1
|
Demaestri C, Pisciotta M, Altunkeser N, Berry G, Hyland H, Breton J, Darling A, Williams B, Bath KG. Central amygdala CRF+ neurons promote heightened threat reactivity following early life adversity in mice. Nat Commun 2024; 15:5522. [PMID: 38951506 PMCID: PMC11217353 DOI: 10.1038/s41467-024-49828-3] [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: 10/31/2023] [Accepted: 06/19/2024] [Indexed: 07/03/2024] Open
Abstract
Failure to appropriately predict and titrate reactivity to threat is a core feature of fear and anxiety-related disorders and is common following early life adversity (ELA). A population of neurons in the lateral central amygdala (CeAL) expressing corticotropin releasing factor (CRF) have been proposed to be key in processing threat of different intensities to mediate active fear expression. Here, we use in vivo fiber photometry to show that ELA results in sex-specific changes in the activity of CeAL CRF+ neurons, yielding divergent mechanisms underlying the augmented startle in ELA mice, a translationally relevant behavior indicative of heightened threat reactivity and hypervigilance. Further, chemogenic inhibition of CeAL CRF+ neurons selectively diminishes startle and produces a long-lasting suppression of threat reactivity. These findings identify a mechanism for sex-differences in susceptibility for anxiety following ELA and have broad implications for understanding the neural circuitry that encodes and gates the behavioral expression of fear.
Collapse
Affiliation(s)
- Camila Demaestri
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, USA
| | - Margaux Pisciotta
- Department of Neuroscience and Behavior, Barnard College of Columbia University, New York, NY, USA
| | - Naira Altunkeser
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - Georgia Berry
- Division of Developmental Neuroscience, Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
| | - Hannah Hyland
- Division of Developmental Neuroscience, Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
| | - Jocelyn Breton
- Division of Developmental Neuroscience, Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Darling
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - Brenna Williams
- Doctoral Program in Cellular and Molecular Physiology & Biophysics, Columbia University, New York, NY, USA
| | - Kevin G Bath
- Division of Developmental Neuroscience, Research Foundation for Mental Hygiene, New York State Psychiatric Institute, New York, NY, USA.
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
2
|
Alcantara-Gonzalez D, Kennedy M, Criscuolo C, Botterill J, Scharfman HE. Increased excitability of dentate gyrus mossy cells occurs early in life in the Tg2576 model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579729. [PMID: 38645244 PMCID: PMC11027210 DOI: 10.1101/2024.02.09.579729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
INTRODUCTION Hyperexcitability in Alzheimer's disease (AD) emerge early and contribute to disease progression. The dentate gyrus (DG) is implicated in hyperexcitability in AD. We hypothesized that mossy cells (MCs), regulators of DG excitability, contribute to early hyperexcitability in AD. Indeed, MCs generate hyperexcitability in epilepsy. METHODS Using the Tg2576 model and WT mice (∼1month-old), we compared MCs electrophysiologically, assessed c-Fos activity marker, Aβ expression and mice performance in a hippocampal-dependent memory task. RESULTS Tg2576 MCs exhibit increased spontaneous excitatory events and decreased inhibitory currents, increasing the charge transfer excitation/inhibition ratio. Tg2576 MC intrinsic excitability was enhanced, and showed higher c-Fos, intracellular Aβ expression, and axon sprouting. Granule cells only showed changes in synaptic properties, without intrinsic changes. The effects occurred before a memory task is affected. DISCUSSION Early electrophysiological and morphological alterations in Tg2576 MCs are consistent with enhanced excitability, suggesting an early role in DG hyperexcitability and AD pathophysiology. HIGHLIGHTS ∘ MCs from 1 month-old Tg2576 mice had increased spontaneous excitatory synaptic input. ∘ Tg2576 MCs had reduced spontaneous inhibitory synaptic input. ∘ Several intrinsic properties were abnormal in Tg2576 MCs. ∘ Tg2576 GCs had enhanced synaptic excitation but no changes in intrinsic properties. ∘ Tg2576 MCs exhibited high c-Fos expression, soluble Aβ and axonal sprouting.
