin
- Department of Psychology & Neuroscience, Temple University, Philadelphia, PA, United States
| | - Lisa A. Briand
- Department of Psychology & Neuroscience, Temple University, Philadelphia, PA, United States
- Neuroscience Program, Temple University, Philadelphia, PA, United States
| |
Collapse
|
7
|
Castillo-Vazquez SK, Massieu L, Rincón-Heredia R, García-de la Torre P, Quiroz-Baez R, Gomez-Verjan JC, Rivero-Segura NA. Glutamatergic Neurotransmission in Aging and Neurodegenerative Diseases: A Potential Target to Improve Cognitive Impairment in Aging. Arch Med Res 2024; 55:103039. [PMID: 38981341 DOI: 10.1016/j.arcmed.2024.103039] [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: 01/11/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
Aging is characterized by the decline in many of the individual's capabilities. It has been recognized that the brain undergoes structural and functional changes during aging that are occasionally associated with the development of neurodegenerative diseases. In this sense, altered glutamatergic neurotransmission, which involves the release, binding, reuptake, and degradation of glutamate (Glu) in the brain, has been widely studied in physiological and pathophysiological aging. In particular, changes in glutamatergic neurotransmission are exacerbated during neurodegenerative diseases and are associated with cognitive impairment, characterized by difficulties in memory, learning, concentration, and decision-making. Thus, in the present manuscript, we aim to highlight the relevance of glutamatergic neurotransmission during cognitive impairment to develop novel strategies to prevent, ameliorate, or delay cognitive decline. To achieve this goal, we provide a comprehensive review of the changes reported in glutamatergic neurotransmission components, such as Glu transporters and receptors during physiological aging and in the most studied neurodegenerative diseases. Finally, we describe the current therapeutic strategies developed to target glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Selma Karime Castillo-Vazquez
- Dirección de Investigación, Instituto Nacional de Geriatría, Mexico City, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lourdes Massieu
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ruth Rincón-Heredia
- Unidad de Imagenología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Paola García-de la Torre
- 4 Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City Mexico
| | - Ricardo Quiroz-Baez
- Dirección de Investigación, Instituto Nacional de Geriatría, Mexico City, Mexico
| | | | | |
Collapse
|
8
|
Liu F, Liu Y, Shen X, Du J, Zhang H, Hou X. Ovariectomy exacerbates the disturbance of excitation- inhibition balance in the brain of APP/PS-1/tau mice. Front Mol Neurosci 2024; 17:1391082. [PMID: 39262829 PMCID: PMC11389216 DOI: 10.3389/fnmol.2024.1391082] [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: 02/24/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024] Open
Abstract
Introduction The prevalence of Alzheimer's disease (AD) is significantly gender-differentiated, with the number of female AD patients far exceeding that of males, accounting for two-thirds of the total prevalence. Although postmenopausal AD mice have been shown to have more prominent pathologic features and memory impairments than normal AD mice, the relevant molecular mechanisms leading to these outcomes have not been well elucidated. In the present study, we used the disturbance of excitation-inhibition balance in the postmenopausal brain as an entry point to explore the link between estrogen deficiency, disorders of the glutamatergic-GABAergic nervous system, and memory impairment. Methods Wild-type (WT) mice and APP/PS1/tau (3 × Tg-AD) mice (10 months old) were randomly divided into four groups: WT+Sham group, WT+OVX group, 3 × Tg-AD+Sham group and 3 × Tg-AD+OVX group. Ovariectomy (OVX) was performed in the WT+OVX group and the 3 × Tg-AD+OVX group, and sham surgery was performed in the WT+Sham group and the 3 × Tg-AD+Sham group. The learning and memory ability and the anxiety and depression-like behavior changes of mice were evaluated by behavioral experiments, and the association between estrogen-estrogen receptors pathway and glutamatergic/GABAergic nervous system and female AD was evaluated by neurochemical experiments. Results In WT and 3 × Tg-AD mice, OVX resulted in impaired learning and memory abilities and anxiety and depression-like behaviors; reduced estrogen levels and downregulated the expression of estrogen receptors; upregulated the expression of amyloid-β, amyloid precursor protein, presenilin 1, and p-tau; upregulated the expression of Bcl-2-associated X protein and downregulated the expression of B-cell lymphoma-2, promoting cell apoptosis; reduced the number of neuronal dendrites and downregulated the expression of postsynaptic density protein-95; more importantly, OVX increased brain glutamate levels but downregulated the expression of N-methyl-D-aspartate receptor-2B, excitatory amino acid transporter 1, excitatory amino acid transporter 2, γ-aminobutyric acid receptor-A and γ-aminobutyric acid receptor-B. Conclusion Our results suggested that OVX-induced estrogen-estrogen receptors pathway disruption caused learning and memory impairment and anxiety and depression-like behaviors, upregulated the expression of AD pathological markers, promoted apoptosis, destroyed neuronal structure, and most importantly, caused glutamatergic/GABAergic nervous system disorders.
Collapse
Affiliation(s)
- Fuwang Liu
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yanman Liu
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuri Shen
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiarui Du
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hanting Zhang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong, China
| | - Xueqin Hou
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
9
|
Dutra-Tavares AC, Couto LA, Souza TP, Bandeira-Martins A, Silva JO, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Nicotine's Effects on Schizophrenia-like Symptoms in a Mice Model: Time Matters. Brain Sci 2024; 14:855. [PMID: 39335351 PMCID: PMC11430416 DOI: 10.3390/brainsci14090855] [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: 07/25/2024] [Revised: 08/12/2024] [Accepted: 08/18/2024] [Indexed: 09/30/2024] Open
Abstract
Tobacco consumption in schizophrenia (SCHZ) patients is highly prevalent. Data support the occurrence of sequential events during comorbidity establishment, and both smoking first, SCHZ second and SCHZ first, smoking second sequences have been proposed. To investigate whether these two possibilities lead to distinct outcomes of comorbidity, we used a phencyclidine-induced SCHZ model and nicotine exposure as a surrogate of smoking. C57Bl/6 mice were submitted to a protocol that either began with 4 days of phencyclidine exposure or 4 days of nicotine exposure. This period was followed by 5 days of combined phencyclidine + nicotine exposure. Locomotor sensitization and pre-pulse inhibition (PPI) were assessed due to their well-known associations with SCHZ as opposed to rearing, an unrelated behavior. Nicotine priming potentiated phencyclidine-evoked sensitization. However, nicotine exposure after SCHZ modeling did not interfere with phencyclidine's effects. In the PPI test, nicotine after SCHZ modeling worsened the phencyclidine-evoked deficiency in males. In contrast, nicotine priming had no effects. Regarding rearing, nicotine priming failed to interfere with phencyclidine-mediated inhibition. Similarly, phencyclidine priming did not modify nicotine-mediated inhibition. The present results indicate that the sequence, either SCHZ-first or nicotine-first, differentially impacts comorbidity outcomes, a finding that is relevant for the identification of mechanisms of nicotine interference in the neurobiology of SCHZ.
Collapse
Affiliation(s)
- Ana Carolina Dutra-Tavares
- Departamento de Ciências Biomédicas e Saúde, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Cabo Frio 28905-320, RJ, Brazil;
| | - Luciana Araújo Couto
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Thainá P. Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Anais Bandeira-Martins
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Juliana Oliveira Silva
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Claudio C. Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores, UERJ, São Gonçalo 24435-005, RJ, Brazil;
| | - Alex C. Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro UERJ, Rio de Janeiro 20550-170, RJ, Brazil (T.P.S.); (A.B.-M.); (J.O.S.); (C.C.F.); (Y.A.-V.)
| |
Collapse
|
10
|
Mazza M, De Berardis D, Marano G. Keep in mind sex differences when prescribing psychotropic drugs. World J Psychiatry 2024; 14:194-198. [PMID: 38464773 PMCID: PMC10921286 DOI: 10.5498/wjp.v14.i2.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/02/2023] [Accepted: 01/02/2024] [Indexed: 02/06/2024] Open
Abstract
Women represent the majority of patients with psychiatric diagnoses and also the largest users of psychotropic drugs. There are inevitable differences in efficacy, side effects and long-term treatment response between men and women. Psychopharmacological research needs to develop adequately powered animal and human trials aimed to consider pharmacokinetics and pharmacodynamics of central nervous system drugs in both male and female subjects. Healthcare professionals have the responsibility to prescribe sex-specific psychopharmacotherapies with a priority to differentiate between men and women in order to minimize adverse drugs reactions, to maximize therapeutic effectiveness and to provide personalized management of care.
Collapse
Affiliation(s)
- Marianna Mazza
- Department of Neurosciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | | | - Giuseppe Marano
- Department of Neurosciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| |
Collapse
|
11
|
Lynch MA. A case for seeking sex-specific treatments in Alzheimer's disease. Front Aging Neurosci 2024; 16:1346621. [PMID: 38414633 PMCID: PMC10897030 DOI: 10.3389/fnagi.2024.1346621] [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: 11/29/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024] Open
Abstract
There is no satisfactory explanation for the sex-related differences in the incidence of many diseases and this is also true of Alzheimer's disease (AD), where females have a higher lifetime risk of developing the disease and make up about two thirds of the AD patient population. The importance of understanding the cause(s) that account for this disproportionate distribution cannot be overestimated, and is likely to be a significant factor in the search for therapeutic strategies that will combat the disease and, furthermore, potentially point to a sex-targeted approach to treatment. This review considers the literature in the context of what is known about the impact of sex on processes targeted by drugs that are in clinical trial for AD, and existing knowledge on differing responses of males and females to these drugs. Current knowledge strongly supports the view that trials should make assessing sex-related difference in responses a priority with a focus on exploring the sex-stratified treatments.
Collapse
|
12
|
Gonsalves MA, White TL, Barredo J, DeMayo MM, DeLuca E, Harris AD, Carpenter LL. Cortical glutamate, Glx, and total N-acetylaspartate: potential biomarkers of repetitive transcranial magnetic stimulation treatment response and outcomes in major depression. Transl Psychiatry 2024; 14:5. [PMID: 38184652 PMCID: PMC10771455 DOI: 10.1038/s41398-023-02715-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 01/08/2024] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is an effective treatment for individuals with major depressive disorder (MDD) who have not improved with standard therapies. However, only 30-45% of patients respond to rTMS. Predicting response to rTMS will benefit both patients and providers in terms of prescribing and targeting treatment for maximum efficacy and directing resources, as individuals with lower likelihood of response could be redirected to more suitable treatment alternatives. In this exploratory study, our goal was to use proton magnetic resonance spectroscopy to examine how glutamate (Glu), Glx, and total N-acetylaspartate (tNAA) predict post-rTMS changes in overall MDD severity and symptoms, and treatment response. Metabolites were measured in a right dorsal anterior cingulate cortex voxel prior to a standard course of 10 Hz rTMS to the left DLPFC in 25 individuals with MDD. MDD severity and symptoms were evaluated via the Inventory of Depression Symptomatology Self-Report (IDS-SR). rTMS response was defined as ≥50% change in full-scale IDS-SR scores post treatment. Percent change in IDS-SR symptom domains were evaluated using principal component analysis and established subscales. Generalized linear and logistic regression models were used to evaluate the relationship between baseline Glu, Glx, and tNAA and outcomes while controlling for age and sex. Participants with baseline Glu and Glx levels in the lower range had greater percent change in full scale IDS-SR scores post-treatment (p < 0.001), as did tNAA (p = 0.007). Low glutamatergic metabolite levels also predicted greater percent change in mood/cognition symptoms (p ≤ 0.001). Low-range Glu, Glx, and tNAA were associated with greater improvement on the immuno-metabolic subscale (p ≤ 0.003). Baseline Glu predicted rTMS responder status (p = 0.025) and had an area under the receiving operating characteristic curve of 0.81 (p = 0.009), demonstrating excellent discriminative ability. Baseline Glu, Glx, and tNAA significantly predicted MDD improvement after rTMS; preliminary evidence also demonstrates metabolite association with symptom subdomain improvement post-rTMS. This work provides feasibility for a personalized medicine approach to rTMS treatment selection, with individuals with Glu levels in the lower range potentially being the best candidates.