Collapse
|
3
|
Mohammadi-Pilehdarboni H, Shenagari M, Joukar F, Naziri H, Mansour-Ghanaei F. Alzheimer's disease and microorganisms: the non-coding RNAs crosstalk. Front Cell Neurosci 2024; 17:1256100. [PMID: 38249527 PMCID: PMC10796784 DOI: 10.3389/fncel.2023.1256100] [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: 07/10/2023] [Accepted: 10/25/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's disease (AD) is a complex, multifactorial disorder, influenced by a multitude of variables ranging from genetic factors, age, and head injuries to vascular diseases, infections, and various other environmental and demographic determinants. Among the environmental factors, the role of the microbiome in the genesis of neurodegenerative disorders (NDs) is gaining increased recognition. This paradigm shift is substantiated by an extensive body of scientific literature, which underscores the significant contributions of microorganisms, encompassing viruses and gut-derived bacteria, to the pathogenesis of AD. The mechanism by which microbial infection exerts its influence on AD hinges primarily on inflammation. Neuroinflammation, activated in response to microbial infections, acts as a defense mechanism for the brain but can inadvertently lead to unexpected neuropathological perturbations, ultimately contributing to NDs. Given the ongoing uncertainty surrounding the genetic factors underpinning ND, comprehensive investigations into environmental factors, particularly the microbiome and viral agents, are imperative. Recent advances in neuroscientific research have unveiled the pivotal role of non-coding RNAs (ncRNAs) in orchestrating various pathways integral to neurodegenerative pathologies. While the upstream regulators governing the pathological manifestations of microorganisms remain elusive, an in-depth exploration of the nuanced role of ncRNAs holds promise for the development of prospective therapeutic interventions. This review aims to elucidate the pivotal role of ncRNAs as master modulators in the realm of neurodegenerative conditions, with a specific focus on Alzheimer's disease.
Collapse
Affiliation(s)
- Hanieh Mohammadi-Pilehdarboni
- Faculty of Medicine and Dentistry and the School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shenagari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Hamed Naziri
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
4
|
Xie M, Huang Y, Cai W, Zhang B, Huang H, Li Q, Qin P, Han J. Neurobiological Underpinnings of Hyperarousal in Depression: A Comprehensive Review. Brain Sci 2024; 14:50. [PMID: 38248265 PMCID: PMC10813043 DOI: 10.3390/brainsci14010050] [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: 12/11/2023] [Revised: 12/26/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
Patients with major depressive disorder (MDD) exhibit an abnormal physiological arousal pattern known as hyperarousal, which may contribute to their depressive symptoms. However, the neurobiological mechanisms linking this abnormal arousal to depressive symptoms are not yet fully understood. In this review, we summarize the physiological and neural features of arousal, and review the literature indicating abnormal arousal in depressed patients. Evidence suggests that a hyperarousal state in depression is characterized by abnormalities in sleep behavior, physiological (e.g., heart rate, skin conductance, pupil diameter) and electroencephalography (EEG) features, and altered activity in subcortical (e.g., hypothalamus and locus coeruleus) and cortical regions. While recent studies highlight the importance of subcortical-cortical interactions in arousal, few have explored the relationship between subcortical-cortical interactions and hyperarousal in depressed patients. This gap limits our understanding of the neural mechanism through which hyperarousal affects depressive symptoms, which involves various cognitive processes and the cerebral cortex. Based on the current literature, we propose that the hyperconnectivity in the thalamocortical circuit may contribute to both the hyperarousal pattern and depressive symptoms. Future research should investigate the relationship between thalamocortical connections and abnormal arousal in depression, and explore its implications for non-invasive treatments for depression.
Collapse
Affiliation(s)
- Musi Xie
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, School of Psychology, Center for Studies of Psychological Application, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou 510631, China; (M.X.); (Y.H.)
| | - Ying Huang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, School of Psychology, Center for Studies of Psychological Application, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou 510631, China; (M.X.); (Y.H.)
| | - Wendan Cai
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou 510631, China; (W.C.); (B.Z.); (H.H.)
| | - Bingqi Zhang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou 510631, China; (W.C.); (B.Z.); (H.H.)
| | - Haonan Huang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou 510631, China; (W.C.); (B.Z.); (H.H.)
| | - Qingwei Li
- Department of Psychiatry, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China;
| | - Pengmin Qin
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, School of Psychology, Center for Studies of Psychological Application, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou 510631, China; (M.X.); (Y.H.)
- Pazhou Laboratory, Guangzhou 510330, China
| | - Junrong Han
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou 510631, China; (W.C.); (B.Z.); (H.H.)
| |
Collapse
|
5
|
Mueller SG. Traumatic Brain Injury and Post-Traumatic Stress Disorder and Their Influence on Development and Pattern of Alzheimer's Disease Pathology in Later Life. J Alzheimers Dis 2024; 98:1427-1441. [PMID: 38552112 DOI: 10.3233/jad-231183] [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] [Indexed: 04/20/2024]
Abstract
Background Traumatic brain injury (TBI) and posttraumatic stress disorder (PTSD) are potential risk factors for the development of dementia including Alzheimer's disease (AD) in later life. The findings of studies investigating this question are inconsistent though. Objective To investigate if these inconsistencies are caused by the existence of subgroups with different vulnerability for AD pathology and if these subgroups are characterized by atypical tau load/atrophy pattern. Methods The MRI and PET data of 89 subjects with or without previous TBI and/or PTSD from the DoD ADNI database were used to calculate an age-corrected gray matter tau mismatch metric (ageN-T mismatch-score and matrix) for each subject. This metric provides a measure to what degree regional tau accumulation drives regional gray matter atrophy (matrix) and can be used to calculate a summary score (score) reflecting the severity of AD pathology in an individual. Results The ageN-T mismatch summary score was positively correlated with whole brain beta-amyloid load and general cognitive function but not with PTSD or TBI severity. Hierarchical cluster analysis identified five different spatial patterns of tau-gray matter interactions. These clusters reflected the different stages of the typical AD tau progression pattern. None was exclusively associated with PTSD and/or TBI. Conclusions These findings suggest that a) although subsets of patients with PTSD and/or TBI develop AD-pathology, a history of TBI or PTSD alone or both is not associated with a significantly higher risk to develop AD pathology in later life. b) remote TBI or PTSD do not modify the typical AD pathology distribution pattern.
Collapse
Affiliation(s)
- Susanne G Mueller
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
6
|
Hodes GE, Bangasser D, Sotiropoulos I, Kokras N, Dalla C. Sex Differences in Stress Response: Classical Mechanisms and Beyond. Curr Neuropharmacol 2024; 22:475-494. [PMID: 37855285 PMCID: PMC10845083 DOI: 10.2174/1570159x22666231005090134] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 10/20/2023] Open
Abstract
Neuropsychiatric disorders, which are associated with stress hormone dysregulation, occur at different rates in men and women. Moreover, nowadays, preclinical and clinical evidence demonstrates that sex and gender can lead to differences in stress responses that predispose males and females to different expressions of similar pathologies. In this curated review, we focus on what is known about sex differences in classic mechanisms of stress response, such as glucocorticoid hormones and corticotrophin-releasing factor (CRF), which are components of the hypothalamicpituitary- adrenal (HPA) axis. Then, we present sex differences in neurotransmitter levels, such as serotonin, dopamine, glutamate and GABA, as well as indices of neurodegeneration, such as amyloid β and Tau. Gonadal hormone effects, such as estrogens and testosterone, are also discussed throughout the review. We also review in detail preclinical data investigating sex differences caused by recentlyrecognized regulators of stress and disease, such as the immune system, genetic and epigenetic mechanisms, as well neurosteroids. Finally, we discuss how understanding sex differences in stress responses, as well as in pharmacology, can be leveraged into novel, more efficacious therapeutics for all. Based on the supporting evidence, it is obvious that incorporating sex as a biological variable into preclinical research is imperative for the understanding and treatment of stress-related neuropsychiatric disorders, such as depression, anxiety and Alzheimer's disease.
Collapse
Affiliation(s)
| | - Debra Bangasser
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences & Applications NCSR “Demokritos”, Athens, Greece
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
7
|
Yang X, Geng F. Corticotropin-releasing factor signaling and its potential role in the prefrontal cortex-dependent regulation of anxiety. J Neurosci Res 2023; 101:1781-1794. [PMID: 37592912 DOI: 10.1002/jnr.25238] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/08/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Abstract
A large body of literature has highlighted the significance of the corticotropin-releasing factor (CRF) system in the regulation of neuropsychiatric diseases. Anxiety disorders are among the most common neuropsychiatric disorders. An increasing number of studies have demonstrated that the CRF family mediates and regulates the development and maintenance of anxiety. Thus, the CRF family is considered to be a potential target for the treatment of anxiety disorders. The prefrontal cortex (PFC) plays a role in the occurrence and development of anxiety, and both CRF and CRF-R1 are widely expressed in the PFC. This paper begins by reviewing CRF-related signaling pathways and their different roles in anxiety and related processes. Then, the role of the CRF system in other neuropsychiatric diseases is reviewed and the potential role of PFC CRF signaling in the regulation of anxiety disorders is discussed. Although other signaling pathways are potentially involved in the process of anxiety, CRF in the PFC primarily modulates anxiety disorders through the activation of corticotropin-releasing factor type1 receptors (CRF-R1) and the excitation of the cAMP/PKA signaling pathway. Moreover, the main signaling pathways of CRF involved in sex differentiation in the PFC appear to be different. In summary, this review suggests that the CRF system in the PFC plays a critical role in the occurrence of anxiety. Thus, CRF signaling is of great significance as a potential target for the treatment of stress-related disorders in the future.