Collapse
Affiliation(s)
- Meghan A Gonsalves
- Neuroscience Graduate Program, Brown University, Providence, RI, USA.
- Butler Hospital Neuromodulation Research Facility, Providence, RI, USA.
- Center of Biomedical Research Excellence (COBRE) for Neuromodulation, Butler Hospital, Providence, RI, USA.
| | - Tara L White
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
- Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, RI, USA
- Carney Institute for Brain Sciences, Brown University, Providence, RI, USA
| | - Jennifer Barredo
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
- Providence VA Medical Center, Providence, RI, USA
- Clinical Neuroimaging Research Core, Brown University, Providence, RI, USA
| | - Marilena M DeMayo
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Emily DeLuca
- Clinical Neuroimaging Research Core, Brown University, Providence, RI, USA
| | - Ashley D Harris
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Linda L Carpenter
- Butler Hospital Neuromodulation Research Facility, Providence, RI, USA
- Center of Biomedical Research Excellence (COBRE) for Neuromodulation, Butler Hospital, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
13
|
Tripathi MK, Ojha SK, Kartawy M, Khaliulin I, Hamoudi W, Amal H. Mutations associated with autism lead to similar synaptic and behavioral alterations in both sexes of male and female mouse brain. Sci Rep 2024; 14:10. [PMID: 38177238 PMCID: PMC10766975 DOI: 10.1038/s41598-023-50248-4] [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: 12/17/2023] [Indexed: 01/06/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder based on synaptic abnormalities. The estimated prevalence rate of male individuals diagnosed with ASD prevails over females is in a proportion of 4:1. Consequently, males remain the main focus in ASD studies in clinical and experimental settings. Meanwhile, some studies point to an underestimation of this disorder in females. In this work, we studied the sex differences of the synaptic and behavioral phenotypes of ASD mouse models. Juvenile male and female Shank3Δ4-22 and Cntnap2-/- mutant mice and their WT littermates were used in the experiments. The animals were subjected to a Three-Chamber Sociability Test, then euthanized, and the whole cortex was used for the evaluation of the synaptic phenotype. Protein levels of glutamatergic (NR1) and GABAergic (GAD1 and VGAT) neuronal markers were measured. Protein level of synaptophysin (Syp) was also measured. Dendritic spine density in somatosensory neurons was analyzed by Golgi staining methods. Spine Density and GAD1, NR1, VGAT, and Syp levels were significantly reduced in Shank3Δ4-22 and Cntnap2-/- mice compared to the control group irrespective of sex, indicating impaired synaptic development in the mutant mice. These results were consistent with the lack of differences in the three-chamber sociability test between male and female mice. In conclusion, female ASD mice of both mutations undergo similar synaptic aberrations as their male counterparts and need to be studied along with the male animals. Finally, this work urges the psychiatry scientific community to use both sexes in their investigations.
Collapse
Affiliation(s)
- Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
14
|
Cubello J, Marvin E, Conrad K, Merrill AK, George JV, Welle K, Jackson BP, Chalupa D, Oberdörster G, Sobolewski M, Cory-Slechta DA. The contributions of neonatal inhalation of copper to air pollution-induced neurodevelopmental outcomes in mice. Neurotoxicology 2024; 100:55-71. [PMID: 38081392 PMCID: PMC10842733 DOI: 10.1016/j.neuro.2023.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Exposures to ambient ultrafine particle (UFP) air pollution (AP) during the early postnatal period in mice (equivalent to human third trimester brain development) produce male-biased changes in brain structure, including ventriculomegaly, reduced brain myelination, alterations in neurotransmitters and glial activation, as well as impulsive-like behavioral characteristics, all of which are also features characteristic of male-biased neurodevelopmental disorders (NDDs). The purpose of this study was to ascertain the extent to which inhaled Cu, a common contaminant of AP that is also dysregulated across multiple NDDs, might contribute to these phenotypes. For this purpose, C57BL/6J mice were exposed from postnatal days 4-7 and 10-13 for 4 hr/day to inhaled copper oxide (CuxOy) nanoparticles at an environmentally relevant concentration averaging 171.9 ng/m3. Changes in brain metal homeostasis and neurotransmitter levels were determined following termination of exposure (postnatal day 14), while behavioral changes were assessed in adulthood. CuxOy inhalation modified cortical metal homeostasis and produced male-biased disruption of striatal neurotransmitters, with marked increases in dopaminergic function, as well as excitatory/inhibitory imbalance and reductions in serotonergic function. Impulsive-like behaviors in a fixed ratio (FR) waiting-for-reward schedule and a fixed interval (FI) schedule of food reward occurred in both sexes, but more prominently in males, effects which could not be attributed to altered locomotor activity or short-term memory. Inhaled Cu as from AP exposures, at environmentally relevant levels experienced during development, may contribute to impaired brain function, as shown by its ability to disrupt brain metal homeostasis and striatal neurotransmission. In addition, its ability to evoke impulsive-like behavior, particularly in male offspring, may be related to striatal dopaminergic dysfunction that is known to mediate such behaviors. As such, regulation of air Cu levels may be protective of public health.
Collapse
Affiliation(s)
- Janine Cubello
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Elena Marvin
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Katherine Conrad
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Alyssa K Merrill
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jithin V George
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kevin Welle
- Proteomics Core, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brian P Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - David Chalupa
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Günter Oberdörster
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
15
|
Johnson CS, Chapp AD, Lind EB, Thomas MJ, Mermelstein PG. Sex differences in mouse infralimbic cortex projections to the nucleus accumbens shell. Biol Sex Differ 2023; 14:87. [PMID: 38082417 PMCID: PMC10712109 DOI: 10.1186/s13293-023-00570-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The nucleus accumbens (NAc) is an important region in motivation and reward. Glutamatergic inputs from the infralimbic cortex (ILC) to the shell region of the NAc (NAcSh) have been implicated in driving the motivation to seek reward through repeated action-based behavior. While this has primarily been studied in males, observed sex differences in motivational circuitry and behavior suggest that females may be more sensitive to rewarding stimuli. These differences have been implicated for the observed vulnerability in women to substance use disorders. METHODS We used an optogenetic self-stimulation task in addition to ex vivo electrophysiological recordings of NAcSh neurons in mouse brain slices to investigate potential sex differences in ILC-NAcSh circuitry in reward-seeking behavior. Glutamatergic neurons in the ILC were infected with an AAV delivering DNA encoding for channelrhodopsin. Entering the designated active corner of an open field arena resulted in photostimulation of the ILC terminals in the NAcSh. Self-stimulation occurred during two consecutive days of testing over three consecutive weeks: first for 10 Hz, then 20 Hz, then 30 Hz. Whole-cell recordings of medium spiny neurons in the NAcSh assessed both optogenetically evoked local field potentials and intrinsic excitability. RESULTS Although both sexes learned to seek the active zone, within the first day, females entered the zone more than males, resulting in a greater amount of photostimulation. Increasing the frequency of optogenetic stimulation amplified female reward-seeking behavior. Males were less sensitive to ILC stimulation, with higher frequencies and repeated days required to increase male reward-seeking behavior. Unexpectedly, ex vivo optogenetic local field potentials in the NAcSh were greater in slices from male animals. In contrast, female medium-spiny neurons (MSNs) displayed significantly greater intrinsic neuronal excitability. CONCLUSIONS Taken together, these data indicate that there are sex differences in the motivated behavior driven by glutamate within the ILC-NAcSh circuit. Though glutamatergic signaling was greater in males, heightened intrinsic excitability in females appears to drive this sex difference.
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
| | - Andrew D Chapp
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Erin B Lind
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Mark J Thomas
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience, School of Medicine, University of Minnesota, 4-140 Jackson Hall, 321 Church St SE, Minneapolis, MN, 55455, USA.
- Medical Discovery Team on Addiction, University of Minnesota, 3-432 McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
16
|
Pengsuwankasem N, Sittiprapaporn P, Rattanabun W, Sangmanee N, Wongsuphasawat K, Rintra J, Nararatwanchai T, Sarikaphuti A, Pandii W. The effect of short daytime napping on cognitive function, sleep quality, and quality of life in mild cognitive impairment patients. Neurosci Lett 2023; 817:137499. [PMID: 37838328 DOI: 10.1016/j.neulet.2023.137499] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/16/2023]
Abstract
Mild cognitive impairment (MCI) is a neurological disorder that can increase the risk of Alzheimer's disease (AD) by three to five times more than those with normal cognition. To better understand the effects of daytime napping on MCI patients, a study was conducted to measure the effects of short naps on cognitive function, sleep quality, and quality of life. In total, 38 participants were asked to take 20-minute naps between 1:00p.m. and 3:00p.m. three times a week for eight weeks. The cognitive function of the participants was measured using the Thai version of the Montreal Cognitive Assessment (Thai-MoCA), their sleep quality was measured using the Thai version of the Pittsburgh Sleep Quality Index (Thai-PSQI), and their quality of life was measured using the Thai version of the Quality of Life (Thai-QoL) questionnaire. After the 8-week period, the participants' scores for the Thai-MoCA and the Thai-QoL questionnaire showed a significant improvement (p < 0.001 for both), while the Thai-PSQI decreased significantly (p = 0.012). This suggests that taking short daytime naps can help to improve the cognitive function, sleep quality, and quality of life of MCI patients.
Collapse
Affiliation(s)
- Nuttapon Pengsuwankasem
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand; Department of Anti-Aging and Regenerative Science, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| | - Phakkharawat Sittiprapaporn
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand; Department of Anti-Aging and Regenerative Science, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| | - Warongporn Rattanabun
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand; Department of Anti-Aging and Regenerative Science, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| | - Nipapan Sangmanee
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand; Department of Anti-Aging and Regenerative Medicine, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| | - Karnt Wongsuphasawat
- Department of Anti-Aging and Regenerative Science, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| | - Jarasphol Rintra
- Department of Anti-Aging and Regenerative Medicine, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| | - Thamthiwat Nararatwanchai
- Department of Anti-Aging and Regenerative Medicine, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| | - Araya Sarikaphuti
- Department of Anti-Aging and Regenerative Science, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| | - Wongdyan Pandii
- Department of Anti-Aging and Regenerative Science, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand.
| |
Collapse
|
17
|
O'Neill J, Diaz MP, Alger JR, Pochon JB, Ghahremani D, Dean AC, Tyndale RF, Petersen N, Marohnic S, Karaiskaki A, London ED. Smoking, tobacco dependence, and neurometabolites in the dorsal anterior cingulate cortex. Mol Psychiatry 2023; 28:4756-4765. [PMID: 37749232 PMCID: PMC10914613 DOI: 10.1038/s41380-023-02247-0] [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: 03/31/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023]
Abstract
Cigarette smoking has a major impact on global health and morbidity, and positron emission tomographic research has provided evidence for reduced inflammation in the human brain associated with cigarette smoking. Given the consequences of inflammatory dysfunction for health, the question of whether cigarette smoking affects neuroinflammation warrants further investigation. The goal of this project therefore was to validate and extend evidence of hypoinflammation related to smoking, and to examine the potential contribution of inflammation to clinical features of smoking. Using magnetic resonance spectroscopy, we measured levels of neurometabolites that are putative neuroinflammatory markers. N-acetyl compounds (N-acetylaspartate + N-acetylaspartylglutamate), glutamate, creatine, choline-compounds (phosphocholine + glycerophosphocholine), and myo-inositol, have all been linked to neuroinflammation, but they have not been examined as such with respect to smoking. We tested whether people who smoke cigarettes have brain levels of these metabolites consistent with decreased neuroinflammation, and whether clinical features of smoking are associated with levels of these metabolites. The dorsal anterior cingulate cortex was chosen as the region-of-interest because of previous evidence linking it to smoking and related states. Fifty-four adults who smoked daily maintained overnight smoking abstinence before testing and were compared with 37 nonsmoking participants. Among the smoking participants, we tested for associations of metabolite levels with tobacco dependence, smoking history, craving, and withdrawal. Levels of N-acetyl compounds and glutamate were higher, whereas levels of creatine and choline compounds were lower in the smoking group as compared with the nonsmoking group. In the smoking group, glutamate and creatine levels correlated negatively with tobacco dependence, and creatine correlated negatively with lifetime smoking, but none of the metabolite levels correlated with craving or withdrawal. The findings indicate a link between smoking and a hypoinflammatory state in the brain, specifically in the dorsal anterior cingulate cortex. Smoking may thereby increase vulnerability to infection and brain injury.