Collapse
Affiliation(s)
- Xin Yang
- Department of Physiology, Shantou University Medical College, Shantou, China
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fei Geng
- Department of Physiology, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| |
Collapse
|
8
|
Reyna NC, Clark BJ, Hamilton DA, Pentkowski NS. Anxiety and Alzheimer's disease pathogenesis: focus on 5-HT and CRF systems in 3xTg-AD and TgF344-AD animal models. Front Aging Neurosci 2023; 15:1251075. [PMID: 38076543 PMCID: PMC10699143 DOI: 10.3389/fnagi.2023.1251075] [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: 06/30/2023] [Accepted: 10/25/2023] [Indexed: 02/12/2024] Open
Abstract
Dementia remains one of the leading causes of morbidity and mortality in older adults. Alzheimer's disease (AD) is the most common type of dementia, affecting over 55 million people worldwide. AD is characterized by distinct neurobiological changes, including amyloid-beta protein deposits and tau neurofibrillary tangles, which cause cognitive decline and subsequent behavioral changes, such as distress, insomnia, depression, and anxiety. Recent literature suggests a strong connection between stress systems and AD progression. This presents a promising direction for future AD research. In this review, two systems involved in regulating stress and AD pathogenesis will be highlighted: serotonin (5-HT) and corticotropin releasing factor (CRF). Throughout the review, we summarize critical findings in the field while discussing common limitations with two animal models (3xTg-AD and TgF344-AD), novel pharmacotherapies, and potential early-intervention treatment options. We conclude by highlighting promising future pharmacotherapies and translational animal models of AD and anxiety.
Collapse
Affiliation(s)
- Nicole C. Reyna
- Department of Psychology, University of New Mexico, Albuquerque, NM, United States
| | | | | | | |
Collapse
|
9
|
Edwards HM, Wallace CE, Gardiner WD, Doherty BM, Harrigan RT, Yuede KM, Yuede CM, Cirrito JR. Sex-dependent effects of acute stress on amyloid-β in male and female mice. Brain 2023; 146:2268-2274. [PMID: 37127299 PMCID: PMC10232275 DOI: 10.1093/brain/awad052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/09/2022] [Accepted: 01/21/2023] [Indexed: 05/03/2023] Open
Abstract
The risk of developing Alzheimer's disease is mediated by a combination of genetics and environmental factors, such as stress, sleep abnormalities and traumatic brain injury. Women are at a higher risk of developing Alzheimer's disease than men, even when controlling for differences in lifespan. Women are also more likely to report high levels of stress than men. Sex differences in response to stress may play a role in the increased risk of Alzheimer's disease in women. In this study, we use in vivo microdialysis to measure levels of Aβ in response to acute stress in male and female mice. We show that Aβ levels are altered differently between female and male mice (APP/PS1 and wild-type) in response to stress, with females showing significantly increased levels of Aβ while most males do not show a significant change. This response is mediated through β-arrestin involvement in Corticotrophin Releasing Factor receptor signalling pathway differences in male and female mice as male mice lacking β-arrestin show increase in Aβ in response to stress similar to females.
Collapse
Affiliation(s)
- Hannah M Edwards
- Department of Neurology, Knight Alzheimer’s Disease Research Center, Hope Center for Neurological Disorders, St. Louis, MO 63108, USA
| | - Clare E Wallace
- Department of Neurology, Knight Alzheimer’s Disease Research Center, Hope Center for Neurological Disorders, St. Louis, MO 63108, USA
| | - Woodrow D Gardiner
- Department of Neurology, Knight Alzheimer’s Disease Research Center, Hope Center for Neurological Disorders, St. Louis, MO 63108, USA
| | - Brookelyn M Doherty
- Department of Neurology, Knight Alzheimer’s Disease Research Center, Hope Center for Neurological Disorders, St. Louis, MO 63108, USA
| | - Ryan T Harrigan
- Department of Neurology, Knight Alzheimer’s Disease Research Center, Hope Center for Neurological Disorders, St. Louis, MO 63108, USA
| | - Kayla M Yuede
- Department of Neurology, Knight Alzheimer’s Disease Research Center, Hope Center for Neurological Disorders, St. Louis, MO 63108, USA
| | - Carla M Yuede
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John R Cirrito
- Department of Neurology, Knight Alzheimer’s Disease Research Center, Hope Center for Neurological Disorders, St. Louis, MO 63108, USA
| |
Collapse
|
10
|
Bates MLS, Arner JR, Curtis AL, Valentino R, Bhatnagar S. Sex-specific alterations in corticotropin-releasing factor regulation of coerulear-cortical network activity. Neuropharmacology 2023; 223:109317. [PMID: 36334761 DOI: 10.1016/j.neuropharm.2022.109317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
The locus coeruleus (LC)-norepinephrine system is a stress responsive system that regulates arousal and cognitive functions through extensive projections, including to the prefrontal cortex. LC-cortical circuits are activated by stressors, and this activation is thought to contribute to stress-induced impairments in executive function. Because corticotropin-releasing factor (CRF) is a mediator of stress-induced LC activation, we examined the effects of CRF administered into the LC of male and female rats on network activity of two functionally distinct regions of the PFC, the medial PFC (mPFC) and the orbitofrontal cortex (OFC). Network activity, measured as local field potentials, was recorded in awake animals before and after intra-LC infusion of aCSF or CRF (2 or 20 ng). CRF had qualitatively distinct effects on network activity in males and females with respect to dose, region and timecourse. CRF (20 ng) produced a prominent theta oscillation (7-9 Hz) selectively in female rats shortly after LC infusion and 20 min later. In contrast, in male rats, CRF (2 and 20 ng) decreased the amplitude of power in the 4-6 Hz range in the mPFC 10 min after injection. Lastly, CRF (20 ng) increased mPFC-OFC coherence in females and decreased mPFC-OFC coherence in males. In sum, these results show sex differences in CRF modulation of the LC-norepinephrine system that regulates prefrontal cortical networks, which may underlie sex differences in cognitive and behavioral responses to stress.
Collapse
Affiliation(s)
- M L Shawn Bates
- Department of Psychology, California State University, Chico, 400 W. First St, Chico, CA, 95929, USA
| | - Jay R Arner
- Division of Stress Neurobiology, Department of Anesthesiology and Critical Care, Abramson Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Andre L Curtis
- Division of Stress Neurobiology, Department of Anesthesiology and Critical Care, Abramson Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Rita Valentino
- Division of Stress Neurobiology, Department of Anesthesiology and Critical Care, Abramson Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Philadelphia, PA, 19104, USA; The Perelman School of Medicine, University of Pennsylvania, USA
| | - Seema Bhatnagar
- Division of Stress Neurobiology, Department of Anesthesiology and Critical Care, Abramson Research Center, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Philadelphia, PA, 19104, USA; The Perelman School of Medicine, University of Pennsylvania, USA.
| |
Collapse
|
11
|
Dioli C, Papadimitriou G, Megalokonomou A, Marques C, Sousa N, Sotiropoulos I. Chronic Stress, Depression, and Alzheimer's Disease: The Triangle of Oblivion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1423:303-315. [PMID: 37525058 DOI: 10.1007/978-3-031-31978-5_31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Chronic stress and high levels of the main stress hormones, and glucocorticoids (GC), are implicated in susceptibility to brain pathologies such as depression and Alzheimer's disease (AD), as they promote neural plasticity damage and glial reactivity, which can lead to dendritic/synaptic loss, reduced neurogenesis, mood deficits, and impaired cognition. Moreover, depression is implicated in the development of AD with chronic stress being a potential link between both disorders via common neurobiological underpinnings. Hereby, we summarize and discuss the clinical and preclinical evidence related to the detrimental effect of chronic stress as a precipitator of AD through the activation of pathological mechanisms leading to the accumulation of amyloid β (Aβ) and Tau protein. Given that the modern lifestyle increasingly exposes individuals to high stress loads, it is clear that understanding the mechanistic link(s) between chronic stress, depression, and AD pathogenesis may facilitate the treatment of AD and other stress-related disorders.