Collapse
Affiliation(s)
- Joseph O'Neill
- Division of Child & Adolescent Psychiatry, Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA
| | - Maylen Perez Diaz
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
- Biogen, Inc., Nashville, TN, USA
| | - Jeffry R Alger
- Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jean-Baptiste Pochon
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Dara Ghahremani
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Andrew C Dean
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Rachel F Tyndale
- Department of Pharmacology & Toxicology, and Department of Psychiatry, University of Toronto, and Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - Nicole Petersen
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Shane Marohnic
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Andrea Karaiskaki
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Edythe D London
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA.
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Pentz AB, Timpe CMF, Normann EM, Slapø NB, Melle I, Lagerberg TV, Steen NE, Westlye LT, Jönsson EG, Haukvik UK, Moberget T, Andreassen OA, Elvsåshagen T. Mismatch negativity in schizophrenia spectrum and bipolar disorders: Group and sex differences and associations with symptom severity. Schizophr Res 2023; 261:80-93. [PMID: 37716205 DOI: 10.1016/j.schres.2023.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/15/2023] [Accepted: 09/04/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Research increasingly implicates glutamatergic dysfunction in the pathophysiologies of psychotic disorders. Auditory mismatch negativity (MMN) is an electroencephalography (EEG) waveform linked to glutamatergic neurotransmission and is consistently attenuated in schizophrenia (SCZ). MMN consists of two subcomponents, the repetition positivity (RP) and deviant negativity (DN) possibly reflecting different neural mechanisms. However, whether MMN reduction is present across different psychotic disorders, linked to distinct symptom clusters, or related to sex remain to be clarified. METHODS Four hundred participants including healthy controls (HCs; n = 296) and individuals with SCZ (n = 39), bipolar disorder (BD) BD typeI (n = 35), or BD type II (n = 30) underwent a roving MMN paradigm and clinical evaluation. MMN, RP and DN as well their memory traces were recorded at the FCZ electrode. Analyses of variance and linear regression models were used both transdiagnostically and within clinical groups. RESULTS MMN was reduced in SCZ compared to BD (p = 0.006, d = 0.55) and to HCs (p < 0.001, d = 0.63). There was a significant group × sex interaction (p < 0.003) and the MMN impairment was only detected in males with SCZ. MMN amplitude correlated positively with Positive and Negative Syndrome Scale total score and negatively with Global Assessment of Functioning Scale score. The deviant negativity was impaired in males with SCZ. No group differences in memory trace indices of the MMN, DN, or RP. CONCLUSION MMN was attenuated in SCZ and correlated with greater severity of psychotic symptoms and lower level of functioning. Our results may indicate sex-dependent differences of glutamatergic function in SCZ.
Collapse
Affiliation(s)
- Atle Bråthen Pentz
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway.
| | - Clara Maria Fides Timpe
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | | | - Nora Berz Slapø
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway
| | - Ingrid Melle
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway
| | - Trine Vik Lagerberg
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway
| | - Nils Eiel Steen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Lars T Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Erik G Jönsson
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Centre for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Sciences, Stockholm Region, Stockholm, Sweden
| | - Unn K Haukvik
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Adult Psychiatry, Institute of Clinical Medicine, University of Oslo, Norway; Department of Forensic Psychiatry Research, Oslo University Hospital, Norway
| | - Torgeir Moberget
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway
| | - Torbjørn Elvsåshagen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
19
|
Dutra-Tavares AC, Souza TP, Silva JO, Semeão KA, Mello FF, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Neonatal phencyclidine as a model of sex-biased schizophrenia symptomatology in adolescent mice. Psychopharmacology (Berl) 2023; 240:2111-2129. [PMID: 37530885 DOI: 10.1007/s00213-023-06434-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/22/2023] [Indexed: 08/03/2023]
Abstract
Sex-biased differences in schizophrenia are evident in several features of the disease, including symptomatology and response to pharmacological treatments. As a neurodevelopmental disorder, these differences might originate early in life and emerge later during adolescence. Considering that the disruption of the glutamatergic system during development is known to contribute to schizophrenia, we hypothesized that the neonatal phencyclidine model could induce sex-dependent behavioral and neurochemical changes associated with this disorder during adolescence. C57BL/6 mice received either saline or phencyclidine (5, 10, or 20 mg/kg) on postnatal days (PN) 7, 9, and 11. Behavioral assessment occurred in late adolescence (PN48-50), when mice were submitted to the open field, social interaction, and prepulse inhibition tests. Either olanzapine or saline was administered before each test. The NMDAR obligatory GluN1 subunit and the postsynaptic density protein 95 (PSD-95) were evaluated in the frontal cortex and hippocampus at early (PN30) and late (PN50) adolescence. Neonatal phencyclidine evoked dose-dependent deficits in all analyzed behaviors and males were more susceptible. Males also had reduced GluN1 expression in the frontal cortex at PN30. There were late-emergent effects at PN50. Cortical GluN1 was increased in both sexes, while phencyclidine increased cortical and decreased hippocampal PSD-95 in females. Olanzapine failed to mitigate most phencyclidine-evoked alterations. In some instances, this antipsychotic aggravated the deficits or potentiated subthreshold effects. These results lend support to the use of neonatal phencyclidine as a sex-biased neurodevelopmental preclinical model of schizophrenia. Olanzapine null effects and deleterious outcomes suggest that its use during adolescence should be further evaluated.
Collapse
Affiliation(s)
- Ana Carolina Dutra-Tavares
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Thainá P Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Juliana O Silva
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Keila A Semeão
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Felipe F Mello
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Claudio C Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores da Universidade do Estado do Rio de Janeiro (UERJ), RJ, São Gonçalo, Brazil
| | - Alex C Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil.
| |
Collapse
|
20
|
Rodríguez-Vega A, Dutra-Tavares AC, Souza TP, Semeão KA, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Nicotine Exposure in a Phencyclidine-Induced Mice Model of Schizophrenia: Sex-Selective Medial Prefrontal Cortex Protein Markers of the Combined Insults in Adolescent Mice. Int J Mol Sci 2023; 24:14634. [PMID: 37834084 PMCID: PMC10572990 DOI: 10.3390/ijms241914634] [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: 07/19/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Tobacco misuse as a comorbidity of schizophrenia is frequently established during adolescence. However, comorbidity markers are still missing. Here, the method of label-free proteomics was used to identify deregulated proteins in the medial prefrontal cortex (prelimbic and infralimbic) of male and female mice modelled to schizophrenia with a history of nicotine exposure during adolescence. Phencyclidine (PCP), used to model schizophrenia (SCHZ), was combined with an established model of nicotine minipump infusions (NIC). The combined insults led to worse outcomes than each insult separately when considering the absolute number of deregulated proteins and that of exclusively deregulated ones. Partially shared Reactome pathways between sexes and between PCP, NIC and PCPNIC groups indicate functional overlaps. Distinctively, proteins differentially expressed exclusively in PCPNIC mice reveal unique effects associated with the comorbidity model. Interactome maps of these proteins identified sex-selective subnetworks, within which some proteins stood out: for females, peptidyl-prolyl cis-trans isomerase (Fkbp1a) and heat shock 70 kDa protein 1B (Hspa1b), both components of the oxidative stress subnetwork, and gamma-enolase (Eno2), a component of the energy metabolism subnetwork; and for males, amphiphysin (Amph), a component of the synaptic transmission subnetwork. These are proposed to be further investigated and validated as markers of the combined insult during adolescence.
Collapse
Affiliation(s)
- Andrés Rodríguez-Vega
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Ana Carolina Dutra-Tavares
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Thainá P. Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Keila A. Semeão
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Claudio C. Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores da Universidade do Estado do Rio de Janeiro, São Gonçalo 24435-005, RJ, Brazil;
| | - Alex C. Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| |
Collapse
|
21
|
Abghari M, Vu JTCM, Eckberg N, Aldana BI, Kohlmeier KA. Decanoic Acid Rescues Differences in AMPA-Mediated Calcium Rises in Hippocampal CA1 Astrocytes and Neurons in the 5xFAD Mouse Model of Alzheimer's Disease. Biomolecules 2023; 13:1461. [PMID: 37892143 PMCID: PMC10604953 DOI: 10.3390/biom13101461] [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: 08/29/2023] [Revised: 09/13/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Alzheimer's disease (AD), a devastating neurodegenerative disease characterized by cognitive dysfunctions, is associated with high levels of amyloid beta 42 (Aβ42), which is believed to play a role in cellular damage and signaling changes in AD. Decanoic acid has been shown to be therapeutic in AD. Glutamatergic signaling within neurons and astrocytes of the CA1 region of the hippocampus is critical in cognitive processes, and previous work has indicated deficiencies in this signaling in a mouse model of AD. In this study, we investigated glutamate-mediated signaling by evaluating AMPA-mediated calcium rises in female and male CA1 neurons and astrocytes in a mouse model of AD and examined the potential of decanoic acid to normalize this signaling. In brain slices from 5xFAD mice in which there are five mutations leading to increasing levels of Aβ42, AMPA-mediated calcium transients in CA1 neurons and astrocytes were significantly lower than that seen in wildtype controls in both females and males. Interestingly, incubation of 5xFAD slices in decanoic acid restored AMPA-mediated calcium levels in neurons and astrocytes in both females and males to levels indistinguishable from those seen in wildtype, whereas similar exposure to decanoic acid did not result in changes in AMPA-mediated transients in neurons or astrocytes in either sex in the wildtype. Our data indicate that one mechanism by which decanoic acid could improve cognitive functioning is through normalizing AMPA-mediated signaling in CA1 hippocampal cells.
Collapse
|
22
|
Birmingham EA, Wickens MM, Kirkland JM, Knouse MC, McGrath AG, Briand LA. Circulating ovarian hormones interact with protein interacting with C kinase (PICK1) within the medial prefrontal cortex to influence cocaine seeking in female mice. Horm Behav 2023; 155:105408. [PMID: 37541099 PMCID: PMC10543586 DOI: 10.1016/j.yhbeh.2023.105408] [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: 03/24/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 08/06/2023]
Abstract
Protein interacting with C kinase 1 (PICK1) is an AMPA receptor binding protein that works in conjunction with glutamate receptor interacting protein (GRIP) to balance the number of GluA2-containing AMPARs in the synapse. In male mice, disrupting PICK1 in the medial prefrontal cortex (mPFC) leads to a decrease in cue-induced cocaine seeking and disrupting GRIP in the mPFC has the opposing effect, consistent with other evidence that removal of GluA2-containing AMPARs potentiates reinstatement. However, PICK1 does not seem to play the same role in female mice, as knockdown of either PICK1 or GRIP in the mPFC leads to similar increases in cue-induced cocaine seeking. These previous findings indicate that the role of PICK1 in the prefrontal cortex is sex specific. The goal of the current study was to examine whether ovarian hormones contribute to the effect of prefrontal PICK1 knockdown on reinstatement of cocaine seeking. While we replicated the increased cue-induced cocaine seeking in prefrontal PICK1 knockdown sham mice, we did not see any difference between the GFP control mice and PICK1 knockdowns following ovariectomy. However, this effect was driven primarily by an increase in cocaine seeking in ovariectomized GFP control mice while there was no effect ovariectomy in PICK1 knockdown mice. Taken together, these findings suggest that circulating ovarian hormones interact with the effects of PICK1 on cue-induced reinstatement.
Collapse
Affiliation(s)
| | - Megan M Wickens
- Department of Psychology & Neuroscience, Temple University, United States of America
| | - Julia M Kirkland
- Department of Psychology & Neuroscience, Temple University, United States of America
| | - Melissa C Knouse
- Department of Psychology & Neuroscience, Temple University, United States of America
| | - Anna G McGrath
- Department of Psychology & Neuroscience, Temple University, United States of America
| | - Lisa A Briand
- Department of Psychology & Neuroscience, Temple University, United States of America; Neuroscience Program, Temple University, United States of America.
| |
Collapse
|
23
|
Knouse MC, Deutschmann AU, Nenov MN, Wimmer ME, Briand LA. Sex differences in pre- and post-synaptic glutamate signaling in the nucleus accumbens core. Biol Sex Differ 2023; 14:52. [PMID: 37596655 PMCID: PMC10439632 DOI: 10.1186/s13293-023-00537-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/08/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Glutamate signaling within the nucleus accumbens underlies motivated behavior and is involved in psychiatric disease. Although behavioral sex differences in these processes are well-established, the neural mechanisms driving these differences are largely unexplored. In these studies, we examine potential sex differences in synaptic plasticity and excitatory transmission within the nucleus accumbens core. Further understanding of baseline sex differences in reward circuitry will shed light on potential mechanisms driving behavioral differences in motivated behavior and psychiatric disease. METHODS Behaviorally naïve adult male and female Long-Evans rats, C57Bl/6J mice, and constitutive PKMζ knockout mice were killed and tissue containing the nucleus accumbens core was collected for ex vivo slice electrophysiology experiments. Electrophysiology recordings examined baseline sex differences in synaptic plasticity and transmission within this region and the potential role of PKMζ in long-term depression. RESULTS Within the nucleus accumbens core, both female mice and rats exhibit higher AMPA/NMDA ratios compared to male animals. Further, female mice have a larger readily releasable pool of glutamate and lower release probability compared to male mice. No significant sex differences were detected in spontaneous excitatory postsynaptic current amplitude or frequency. Finally, the threshold for induction of long-term depression was lower for male animals than females, an effect that appears to be mediated, in part, by PKMζ. CONCLUSIONS We conclude that there are baseline sex differences in synaptic plasticity and excitatory transmission in the nucleus accumbens core. Our data suggest there are sex differences at multiple levels in this region that should be considered in the development of pharmacotherapies to treat psychiatric illnesses such as depression and substance use disorder.
Collapse
Affiliation(s)
- Melissa C Knouse
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Andre U Deutschmann
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Miroslav N Nenov
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Mathieu E Wimmer
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Lisa A Briand
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA.
- Neuroscience Program, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA.
| |
Collapse
|
24
|
Torrisi SA, Rizzo S, Laudani S, Ieraci A, Drago F, Leggio GM. Acute stress alters recognition memory and AMPA/NMDA receptor subunits in a sex-dependent manner. Neurobiol Stress 2023; 25:100545. [PMID: 37293561 PMCID: PMC10244889 DOI: 10.1016/j.ynstr.2023.100545] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Several studies have consistently reported a detrimental effect of chronic stress on recognition memory. However, the effects of acute stress on this cognitive ability have been poorly investigated. Moreover, despite well-documented sex differences in recognition memory observed in clinical studies, most of the preclinical studies in this field of research have been carried out by using solely male rodents. Here we tested the hypothesis that acute stress could affect the consolidation of different types of recognition memory in a sex-dependent manner. For this purpose, male and female C57BL6/J mice were exposed to 2-h of restrain stress immediately after the training session of both the novel object recognition (NOR) test and novel object location (NOL) tasks. Acute restraint stress did not affect memory performance of male and female mice, after a 4-h delay between the training session and the test phase of both tasks. By contrast, acute restraint stress altered memory performance in a sex-dependent manner, after a 24-h delay. While stressed mice of both sexes were impaired in the NOL test, only male stressed mice were impaired in the NOR test. Because ionotropic glutamate receptors-mediated neurotransmission is essential for shaping recognition memory, we further tested the hypothesis that post training acute stress could induce sex-dependent transcriptional changes of ionotropic glutamate receptor subunits in the dorsal hippocampus. We uncovered that acute stress induced sex-, time- and type of memory-dependent transcriptional changes of N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits. These findings suggest that the effect of acute stress on recognition memory can be strongly biased by multiple factors including sex. These findings also indicate that the same stress-induced memory impairment observed in both sexes can be triggered by different sex-dependent molecular mechanisms. At the therapeutic level, this should not be overlooked in the context of personalized and targeted treatments.
Collapse
Affiliation(s)
- Sebastiano A. Torrisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Silvia Rizzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Samuele Laudani
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, 22060, Novedrate, CO, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
25
|
Kamalmaz N, Ben Bacha A, Alonazi M, Albasher G, Khayyat AIA, El-Ansary A. Unveiling sex-based differences in developing propionic acid-induced features in mice as a rodent model of ASD. PeerJ 2023; 11:e15488. [PMID: 37334116 PMCID: PMC10274690 DOI: 10.7717/peerj.15488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background Males are more likely to develop autism as a neurodevelopmental disorder than females are, although the mechanisms underlying male vulnerability are not fully understood. Therefore, studying the role of autism etiologies considering sex differences in the propionic acid (PPA) rodent model of autism would build greater understanding of how females are protected from autism spectrum disorder, which may be used as a treatment strategy for males with autism. Objectives This study aimed to investigate the sex differences in oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut microbiota impairment as etiological mechanisms for many neurological diseases, with specific reference to autism. Method Forty albino mice were divided into four groups of 10 animals each with two control and two treated groups of both sexes received only phosphate-buffered saline or a neurotoxic dose of PPA (250 mg/kg body weight) for 3 days, respectively. Biochemical markers of energy metabolism, oxidative stress, neuroinflammation, and excitotoxicity were measured in mouse brain homogenates, whereas the presence of pathogenic bacteria was assessed in mouse stool samples. Furthermore, the repetitive behavior, cognitive ability, and physical-neural coordination of the animals were examined. Results Collectively, selected variables related to oxidative stress, glutamate excitotoxicity, neuroinflammation, and gut bacteria were impaired concomitantly with altered behavior in PPA-induced rodent model, with males being more susceptible than females. Conclusion This study explains the role of sex in the higher vulnerability of males to develop autistic biochemical and behavioral features compared with females. Female sex hormones and the higher detoxification capacity and higher glycolytic flux in females serve as neuroprotective contributors in a rodent model of autism.
Collapse
Affiliation(s)
- Nasreen Kamalmaz
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Gadah Albasher
- Zoology Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Arwa Ishaq A. Khayyat
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Kutash LA, Moschonas EH, O'Neil DA, Craine TJ, Iouchmanov AL, Sunleaf CR, Nicholas MA, Grobengieser KO, Patel AK, Toader M, Ranellone TS, Rennerfeldt PL, Cheng JP, Race NS, Kline AE, Bondi CO. Sustained attention performance deficits in the three-choice serial reaction time task in male and female rats after experimental brain trauma. Brain Res 2023; 1808:148336. [PMID: 36948353 PMCID: PMC11037439 DOI: 10.1016/j.brainres.2023.148336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
Impaired attention is central to the cognitive deficits associated with long-term sequelae for many traumatic brain injury (TBI) survivors. Assessing complex sustained attention post-TBI is clinically-relevant and may provide reliable avenues towards developing therapeutic and rehabilitation targets in both males and females. We hypothesized that rats subjected to a moderate TBI will exhibit attentional deficits seen as reduced accuracy and increased distractibility in an operant 3-choice serial reaction time task (3-CSRT), designed as an analogue of the clinical continuous performance test. Upon reaching baseline of 70% accuracy at the 300 ms cue, adult male and female Sprague-Dawley rats were subjected to a controlled cortical impact (2.8 mm deformation at 4 m/s) or sham injury over the right parietal cortex. After two weeks of recovery, they were retested on the 3-CSRT for ten days. Dependent measures include percent accuracy (overall and for each of the three cue ports), percent omissions, as well as latency to instrumental poke and retrieve reward. Results demonstrate that both males and females displayed reduced percent accuracy and increased omissions when re-tested post-TBI on 3-CSRT compared to Sham rats and to their own pre-insult baseline (p's < 0.05). Performance accuracy was impaired consistently throughout the ten days of post-surgery re-testing, suggesting pronounced and long-lasting dysfunction in sustained attention processes. Deficits were specifically more pronounced when the cue was pseudorandomly presented in the left-side cue port (p < 0.05), mirroring clinical hemispatial neglect. These data demonstrate significant and persistent complex attention impairments in both sexes after TBI, rendering identifying efficient therapies for cognitive recovery as pivotal.
Collapse
Affiliation(s)
- Lindsay A Kutash
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eleni H Moschonas
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Darik A O'Neil
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy J Craine
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Anna L Iouchmanov
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carlson R Sunleaf
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melissa A Nicholas
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine O Grobengieser
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aarti K Patel
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mihaela Toader
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tyler S Ranellone
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Piper L Rennerfeldt
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey P Cheng
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas S Race
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Association of Academic Physiatrists Rehabilitation Medicine Scientist Training Program, Owings Mills, MD, USA
| | - Anthony E Kline
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Corina O Bondi
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
27
|
Mendez-Vazquez H, Roach RL, Nip K, Chanda S, Sathler MF, Garver T, Danzman RA, Moseley MC, Roberts JP, Koch ON, Steger AA, Lee R, Arikkath J, Kim S. The autism-associated loss of δ-catenin functions disrupts social behavior. Proc Natl Acad Sci U S A 2023; 120:e2300773120. [PMID: 37216537 PMCID: PMC10235948 DOI: 10.1073/pnas.2300773120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 05/01/2023] [Indexed: 05/24/2023] Open
Abstract
δ-catenin is expressed in excitatory synapses and functions as an anchor for the glutamatergic AMPA receptor (AMPAR) GluA2 subunit in the postsynaptic density. The glycine 34 to serine (G34S) mutation in the δ-catenin gene has been found in autism spectrum disorder (ASD) patients and results in loss of δ-catenin functions at excitatory synapses, which is presumed to underlie ASD pathogenesis in humans. However, how the G34S mutation causes loss of δ-catenin functions to induce ASD remains unclear. Here, using neuroblastoma cells, we identify that the G34S mutation increases glycogen synthase kinase 3β (GSK3β)-dependent δ-catenin degradation to reduce δ-catenin levels, which likely contributes to the loss of δ-catenin functions. Synaptic δ-catenin and GluA2 levels in the cortex are significantly decreased in mice harboring the δ-catenin G34S mutation. The G34S mutation increases glutamatergic activity in cortical excitatory neurons while it is decreased in inhibitory interneurons, indicating changes in cellular excitation and inhibition. δ-catenin G34S mutant mice also exhibit social dysfunction, a common feature of ASD. Most importantly, pharmacological inhibition of GSK3β activity reverses the G34S-induced loss of δ-catenin function effects in cells and mice. Finally, using δ-catenin knockout mice, we confirm that δ-catenin is required for GSK3β inhibition-induced restoration of normal social behavior in δ-catenin G34S mutant animals. Taken together, we reveal that the loss of δ-catenin functions arising from the ASD-associated G34S mutation induces social dysfunction via alterations in glutamatergic activity and that GSK3β inhibition can reverse δ-catenin G34S-induced synaptic and behavioral deficits.