Collapse
Affiliation(s)
- Chrysoula Dioli
- Institute of Biosciences and Applications, NCSR Demokritos, Athens, Greece
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | | | - Carlos Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ioannis Sotiropoulos
- Institute of Biosciences and Applications, NCSR Demokritos, Athens, Greece.
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
12
|
Dos Santos Claro PA, Silbermins M, Inda C, Silberstein S. CRHR1 endocytosis: Spatiotemporal regulation of receptor signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:229-260. [PMID: 36813360 DOI: 10.1016/bs.pmbts.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Corticotropin releasing hormone (CRH) is crucial for basal and stress-initiated reactions in the hypothalamic-pituitary-adrenal axis (HPA) and extrahypothalamic brain circuits, where it acts as a neuromodulator to organize behavioral and humoral responses to stress. We review and describe cellular components and molecular mechanisms involved in CRH system signaling through G protein-coupled receptors (GPCRs) CRHR1 and CRHR2, under the current view of GPCR signaling from the plasma membrane but also from intracellular compartments, which establish the bases of signal resolution in space and time. Focus is placed on latest studies of CRHR1 signaling in physiologically significant contexts of the neurohormone function that disclosed new mechanistic features of cAMP production and ERK1/2 activation. We also introduce in a brief overview the pathophysiological function of the CRH system, underlining the need for a complete characterization of CRHRs signaling to design new and specific therapies for stress-related disorders.
Collapse
Affiliation(s)
- Paula A Dos Santos Claro
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Micaela Silbermins
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina Inda
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Octamer SRL, Buenos Aires, Argentina
| | - Susana Silberstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
13
|
Boyd RJ, Avramopoulos D, Jantzie LL, McCallion AS. Neuroinflammation represents a common theme amongst genetic and environmental risk factors for Alzheimer and Parkinson diseases. J Neuroinflammation 2022; 19:223. [PMID: 36076238 PMCID: PMC9452283 DOI: 10.1186/s12974-022-02584-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/23/2022] [Indexed: 11/21/2022] Open
Abstract
Multifactorial diseases are characterized by inter-individual variation in etiology, age of onset, and penetrance. These diseases tend to be relatively common and arise from the combined action of genetic and environmental factors; however, parsing the convoluted mechanisms underlying these gene-by-environment interactions presents a significant challenge to their study and management. For neurodegenerative disorders, resolving this challenge is imperative, given the enormous health and societal burdens they impose. The mechanisms by which genetic and environmental effects may act in concert to destabilize homeostasis and elevate risk has become a major research focus in the study of common disease. Emphasis is further being placed on determining the extent to which a unifying biological principle may account for the progressively diminishing capacity of a system to buffer disease phenotypes, as risk for disease increases. Data emerging from studies of common, neurodegenerative diseases are providing insights to pragmatically connect mechanisms of genetic and environmental risk that previously seemed disparate. In this review, we discuss evidence positing inflammation as a unifying biological principle of homeostatic destabilization affecting the risk, onset, and progression of neurodegenerative diseases. Specifically, we discuss how genetic variation associated with Alzheimer disease and Parkinson disease may contribute to pro-inflammatory responses, how such underlying predisposition may be exacerbated by environmental insults, and how this common theme is being leveraged in the ongoing search for effective therapeutic interventions.
Collapse
Affiliation(s)
- Rachel J Boyd
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Dimitri Avramopoulos
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lauren L Jantzie
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Kennedy Krieger Institute, Baltimore, MD, 21205, USA
| | - Andrew S McCallion
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
14
|
Andrews EJ, Martini AC, Head E. Exploring the role of sex differences in Alzheimer's disease pathogenesis in Down syndrome. Front Neurosci 2022; 16:954999. [PMID: 36033603 PMCID: PMC9411995 DOI: 10.3389/fnins.2022.954999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/25/2022] [Indexed: 11/14/2022] Open
Abstract
Women are disproportionately affected by Alzheimer's disease (AD), yet little is known about sex-specific effects on the development of AD in the Down syndrome (DS) population. DS is caused by a full or partial triplication of chromosome 21, which harbors the amyloid precursor protein (APP) gene, among others. The majority of people with DS in their early- to mid-40s will accumulate sufficient amyloid-beta (Aβ) in their brains along with neurofibrillary tangles (NFT) for a neuropathological diagnosis of AD, and the triplication of the APP gene is regarded as the main cause. Studies addressing sex differences with age and impact on dementia in people with DS are inconsistent. However, women with DS experience earlier age of onset of menopause, marked by a drop in estrogen, than women without DS. This review focuses on key sex differences observed with age and AD in people with DS and a discussion of possible underlying mechanisms that could be driving or protecting from AD development in DS. Understanding how biological sex influences the brain will lead to development of dedicated therapeutics and interventions to improve the quality of life for people with DS and AD.