Collapse
Affiliation(s)
| | - Regan L. Roach
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| | - Kaila Nip
- Cellular and Molecular Biology Program, Colorado State UniversityFort CollinsCO80523
| | - Soham Chanda
- Cellular and Molecular Biology Program, Colorado State UniversityFort CollinsCO80523
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, CO80523
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO80523
| | - Matheus F. Sathler
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| | - Tyler Garver
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, CO80523
| | - Rosaline A. Danzman
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| | - Madeleine C. Moseley
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, CO80523
| | - Jessica P. Roberts
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, CO80523
| | - Olivia N. Koch
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| | | | - Rahmi Lee
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| | - Jyothi Arikkath
- Developmental Neuroscience, Munore-Meyer Institute, University of Nebraska Medical Center, Omaha, NE68198
| | - Seonil Kim
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
- Cellular and Molecular Biology Program, Colorado State UniversityFort CollinsCO80523
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, CO80523
| |
Collapse
|
28
|
Bölte S, Neufeld J, Marschik PB, Williams ZJ, Gallagher L, Lai MC. Sex and gender in neurodevelopmental conditions. Nat Rev Neurol 2023; 19:136-159. [PMID: 36747038 PMCID: PMC10154737 DOI: 10.1038/s41582-023-00774-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2023] [Indexed: 02/08/2023]
Abstract
Health-related conditions often differ qualitatively or quantitatively between individuals of different birth-assigned sexes and gender identities, and/or with different gendered experiences, requiring tailored care. Studying the moderating and mediating effects of sex-related and gender-related factors on impairment, disability, wellbeing and health is of paramount importance especially for neurodivergent individuals, who are diagnosed with neurodevelopmental conditions with uneven sex/gender distributions. Researchers have become aware of the myriad influences that sex-related and gender-related variables have on the manifestations of neurodevelopmental conditions, and contemporary work has begun to investigate the mechanisms through which these effects are mediated. Here we describe topical concepts of sex and gender science, summarize current knowledge, and discuss research and clinical challenges related to autism, attention-deficit/hyperactivity disorder and other neurodevelopmental conditions. We consider sex and gender in the context of epidemiology, behavioural phenotypes, neurobiology, genetics, endocrinology and neighbouring disciplines. The available evidence supports the view that sex and gender are important contributors to the biological and behavioural variability in neurodevelopmental conditions. Methodological caveats such as frequent conflation of sex and gender constructs, inappropriate measurement of these constructs and under-representation of specific demographic groups (for example, female and gender minority individuals and people with intellectual disabilities) limit the translational potential of research so far. Future research and clinical implementation should integrate sex and gender into next-generation diagnostics, mechanistic investigations and support practices.
Collapse
Affiliation(s)
- Sven Bölte
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research; Department of Women's and Children's Health, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.
- Child and Adolescent Psychiatry, Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.
- Curtin Autism Research Group, Curtin School of Allied Health, Curtin University, Perth, WA, Australia.
| | - Janina Neufeld
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research; Department of Women's and Children's Health, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
- Swedish Collegium for Advanced Study (SCAS), Uppsala, Sweden
| | - Peter B Marschik
- Center of Neurodevelopmental Disorders (KIND), Centre for Psychiatry Research; Department of Women's and Children's Health, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen and Leibniz ScienceCampus Primate Cognition, Göttingen, Germany
- iDN - interdisciplinary Developmental Neuroscience, Division of Phoniatrics, Medical University of Graz, Graz, Austria
| | - Zachary J Williams
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Frist Center for Autism and Innovation, Vanderbilt University, Nashville, TN, USA
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Child and Youth Mental Health Collaborative at the Centre for Addiction and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, and Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Meng-Chuan Lai
- Child and Youth Mental Health Collaborative at the Centre for Addiction and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, and Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK.
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.
| |
Collapse
|
29
|
Jalewa J, Todd J, Michie PT, Hodgson DM, Harms L. The effect of schizophrenia risk factors on mismatch responses in a rat model. Psychophysiology 2023; 60:e14175. [PMID: 36087044 PMCID: PMC10909418 DOI: 10.1111/psyp.14175] [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: 12/21/2021] [Revised: 06/28/2022] [Accepted: 08/04/2022] [Indexed: 01/06/2023]
Abstract
Reduced mismatch negativity (MMN), a robust finding in schizophrenia, has prompted interest in MMN as a preclinical biomarker of schizophrenia. The rat brain can generate human-like mismatch responses (MMRs) which therefore enables the exploration of the neurobiology of reduced MMRs. Given epidemiological evidence that two developmental factors, maternal infection and adolescent cannabis use, increase the risk of schizophrenia, we determined the effect of these two developmental risk factors on rat MMR amplitude in different auditory contexts. MMRs were assessed in awake adult male and female Wistar rats that were offspring of pregnant dams treated with either a viral infection mimetic (poly I:C) inducing maternal immune activation (MIA) or saline control. In adolescence, subgroups of the prenatal treatment groups were exposed to either a synthetic cannabinoid (adolescent cannabinoid exposure: ACE) or vehicle. The context under which MMRs were obtained was manipulated by employing two different oddball paradigms, one that manipulated the physical difference between rare and common auditory stimuli, and another that manipulated the probability of the rare stimulus. The design of the multiple stimulus sequences across the two paradigms also allowed an investigation of context on MMRs to two identical stimulus sequences. Male offspring exposed to each of the risk factors for schizophrenia (MIA, ACE or both) showed a reduction in MMR, which was evident only in the probability paradigm, with no effects seen in the physical difference. Our findings highlight the importance of contextual factors induced by paradigm manipulations and sex for modeling schizophrenia-like MMN impairments in rats.
Collapse
Affiliation(s)
- Jaishree Jalewa
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Juanita Todd
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Patricia T. Michie
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Deborah M. Hodgson
- School of Psychological Sciences, College of Engineering, Science and EnvironmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Lauren Harms
- Hunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
- School of Biomedical Science and Pharmacy, College of Health, Medicine and WellbeingUniversity of NewcastleCallaghanNew South WalesAustralia
| |
Collapse
|
30
|
Quintrell E, Wyrwoll C, Rosenow T, Larcombe A, Kelty E. The effects of acamprosate on maternal and neonatal outcomes in a mouse model of alcohol use disorders. Physiol Behav 2023; 259:114037. [PMID: 36427542 DOI: 10.1016/j.physbeh.2022.114037] [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: 09/26/2022] [Revised: 11/07/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Despite the teratogenic effects of alcohol, little is known about the safety of pharmacotherapies such as acamprosate for the treatment of alcohol use disorders in pregnancy. The aims of this study were to investigate, in a mouse model, the effects of maternally administered acamprosate on maternal and neonatal health, offspring neurodevelopment and behaviour, as well as examine whether acamprosate reduces the neurological harm associated with alcohol consumption in pregnancy. METHODS Dams were randomly allocated to one of four treatment groups: (i) control (water), (ii) acamprosate (1.6 g/L), (iii) alcohol (5% v/v) or (iv) acamprosate and alcohol (1.6 g/L; 5% v/v ethanol) and exposed from 2-weeks pre-pregnancy until postpartum day 7. Gestational outcomes including litter size and sex ratio were assessed, in addition to early-life markers of neurodevelopment. At 8 weeks of age, motor coordination, anxiety, locomotion, and memory of the adult offspring were also examined. RESULTS Exposure to acamprosate did not affect maternal and birth outcomes (mating success, gestational weight gain, litter size, sex ratio), neonatal outcomes (head and body length, postnatal weight) or neurodevelopmental markers (righting reflex and negative geotaxis). Acamprosate exposure did not affect offspring motor control, locomotion or anxiety, however the effects on short-term memory remain uncertain. Prenatal alcohol exposed offspring exhibited various alterations, such as lower postnatal weight, smaller head (p = 0.04) and body lengths (p = 0.046) at postnatal day 70 (males only), increased negative geotaxis speed (p = 0.03), an increased time spent in the inner zone of the open field (p = 0.02). Acamprosate mitigated the effects of alcohol for negative geotaxis at postnatal day 7 (p = 0.01) and female offspring weight at postnatal day 70 (p = 0.03). CONCLUSIONS Overall, we show that prenatal acamprosate exposure was not associated with poor maternal or neonatal health outcomes or impaired neurodevelopment and behaviour. However, acamprosate's effects on short-term memory remain uncertain. We present preliminary evidence to suggest acamprosate displayed some neuroprotective effects against damage caused by in utero alcohol exposure.
Collapse
Affiliation(s)
- Ebony Quintrell
- School of Population and Global Health, University of Western Australia, Nedlands, Western Australia Australia; Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
| | - Caitlin Wyrwoll
- Telethon Kids Institute, Nedlands, Western Australia, Australia; School of Human Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Tim Rosenow
- The Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Alexander Larcombe
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia; Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, Western Australia, Australia
| | - Erin Kelty
- School of Population and Global Health, University of Western Australia, Nedlands, Western Australia Australia.
| |
Collapse
|
31
|
Ochocki AJ, Kenney JW. A gelatin-based feed for precise and non-invasive drug delivery to adult zebrafish. J Exp Biol 2023; 226:286278. [PMID: 36606734 PMCID: PMC10165467 DOI: 10.1242/jeb.245186] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/25/2022] [Indexed: 01/07/2023]
Abstract
Although the use of adult zebrafish as a model organism has increased in recent years, there is room to refine methods, such as drug delivery, to make them less invasive and more precise. Here, we describe the development of a non-invasive gelatin-based feed method that is tailored to animals based on their body mass. The feed was readily eaten by zebrafish (<1 min) with minimal leaching of compound when placed in water (<5% in 5 min). As a proof of principle, we fed fish a NMDA receptor antagonist (MK-801, 4 mg kg-1) prior to the novel tank test. We found that MK-801 caused a general decrease in predator-avoidance/anxiety-like behavior (bottom dwelling) and an increase in locomotion in male fish, but not females. Our simple, easy to prepare and individually tailored gelatin-based feed enables precisely dosed, non-invasive drug delivery to adult-stage zebrafish for the first time.
Collapse
Affiliation(s)
- Aleksander J Ochocki
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Justin W Kenney
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
32
|
Mendez-Vazquez H, Roach RL, Nip K, Sathler MF, Garver T, Danzman RA, Moseley MC, Roberts JP, Koch ON, Steger AA, Lee R, Arikkath J, Kim S. The autism-associated loss of δ-catenin functions disrupts social behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523372. [PMID: 36711484 PMCID: PMC9882145 DOI: 10.1101/2023.01.12.523372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
δ-catenin is expressed in excitatory synapses and functions as an anchor for the glutamatergic AMPA receptor (AMPAR) GluA2 subunit in the postsynaptic density. The glycine 34 to serine (G34S) mutation in the δ-catenin gene is found in autism spectrum disorder (ASD) patients and induces loss of δ-catenin functions at excitatory synapses, which is presumed to underlie ASD pathogenesis in humans. However, how the G34S mutation causes loss of δ-catenin functions to induce ASD remains unclear. Here, using neuroblastoma cells, we discover that the G34S mutation generates an additional phosphorylation site for glycogen synthase kinase 3β (GSK3β). This promotes δ-catenin degradation and causes the reduction of δ-catenin levels, which likely contributes to the loss of δ-catenin functions. Synaptic δ-catenin and GluA2 levels in the cortex are significantly decreased in mice harboring the δ-catenin G34S mutation. The G34S mutation increases glutamatergic activity in cortical excitatory neurons while it is decreased in inhibitory interneurons, indicating changes in cellular excitation and inhibition. δ-catenin G34S mutant mice also exhibit social dysfunction, a common feature of ASD. Most importantly, inhibition of GSK3β activity reverses the G34S-induced loss of δ-catenin function effects in cells and mice. Finally, using δ-catenin knockout mice, we confirm that δ-catenin is required for GSK3β inhibition-induced restoration of normal social behaviors in δ-catenin G34S mutant animals. Taken together, we reveal that the loss of δ-catenin functions arising from the ASD-associated G34S mutation induces social dysfunction via alterations in glutamatergic activity and that GSK3β inhibition can reverse δ-catenin G34S-induced synaptic and behavioral deficits. Significance Statement δ-catenin is important for the localization and function of glutamatergic AMPA receptors at synapses in many brain regions. The glycine 34 to serine (G34S) mutation in the δ-catenin gene is found in autism patients and results in the loss of δ-catenin functions. δ-catenin expression is also closely linked to other autism-risk genes involved in synaptic structure and function, further implying that it is important for the autism pathophysiology. Importantly, social dysfunction is a key characteristic of autism. Nonetheless, the links between δ-catenin functions and social behaviors are largely unknown. The significance of the current research is thus predicated on filling this gap by discovering the molecular, cellular, and synaptic underpinnings of the role of δ-catenin in social behaviors.
Collapse
|
33
|
Harris AD, Amiri H, Bento M, Cohen R, Ching CRK, Cudalbu C, Dennis EL, Doose A, Ehrlich S, Kirov II, Mekle R, Oeltzschner G, Porges E, Souza R, Tam FI, Taylor B, Thompson PM, Quidé Y, Wilde EA, Williamson J, Lin AP, Bartnik-Olson B. Harmonization of multi-scanner in vivo magnetic resonance spectroscopy: ENIGMA consortium task group considerations. Front Neurol 2023; 13:1045678. [PMID: 36686533 PMCID: PMC9845632 DOI: 10.3389/fneur.2022.1045678] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
Magnetic resonance spectroscopy is a powerful, non-invasive, quantitative imaging technique that allows for the measurement of brain metabolites that has demonstrated utility in diagnosing and characterizing a broad range of neurological diseases. Its impact, however, has been limited due to small sample sizes and methodological variability in addition to intrinsic limitations of the method itself such as its sensitivity to motion. The lack of standardization from a data acquisition and data processing perspective makes it difficult to pool multiple studies and/or conduct multisite studies that are necessary for supporting clinically relevant findings. Based on the experience of the ENIGMA MRS work group and a review of the literature, this manuscript provides an overview of the current state of MRS data harmonization. Key factors that need to be taken into consideration when conducting both retrospective and prospective studies are described. These include (1) MRS acquisition issues such as pulse sequence, RF and B0 calibrations, echo time, and SNR; (2) data processing issues such as pre-processing steps, modeling, and quantitation; and (3) biological factors such as voxel location, age, sex, and pathology. Various approaches to MRS data harmonization are then described including meta-analysis, mega-analysis, linear modeling, ComBat and artificial intelligence approaches. The goal is to provide both novice and experienced readers with the necessary knowledge for conducting MRS data harmonization studies.
Collapse
Affiliation(s)
- Ashley D. Harris
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Houshang Amiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mariana Bento
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Ronald Cohen
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Christopher R. K. Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, Los Angeles, CA, United States
| | - Christina Cudalbu
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Emily L. Dennis
- TBI and Concussion Center, Department of Neurology, University of Utah, Salt Lake City, UT, United States
| | - Arne Doose
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ivan I. Kirov
- Department of Radiology, Center for Advanced Imaging Innovation and Research, New York University Grossman School of Medicine, New York, NY, United States
| | - Ralf Mekle
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Eric Porges
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Roberto Souza
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Electrical and Software Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Friederike I. Tam
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Brian Taylor
- Division of Diagnostic Imaging, Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, Los Angeles, CA, United States
| | - Yann Quidé
- School of Psychology, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Elisabeth A. Wilde
- TBI and Concussion Center, Department of Neurology, University of Utah, Salt Lake City, UT, United States
| | - John Williamson
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Alexander P. Lin
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Brenda Bartnik-Olson
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, United States
| |
Collapse
|
34
|
Fabian CB, Seney ML, Joffe ME. Sex differences and hormonal regulation of metabotropic glutamate receptor synaptic plasticity. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 168:311-347. [PMID: 36868632 PMCID: PMC10392610 DOI: 10.1016/bs.irn.2022.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Striking sex differences exist in presentation and incidence of several psychiatric disorders. For example, major depressive disorder is more prevalent in women than men, and women who develop alcohol use disorder progress through drinking milestones more rapidly than men. With regards to psychiatric treatment responses, women respond more favorably to selective serotonin reuptake inhibitors than men, whereas men have better outcomes when prescribed tricyclic antidepressants. Despite such well-documented biases in incidence, presentation, and treatment response, sex as a biological variable has long been neglected in preclinical and clinical research. An emerging family of druggable targets for psychiatric diseases, metabotropic glutamate (mGlu) receptors are G-protein coupled receptors broadly distributed throughout the central nervous system. mGlu receptors confer diverse neuromodulatory actions of glutamate at the levels of synaptic plasticity, neuronal excitability, and gene transcription. In this chapter, we summarize the current preclinical and clinical evidence for sex differences in mGlu receptor function. We first highlight basal sex differences in mGlu receptor expression and function and proceed to describe how gonadal hormones, notably estradiol, regulate mGlu receptor signaling. We then describe sex-specific mechanisms by which mGlu receptors differentially modulate synaptic plasticity and behavior in basal states and models relevant for disease. Finally, we discuss human research findings and highlight areas in need of further research. Taken together, this review emphasizes how mGlu receptor function and expression can differ across sex. Gaining a more complete understanding of how sex differences in mGlu receptor function contribute to psychiatric diseases will be critical in the development of novel therapeutics that are effective in all individuals.
Collapse
Affiliation(s)
- Carly B Fabian
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - Marianne L Seney
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - Max E Joffe
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
35
|
Knouse MC, McGrath AG, Deutschmann AU, Rich MT, Zallar LJ, Rajadhyaksha AM, Briand LA. Sex differences in the medial prefrontal cortical glutamate system. Biol Sex Differ 2022; 13:66. [PMID: 36348414 PMCID: PMC9641904 DOI: 10.1186/s13293-022-00468-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dysregulation in the prefrontal cortex underlies a variety of psychiatric illnesses, including substance use disorder, depression, and anxiety. Despite the established sex differences in prevalence and presentation of these illnesses, the neural mechanisms driving these differences are largely unexplored. Here, we investigate potential sex differences in glutamatergic transmission within the medial prefrontal cortex (mPFC). The goal of these experiments was to determine if there are baseline sex differences in transmission within this region that may underlie sex differences in diseases that involve dysregulation in the prefrontal cortex. METHODS Adult male and female C57Bl/6J mice were used for all experiments. Mice were killed and bilateral tissue samples were taken from the medial prefrontal cortex for western blotting. Both synaptosomal and total GluA1 and GluA2 levels were measured. In a second set of experiments, mice were killed and ex vivo slice electrophysiology was performed on prepared tissue from the medial prefrontal cortex. Spontaneous excitatory postsynaptic currents and rectification indices were measured. RESULTS Females exhibit higher levels of synaptosomal GluA1 and GluA2 in the mPFC compared to males. Despite similar trends, no statistically significant differences are seen in total levels of GluA1 and GluA2. Females also exhibit both a higher amplitude and higher frequency of spontaneous excitatory postsynaptic currents and greater inward rectification in the mPFC compared to males. CONCLUSIONS Overall, we conclude that there are sex differences in glutamatergic transmission in the mPFC. Our data suggest that females have higher levels of glutamatergic transmission in this region. This provides evidence that the development of sex-specific pharmacotherapies for various psychiatric diseases is important to create more effective treatments.
Collapse
Affiliation(s)
- Melissa C. Knouse
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA 19122 USA
| | - Anna G. McGrath
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA 19122 USA
| | - Andre U. Deutschmann
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA 19122 USA
| | - Matthew T. Rich
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Lia J. Zallar
- Department of Pharmacology, Weill Cornell Medicine of Cornell University, New York, NY USA
| | - Anjali M. Rajadhyaksha
- Pediatric Neurology, Pediatrics, Weill Cornell Medicine of Cornell University, New York, NY USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine of Cornell University, New York, NY USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine of Cornell University, New York, NY USA
| | - Lisa A. Briand
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA 19122 USA
- Neuroscience Program, Temple University, Philadelphia, USA
| |
Collapse
|
36
|
Maffioli E, Murtas G, Rabattoni V, Badone B, Tripodi F, Iannuzzi F, Licastro D, Nonnis S, Rinaldi AM, Motta Z, Sacchi S, Canu N, Tedeschi G, Coccetti P, Pollegioni L. Insulin and serine metabolism as sex-specific hallmarks of Alzheimer's disease in the human hippocampus. Cell Rep 2022; 40:111271. [PMID: 36070700 DOI: 10.1016/j.celrep.2022.111271] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 07/01/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022] Open
Abstract
Healthy aging is an ambitious aspiration for humans, but neurodegenerative disorders, such as Alzheimer's disease (AD), strongly affect quality of life. Using an integrated omics approach, we investigate alterations in the molecular composition of postmortem hippocampus samples of healthy persons and individuals with AD. Profound differences are apparent between control and AD male and female cohorts in terms of up- and downregulated metabolic pathways. A decrease in the insulin response is evident in AD when comparing the female with the male group. The serine metabolism (linked to the glycolytic pathway and generating the N-methyl-D-aspartate [NMDA] receptor coagonist D-serine) is also significantly modulated: the D-Ser/total serine ratio represents a way to counteract age-related cognitive decline in healthy men and during AD onset in women. These results show how AD changes and, in certain respects, almost reverses sex-specific proteomic and metabolomic profiles, highlighting how different pathophysiological mechanisms are active in men and women.
Collapse
Affiliation(s)
- Elisa Maffioli
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, 20121 Milano, Italy; CIMAINA, University of Milano, 20121 Milano, Italy
| | - Giulia Murtas
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Valentina Rabattoni
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Beatrice Badone
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy
| | - Farida Tripodi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy
| | - Filomena Iannuzzi
- Department of System Medicine, University of Rome "Tor Vergata," 00133 Rome, Italy
| | | | - Simona Nonnis
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, 20121 Milano, Italy; CIMAINA, University of Milano, 20121 Milano, Italy
| | - Anna Maria Rinaldi
- Department of System Medicine, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Zoraide Motta
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Silvia Sacchi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Nadia Canu
- Department of System Medicine, University of Rome "Tor Vergata," 00133 Rome, Italy; Istituto di Biochimica e Biologia Cellulare (IBBC) CNR, 00015 Monterotondo Scalo, Italy.
| | - Gabriella Tedeschi
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, 20121 Milano, Italy; CIMAINA, University of Milano, 20121 Milano, Italy.
| | - Paola Coccetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy.
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy.
| |
Collapse
|
37
|
Shanmugan S, Seidlitz J, Cui Z, Adebimpe A, Bassett DS, Bertolero MA, Davatzikos C, Fair DA, Gur RE, Gur RC, Larsen B, Li H, Pines A, Raznahan A, Roalf DR, Shinohara RT, Vogel J, Wolf DH, Fan Y, Alexander-Bloch A, Satterthwaite TD. Sex differences in the functional topography of association networks in youth. Proc Natl Acad Sci U S A 2022; 119:e2110416119. [PMID: 35939696 PMCID: PMC9388107 DOI: 10.1073/pnas.2110416119] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/15/2022] [Indexed: 01/16/2023] Open
Abstract
Prior work has shown that there is substantial interindividual variation in the spatial distribution of functional networks across the cerebral cortex, or functional topography. However, it remains unknown whether there are sex differences in the topography of individualized networks in youth. Here, we leveraged an advanced machine learning method (sparsity-regularized non-negative matrix factorization) to define individualized functional networks in 693 youth (ages 8 to 23 y) who underwent functional MRI as part of the Philadelphia Neurodevelopmental Cohort. Multivariate pattern analysis using support vector machines classified participant sex based on functional topography with 82.9% accuracy (P < 0.0001). Brain regions most effective in classifying participant sex belonged to association networks, including the ventral attention, default mode, and frontoparietal networks. Mass univariate analyses using generalized additive models with penalized splines provided convergent results. Furthermore, transcriptomic data from the Allen Human Brain Atlas revealed that sex differences in multivariate patterns of functional topography were spatially correlated with the expression of genes on the X chromosome. These results highlight the role of sex as a biological variable in shaping functional topography.