Collapse
Affiliation(s)
- Elizabeth J. Andrews
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Alessandra C. Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
15
|
Tokatli MR, Sisti LG, Marziali E, Nachira L, Rossi MF, Amantea C, Moscato U, Malorni W. Hormones and Sex-Specific Medicine in Human Physiopathology. Biomolecules 2022; 12:413. [PMID: 35327605 PMCID: PMC8946266 DOI: 10.3390/biom12030413] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/04/2022] [Indexed: 12/11/2022] Open
Abstract
A prodigious increment of scientific evidence in both preclinical and clinical studies is narrowing a major gap in knowledge regarding sex-specific biological responses observed in numerous branches of clinical practices. Some paradigmatic examples include neurodegenerative and mental disorders, immune-related disorders such as pathogenic infections and autoimmune diseases, oncologic conditions, and cardiovascular morbidities. The male-to-female proportion in a population is expressed as sex ratio and varies eminently with respect to the pathophysiology, natural history, incidence, prevalence, and mortality rates. The factors that determine this scenario incorporate both sex-associated biological differences and gender-dependent sociocultural issues. A broad narrative review focused on the current knowledge about the role of hormone regulation in gender medicine and gender peculiarities across key clinical areas is provided. Sex differences in immune response, cardiovascular diseases, neurological disorders, cancer, and COVID-19 are some of the hints reported. Moreover, gender implications in occupational health and health policy are offered to support the need for more personalized clinical medicine and public health approaches to achieve an ameliorated quality of life of patients and better outcomes in population health.
Collapse
Affiliation(s)
| | - Leuconoe Grazia Sisti
- Center for Global Health Research and Studies, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.G.S.); (E.M.); (L.N.); (U.M.)
- National Institute for Health, Migration and Poverty, 00153 Rome, Italy
| | - Eleonora Marziali
- Center for Global Health Research and Studies, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.G.S.); (E.M.); (L.N.); (U.M.)
| | - Lorenza Nachira
- Center for Global Health Research and Studies, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.G.S.); (E.M.); (L.N.); (U.M.)
| | - Maria Francesca Rossi
- Department of Life Sciences and Public Health, Section of Occupational Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.F.R.); (C.A.)
| | - Carlotta Amantea
- Department of Life Sciences and Public Health, Section of Occupational Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.F.R.); (C.A.)
| | - Umberto Moscato
- Center for Global Health Research and Studies, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.G.S.); (E.M.); (L.N.); (U.M.)
- Department of Life Sciences and Public Health, Section of Occupational Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.F.R.); (C.A.)
| | - Walter Malorni
- Course in Pharmacy, University of Tor Vergata, 00133 Rome, Italy;
- Center for Global Health Research and Studies, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.G.S.); (E.M.); (L.N.); (U.M.)
| |
Collapse
|
16
|
Guo L, Zhong MB, Zhang L, Zhang B, Cai D. Sex Differences in Alzheimer's Disease: Insights From the Multiomics Landscape. Biol Psychiatry 2022; 91:61-71. [PMID: 33896621 PMCID: PMC8996342 DOI: 10.1016/j.biopsych.2021.02.968] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) has complex etiologies, and the impact of sex on AD varies over the course of disease development. The literature provides some evidence of sex-specific contributions to AD. However, molecular mechanisms of sex-biased differences in AD remain elusive. Multiomics data in tandem with systems biology approaches offer a new avenue to dissect sex-stratified molecular mechanisms of AD and to develop sex-specific diagnostic and therapeutic strategies for AD. Single-cell transcriptomic datasets and cell deconvolution of bulk tissue transcriptomic data provide additional insights into brain cell type-specific impact on sex-biased differences in AD. In this review, we summarize the impact of sex chromosomes and sex hormones on AD, the impact of sex-biased differences during AD development, and the interplay between sex and a major AD genetic risk factor, the APOE ε4 genotype, through the multiomics landscape. Several sex-biased molecular pathways such as neuroinflammation and bioenergetic metabolism have been identified. The importance of sex chromosome and sex hormones, as well as the associated pathways in AD pathogenesis, is further strengthened by findings from omics studies. Future research efforts should integrate the multiomics data from different brain regions and different cell types using systems biology approaches, and leverage the knowledge into a holistic examination of sex differences in AD. Advances in systems biology technologies and increasingly available large-scale multiomics datasets will facilitate future studies dissecting such complex signaling mechanisms to better understand AD pathogenesis in both sexes, with the ultimate goals of developing efficacious sex- and APOE-stratified preventive and therapeutic interventions for AD.