Collapse
Affiliation(s)
- Sheila Shanmugan
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Jakob Seidlitz
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Zaixu Cui
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Chinese Institute for Brain Research, Beijing,102206, China
| | - Azeez Adebimpe
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Danielle S. Bassett
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104
- Santa Fe Institute, Santa Fe, NM 87501
| | - Maxwell A. Bertolero
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Christos Davatzikos
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Damien A. Fair
- Department of Behavioral Neuroscience, Department of Psychiatry, Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR 97239
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Hongming Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Adam Pines
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Armin Raznahan
- Section on Developmental Neurogenomics Unit, Intramural Research Program, National Institutes of Mental Health, Bethesda, MD 20892
| | - David R. Roalf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Russell T. Shinohara
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104
| | - Jacob Vogel
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Daniel H. Wolf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| | - Aaron Alexander-Bloch
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Theodore D. Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104
- Penn-Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104
- Center for Biomedical Image Computation and Analytics, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
38
|
Winek K, Tzur Y, Soreq H. Biological underpinnings of sex differences in neurological disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 164:27-67. [PMID: 36038206 DOI: 10.1016/bs.irn.2022.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The importance of sex differences in neurological disorders has been increasingly acknowledged in recent clinical and basic research studies, but the complex biology and genetics underlying sex-linked biological heterogeneity and its brain-to-body impact remained incompletely understood. Men and women differ substantially in their susceptibility to certain neurological diseases, in the severity of symptoms, prognosis as well as the nature and efficacy of their response to treatments. The detailed mechanisms underlying these differences, especially at the molecular level, are being addressed in many studies but leave a lot to be further revealed. Here, we provide an overview of recent advances in our understanding of how sex differences in the brain and brain-body signaling contribute to neurological disorders and further present some future prospects entailed in terms of diagnostics and therapeutics.
Collapse
Affiliation(s)
- Katarzyna Winek
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yonat Tzur
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hermona Soreq
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
39
|
Abstract
Brain disease remains a significant health, social, and economic burden with a high failure rate of translation of therapeutics to the clinic. Nanotherapeutics have represented a promising area of technology investment to improve drug bioavailability and delivery to the brain, with several successes for nanotherapeutic use for central nervous system disease that are currently in the clinic. However, renewed and continued research on the treatment of neurological disorders is critically needed. We explore the challenges of drug delivery to the brain and the ways in which nanotherapeutics can overcome these challenges. We provide a summary and overview of general design principles that can be applied to nanotherapeutics for uptake and penetration in the brain. We next highlight remaining questions that limit the translational potential of nanotherapeutics for application in the clinic. Lastly, we provide recommendations for ongoing preclinical research to improve the overall success of nanotherapeutics against neurological disease.
Collapse
Affiliation(s)
- Andrea Joseph
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, Washington, USA;
| |
Collapse
|
40
|
Krentzel AA, Proaño SB, Dorris DM, Setzer B, Meitzen J. The estrous cycle and 17β-estradiol modulate the electrophysiological properties of rat nucleus accumbens core medium spiny neurons. J Neuroendocrinol 2022; 34:e13122. [PMID: 35365910 PMCID: PMC9250601 DOI: 10.1111/jne.13122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 12/03/2022]
Abstract
The nucleus accumbens core is a key nexus within the mammalian brain for integrating the premotor and limbic systems and regulating important cognitive functions such as motivated behaviors. Nucleus accumbens core functions show sex differences and are sensitive to the presence of hormones such as 17β-estradiol (estradiol) in normal and pathological contexts. The primary neuron type of the nucleus accumbens core, the medium spiny neuron (MSN), exhibits sex differences in both intrinsic excitability and glutamatergic excitatory synapse electrophysiological properties. Here, we provide a review of recent literature showing how estradiol modulates rat nucleus accumbens core MSN electrophysiology within the context of the estrous cycle. We review the changes in MSN electrophysiological properties across the estrous cycle and how these changes can be mimicked in response to exogenous estradiol exposure. We discuss in detail recent findings regarding how acute estradiol exposure rapidly modulates excitatory synapse properties in nucleus accumbens core but not caudate-putamen MSNs, which mirror the natural changes seen across estrous cycle phases. These recent insights demonstrate the strong impact of sex-specific estradiol action upon nucleus accumbens core neuron electrophysiology.
Collapse
Affiliation(s)
- Amanda A. Krentzel
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Stephanie B. Proaño
- Neurobiology LaboratoryNational Institute of Environmental Health Sciences, NIHResearch Triangle ParkNCUSA
| | - David M. Dorris
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Beverly Setzer
- Graduate Program for Neuroscience and Department of Biomedical EngineeringBoston UniversityBostonMAUSA
| | - John Meitzen
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNCUSA
- Center for Human Health and the EnvironmentNorth Carolina State UniversityRaleighNCUSA
| |
Collapse
|
41
|
Ganci M, Suleyman E, Butt H, Ball M. Associations between self-reported psychological symptom severity and gut microbiota: further support for the microgenderome. BMC Psychiatry 2022; 22:307. [PMID: 35501777 PMCID: PMC9059404 DOI: 10.1186/s12888-022-03947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Research into the brain-gut-microbiota axis (BGMA) continues to reveal associations between gut microbiota (GM) and psychological symptom expression, inspiring new ways of conceptualising psychological disorders. However, before GM modulation can be touted as a possible auxiliary treatment option, more research is needed as inconsistencies in previous findings regarding these associations are prevalent. Additionally, the concept of the microgenderome, which proposes that GM may interact with sex hormones, has received limited attention in studies using human samples to date. However, such research has demonstrated sex specific associations between GM and psychological symptom expression. METHOD This cross-sectional retrospective study explores associations between GM species (identified through faecal microbial analysis) and symptom severity across four psychological domains (Depressive, Neurocognitive, Stress and Anxiety, and Sleep and Fatigue) for males (N = 1143) and females (N = 3467) separately. RESULTS GM species from several genera including Bifidobacterium, Clostridium, Enterococcus, and Leuconostoc were found to be differentially associated with psychological symptom severity for males and females. As such, the findings of the current study provide support for the concept of the microgenderome. CONCLUSION While further research is needed before their implementation in psychological treatment plans, the current findings suggest that modulation of GM at the species level may hold promise as auxiliary diagnostic or treatment options. These findings may give further insight into a client's presenting problem from a more holistic, multidisciplinary perspective. The clear sex divergence in associations between GM and symptoms give insight into sex discrepancies in susceptibility to psychological disorders.
Collapse
Affiliation(s)
- Michael Ganci
- Psychology Department, Institute for Health and Sport, Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia.
| | - Emra Suleyman
- grid.1019.90000 0001 0396 9544Psychology Department, Institute for Health and Sport, Victoria University, PO Box 14428, Melbourne, VIC 8001 Australia
| | - Henry Butt
- grid.1019.90000 0001 0396 9544Psychology Department, Institute for Health and Sport, Victoria University, PO Box 14428, Melbourne, VIC 8001 Australia ,Bioscreen Yarraville (Aust) Pty Ltd, Melbourne, VIC Australia
| | - Michelle Ball
- grid.1019.90000 0001 0396 9544Psychology Department, Institute for Health and Sport, Victoria University, PO Box 14428, Melbourne, VIC 8001 Australia
| |
Collapse
|
42
|
Kara F, Joers JM, Deelchand DK, Park YW, Przybelski SA, Lesnick TG, Senjem ML, Zeydan B, Knopman DS, Lowe VJ, Vemuri P, Mielke MM, Machulda MM, Jack CR, Petersen RC, Öz G, Kantarci K. 1H MR spectroscopy biomarkers of neuronal and synaptic function are associated with tau deposition in cognitively unimpaired older adults. Neurobiol Aging 2022; 112:16-26. [PMID: 35038671 PMCID: PMC8976711 DOI: 10.1016/j.neurobiolaging.2021.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 12/25/2022]
Abstract
Proton magnetic resonance spectroscopy (1H MRS) may provide information on pathophysiological changes associated with tau deposition in cognitively unimpaired older adults. In this study, the associations of posterior cingulate gyrus tau and amyloid beta (Aβ) deposition on PET with 1H MRS metabolite ratios acquired from bilateral posterior cingulate gyri were investigated in cognitively unimpaired older adults. Participants (n = 40) from the Mayo Clinic Study of Aging underwent single-voxel sLASER 1H MRS from the posterior cingulate gyrus at 3 Tesla, 18F-flortaucipir, and 11C- Pittsburgh Compound B (PiB) PET. An increase in posterior cingulate gyrus tau deposition, but not elevated Aβ, was associated with lower N-acetylaspartate/total creatine (tCr) and glutamate (Glu)/tCr ratios, and sex by tau interaction was observed in association with Glu/tCr. Higher tau levels in cognitively unimpaired older adults are associated with biomarkers of neural and synaptic injury even in the absence of cognitive impairment and these relationships appear to be stronger in women than in men.
Collapse
Affiliation(s)
- Firat Kara
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - James M Joers
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Dinesh K Deelchand
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Young Woo Park
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Burcu Zeydan
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | | | | | - Gülin Öz
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
43
|
Montanari M, Martella G, Bonsi P, Meringolo M. Autism Spectrum Disorder: Focus on Glutamatergic Neurotransmission. Int J Mol Sci 2022; 23:ijms23073861. [PMID: 35409220 PMCID: PMC8998955 DOI: 10.3390/ijms23073861] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Disturbances in the glutamatergic system have been increasingly documented in several neuropsychiatric disorders, including autism spectrum disorder (ASD). Glutamate-centered theories of ASD are based on evidence from patient samples and postmortem studies, as well as from studies documenting abnormalities in glutamatergic gene expression and metabolic pathways, including changes in the gut microbiota glutamate metabolism in patients with ASD. In addition, preclinical studies on animal models have demonstrated glutamatergic neurotransmission deficits and altered expression of glutamate synaptic proteins. At present, there are no approved glutamatergic drugs for ASD, but several ongoing clinical trials are currently focusing on evaluating in autistic patients glutamatergic pharmaceuticals already approved for other conditions. In this review, we provide an overview of the literature concerning the role of glutamatergic neurotransmission in the pathophysiology of ASD and as a potential target for novel treatments.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Department of Systems Neuroscience, University Tor Vergata, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| |
Collapse
|
44
|
Abstract
Most psychiatric illnesses, such as schizophrenia, show profound sex differences in incidence, clinical presentation, course, and outcome. Fortunately, more recently the literature on sex differences and (to a lesser extent) effects of sex steroid hormones is expanding, and in this review we have focused on such studies in psychosis, both from a clinical/epidemiological and preclinical/animal model perspective. We begin by briefly describing the clinical evidence for sex differences in schizophrenia epidemiology, symptomatology, and pathophysiology. We then detail sex differences and sex hormone effects in behavioral animal models of psychosis, specifically psychotropic drug-induced locomotor hyperactivity and disruption of prepulse inhibition. We expand on the preclinical data to include developmental and genetic models of psychosis, such as the maternal immune activation model and neuregulin transgenic animals, respectively. Finally, we suggest several recommendations for future studies, in order to facilitate a better understanding of sex differences in the development of psychosis.
Collapse
|
45
|
Luttenbacher I, Phillips A, Kazemi R, Hadipour AL, Sanghvi I, Martinez J, Adamson MM. Transdiagnostic role of glutamate and white matter damage in neuropsychiatric disorders: A Systematic Review. J Psychiatr Res 2022; 147:324-348. [PMID: 35151030 DOI: 10.1016/j.jpsychires.2021.12.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/08/2021] [Accepted: 12/19/2021] [Indexed: 12/09/2022]
Abstract
Neuropsychiatric disorders including generalized anxiety disorder (GAD), obsessive-compulsive disorder (OCD), major depressive disorder (MDD), bipolar disorder (BD), and schizophrenia (SZ) have been considered distinct categories of diseases despite their overlapping characteristics and symptomatology. We aimed to provide an in-depth review elucidating the role of glutamate/Glx and white matter (WM) abnormalities in these disorders from a transdiagnostic perspective. The PubMed online database was searched for studies published between 2010 and 2021. After careful screening, 401 studies were included. The findings point to decreased levels of glutamate in the Anterior Cingulate Cortex in both SZ and BD, whereas Glx is elevated in the Hippocampus in SZ and MDD. With regard to WM abnormalities, the Corpus Callosum and superior Longitudinal Fascicle were the most consistently identified brain regions showing decreased fractional anisotropy (FA) across all the reviewed disorders, except GAD. Additionally, the Uncinate Fasciculus displayed decreased FA in all disorders, except OCD. Decreased FA was also found in the inferior Longitudinal Fasciculus, inferior Fronto-Occipital Fasciculus, Thalamic Radiation, and Corona Radiata in SZ, BD, and MDD. Decreased FA in the Fornix and Corticospinal Tract were found in BD and SZ patients. The Cingulum and Anterior Limb of Internal Capsule exhibited decreased FA in MDD and SZ patients. The results suggest a gradual increase in severity from GAD to SZ defined by the number of brain regions with WM abnormality which may be partially caused by abnormal glutamate levels. WM damage could thus be considered a potential marker of some of the main neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ines Luttenbacher
- Department of Social & Behavioral Sciences, University of Amsterdam, Amsterdam, Netherlands; Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Angela Phillips
- Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA; Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Reza Kazemi
- Department of Cognitive Psychology, Institute for Cognitive Science Studies, Tehran, Iran
| | - Abed L Hadipour
- Department of Cognitive Sciences, University of Messina, Messina, Italy
| | - Isha Sanghvi
- Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neuroscience, University of Southern California, Los Angeles, CA, USA
| | - Julian Martinez
- Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA; Palo Alto University, Palo Alto, CA, USA
| | - Maheen M Adamson
- Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
46
|
Wang G, Woods C, Johnson MA, Milner TA, Glass MJ. Angiotensin II Infusion Results in Both Hypertension and Increased AMPA GluA1 Signaling in Hypothalamic Paraventricular Nucleus of Male but not Female Mice. Neuroscience 2022; 485:129-144. [PMID: 34999197 PMCID: PMC9116447 DOI: 10.1016/j.neuroscience.2021.12.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 10/19/2022]
Abstract
The hypothalamic paraventricular nucleus (PVN) plays a key role in hypertension, however the signaling pathways that contribute to the adaptability of the PVN during hypertension are uncertain. We present evidence that signaling at the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) GluA1 receptor contributes to increased blood pressure in a model of neurogenic hypertension induced by 14-day slow-pressor angiotensin II (AngII) infusion in male mice. It was found that AngII hypertension was associated with an increase in plasma membrane affiliation of GluA1, but decreased GluA2, in dendritic profiles of PVN neurons expressing the TNFα type 1 receptor, a modulator of AMPA receptor trafficking. The increased plasma membrane GluA1 was paralleled by heightened AMPA currents in PVN-spinal cord projection neurons from AngII-infused male mice. Significantly, elevated AMPA currents in AngII-treated mice were blocked by 1-Naphthyl acetyl spermine trihydrochloride, pointing to the involvement of GluA2-lacking GluA1 receptors in the heightened AMPA signaling in PVN neurons. A further functional role for GluA1 in the PVN was demonstrated by the attenuated hypertensive response following silencing of GluA1 in the PVN of AngII-infused male mice. In female mice, AngII-infusion did not impact blood pressure or plasma membrane localization of GluA1 . Post-translational modifications that increase the plasma membrane localization of AMPA GluA1 and heighten the rapid excitatory signaling actions of glutamate in PVN neurons may serve as a molecular substrate underlying sex differences in hypertension.
Collapse
Affiliation(s)
- Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065
| | - Clara Woods
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065
| | - Megan A. Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065,Harold and Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065
| | - Michael J. Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065,Address correspondence to: Dr. Michael J. Glass, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065; Phone: (646) 962-8253;
| |
Collapse
|
47
|
Male sex bias in early and late onset neurodevelopmental disorders: shared aspects and differences in autism spectrum disorder, attention deficit/hyperactivity disorder, and schizophrenia. Neurosci Biobehav Rev 2022; 135:104577. [DOI: 10.1016/j.neubiorev.2022.104577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/23/2022] [Accepted: 02/11/2022] [Indexed: 12/22/2022]
|
48
|
Loureiro CM, Fachim HA, Harte MK, Dalton CF, Reynolds GP. Subchronic PCP effects on DNA methylation and protein expression of NMDA receptor subunit genes in the prefrontal cortex and hippocampus of female rats. J Psychopharmacol 2022; 36:238-244. [PMID: 35102781 DOI: 10.1177/02698811211069109] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND N-methyl-d-aspartate receptor (NMDAR) dysfunction is implicated in schizophrenia, and NMDAR antagonists, such as phencyclidine (PCP), can induce behaviours that mimic aspects of the disorder. AIMS We investigated DNA methylation of Grin1, Grin2a and Grin2b promoter region and NR1 and NR2 protein expression in the prefrontal cortex (PFC) and hippocampus of adult female Lister-hooded rats following subchronic PCP (scPCP) administration. We also determined whether any alterations were tissue-specific. METHODS Rats were divided into two groups that received vehicle (0.9% saline) or 2 mg/kg PCP twice a day for 7 days (n = 10 per group). After behavioural testing (novel object recognition), to confirm a cognitive deficit, brains were dissected and NMDAR subunit DNA methylation and protein expression were analysed by pyrosequencing and ELISA. Line-1 methylation was determined as a measure of global methylation. Data were analysed using Student's t-test and Pearson correlation. RESULTS The scPCP administration led to Grin1 and Grin2b hypermethylation and reduction in NR1 protein in both PFC and hippocampus. No significant differences were observed in Line-1 or Grin2a methylation and NR2 protein. CONCLUSIONS The scPCP treatment resulted in increased DNA methylation at promoter sites of Grin1 and Grin2b NMDAR subunits in two brain areas implicated in schizophrenia, independent of any global change in DNA methylation, and are similar to our observations in a neurodevelopmental animal model of schizophrenia - social isolation rearing post-weaning. Moreover, these alterations may contribute to the changes in protein expression for NMDAR subunits demonstrating the potential importance of epigenetic mechanisms in schizophrenia.
Collapse
Affiliation(s)
- Camila M Loureiro
- Department of Internal Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Helene A Fachim
- Department of Endocrinology and Metabolism, Salford Royal Foundation Trust, Salford, UK
| | - Michael K Harte
- Division of Pharmacy & Optometry, University of Manchester, Manchester, UK
| | - Caroline F Dalton
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| |
Collapse
|
49
|
Olave FA, Aguayo FI, Román-Albasini L, Corrales WA, Silva JP, González PI, Lagos S, García MA, Alarcón-Mardones M, Rojas PS, Xu X, Cidlowski JA, Aliaga E, Fiedler J. Chronic restraint stress produces sex-specific behavioral and molecular outcomes in the dorsal and ventral rat hippocampus. Neurobiol Stress 2022; 17:100440. [PMID: 35252485 PMCID: PMC8894263 DOI: 10.1016/j.ynstr.2022.100440] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/09/2022] [Accepted: 02/21/2022] [Indexed: 01/24/2023] Open
Abstract
Stress-related disorders display differences at multiple levels according to sex. While most studies have been conducted in male rodents, less is known about comparable outcomes in females. In this study, we found that the chronic restraint stress model (2.5 h/day for 14 days) triggers different somatic responses in male and female adult rats. Chronic restraint produced a loss in sucrose preference and novel location preference in male rats. However, chronic restraint failed to produce loss of sucrose preference in females, while it improved spatial performance. We then characterized the molecular responses associated with these behaviors in the hippocampus, comparing the dorsal and ventral poles. Notably, sex- and hippocampal pole-specific transcriptional signatures were observed, along with a significant concordance between the female ventral and male dorsal profiles. Functional enrichment analysis revealed both shared and specific terms associated with each pole and sex. By looking into signaling pathways that were associated with these terms, we found an ample array of sex differences in the dorsal and, to a lesser extent, in the ventral hippocampus. These differences were mainly present in synaptic TrkB signaling, Akt pathway, and glutamatergic receptors. Unexpectedly, the effects of stress on these pathways were rather minimal and mostly dissociated from the sex-specific behavioral outcomes. Our study suggests that female rats are resilient and males susceptible to the restraint stress exposure in the sucrose preference and object location tests, while the activity of canonical signaling pathways is primarily determined by sex rather than stress in the dorsal and ventral hippocampus.
Collapse
Affiliation(s)
- Felipe A. Olave
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Felipe I. Aguayo
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Luciano Román-Albasini
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Wladimir A. Corrales
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Juan P. Silva
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Pablo I. González
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Sara Lagos
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - María A. García
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Matías Alarcón-Mardones
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
| | - Paulina S. Rojas
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Xiaojiang Xu
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, 27709, USA
| | - John A. Cidlowski
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, 27709, USA
| | - Esteban Aliaga
- Medical Technology School and the Neuropsychology and Cognitive Neurosciences Research Center (CINPSI-Neurocog), Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile
- Corresponding author. Medical Technology School, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile.
| | - Jenny Fiedler
- Laboratory of Neuroplasticity and Neurogenetics. Faculty of Chemical and Pharmaceutical Sciences. Department of Biochemistry and Molecular Biology. Universidad de Chile, Independencia, 8380492, Santiago, Chile
- Corresponding author. Department of Biochemistry and Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago. Chile.
| |
Collapse
|
50
|
Navakkode S, Gaunt JR, Pavon MV, Bansal VA, Abraham RP, Chong YS, Ch'ng TH, Sajikumar S. Sex-specific accelerated decay in time/activity-dependent plasticity and associative memory in an animal model of Alzheimer's disease. Aging Cell 2021; 20:e13502. [PMID: 34796608 PMCID: PMC8672784 DOI: 10.1111/acel.13502] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/02/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical studies have shown that female brains are more predisposed to neurodegenerative diseases such as Alzheimer's disease (AD), but the cellular and molecular mechanisms behind this disparity remain unknown. In several mouse models of AD, synaptic plasticity dysfunction is an early event and appears before significant accumulation of amyloid plaques and neuronal degeneration. However, it is unclear whether sexual dimorphism at the synaptic level contributes to the higher risk and prevalence of AD in females. Our studies on APP/PS1 (APPSwe/PS1dE9) mouse model show that AD impacts hippocampal long‐term plasticity in a sex‐specific manner. Long‐term potentiation (LTP) induced by strong tetanic stimulation (STET), theta burst stimulation (TBS) and population spike timing‐dependent plasticity (pSTDP) show a faster decay in AD females compared with age‐matched AD males. In addition, behavioural tagging (BT), a model of associative memory, is specifically impaired in AD females with a faster decay in memory compared with males. Together with the plasticity and behavioural data, we also observed an upregulation of neuroinflammatory markers, along with downregulation of transcripts that regulate cellular processes associated with synaptic plasticity and memory in females. Immunohistochemistry of AD brains confirms that female APP/PS1 mice carry a higher amyloid plaque burden and have enhanced microglial activation compared with male APP/PS1 mice. Their presence in the diseased mice also suggests a link between the impairment of LTP and the upregulation of the inflammatory response. Overall, our data show that synaptic plasticity and associative memory impairments are more prominent in females and this might account for the faster progression of AD in females.
Collapse
Affiliation(s)
- Sheeja Navakkode
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Department of Physiology National University of Singapore Singapore Singapore
| | - Jessica Ruth Gaunt
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Maria Vazquez Pavon
- Department of Physiology National University of Singapore Singapore Singapore
| | | | - Riya Prasad Abraham
- Department of Physiology National University of Singapore Singapore Singapore
| | - Yee Song Chong
- Department of Physiology National University of Singapore Singapore Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- School of Biological Science Nanyang Technological University Singapore Singapore
| | - Sreedharan Sajikumar
- Department of Physiology National University of Singapore Singapore Singapore
- Healthy Longevity Translational Research Programme Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Life Sciences Institute Neurobiology Programme National University of Singapore Singapore Singapore
| |
Collapse
|