Collapse
Affiliation(s)
- Lei Guo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Margaret B Zhong
- Department of Neuroscience, Barnard College of Columbia University, New York, New York
| | - Larry Zhang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York; Research and Development Service, James J. Peters VA Medical Center, Bronx, New York
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Dongming Cai
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York; Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, New York; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York; Research and Development Service, James J. Peters VA Medical Center, Bronx, New York.
| |
Collapse
|
17
|
Sex differences in anxiety and depression: circuits and mechanisms. Nat Rev Neurosci 2021; 22:674-684. [PMID: 34545241 DOI: 10.1038/s41583-021-00513-0] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 02/08/2023]
Abstract
Epidemiological sex differences in anxiety disorders and major depression are well characterized. Yet the circuits and mechanisms that contribute to these differences are understudied, because preclinical studies have historically excluded female rodents. This oversight is beginning to be addressed, and recent studies that include male and female rodents are identifying sex differences in neurobiological processes that underlie features of these disorders, including conflict anxiety, fear processing, arousal, social avoidance, learned helplessness and anhedonia. These findings allow us to conceptualize various types of sex differences in the brain, which in turn have broader implications for considering sex as a biological variable. Importantly, comparing the sexes could aid in the discovery of novel therapeutics.
Collapse
|
18
|
Locci A, Yan Y, Rodriguez G, Dong H. Sex differences in CRF1, CRF, and CRFBP expression in C57BL/6J mouse brain across the lifespan and in response to acute stress. J Neurochem 2021; 158:943-959. [PMID: 32813270 PMCID: PMC9811412 DOI: 10.1111/jnc.15157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 01/07/2023]
Abstract
Signaling pathways mediated by corticotropin-releasing factor and its receptor 1 (CRF1) play a central role in stress responses. Dysfunction of the CRF system has been associated with neuropsychiatric disorders. However, dynamic changes in the CRF system during brain development and aging are not well investigated. In this study, we characterized CRF1, CRF, and corticotropin-releasing factor binding protein (CRFBP) expression in different brain regions in both male and female C57BL/6J mice from 1 to 18 months of age under basal conditions as well as after an acute 2-hr-restraint stress. We found that CRF and CRF1 levels tended to increase in the hippocampus and hypothalamus, and to decrease in the prefrontal cortex with aging, especially at 18 months of age, whereas CRFBP expression followed an opposite direction in these brain areas. We also observed area-specific sex differences in the expression of these three proteins. For example, CRF expression was lower in females than in males in all the brain regions examined except the prefrontal cortex. After acute stress, CRF and CRF1 were up-regulated at 1, 6, and 12 months of age, and down-regulated at 18 months of age. Females showed more robust changes compared to males of the same age. CRFBP expression either decreased or remained unchanged in most of the brain areas following acute stress. Our findings suggest that brain CRF1, CRF, and CRFBP expression changes dynamically across the lifespan and under stress condition in a sex- and regional-specific manner. Sex differences in the CRF system in response to stress may contribute to the etiology of stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Andrea Locci
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yan Yan
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
19
|
Grubisha MJ, Sweet RA, MacDonald ML. Investigating Post-translational Modifications in Neuropsychiatric Disease: The Next Frontier in Human Post-mortem Brain Research. Front Mol Neurosci 2021; 14:689495. [PMID: 34335181 PMCID: PMC8322442 DOI: 10.3389/fnmol.2021.689495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/18/2021] [Indexed: 12/27/2022] Open
Abstract
Gene expression and translation have been extensively studied in human post-mortem brain tissue from subjects with psychiatric disease. Post-translational modifications (PTMs) have received less attention despite their implication by unbiased genetic studies and importance in regulating neuronal and circuit function. Here we review the rationale for studying PTMs in psychiatric disease, recent findings in human post-mortem tissue, the required controls for these types of studies, and highlight the emerging mass spectrometry approaches transforming this research direction.
Collapse
Affiliation(s)
- Melanie J. Grubisha
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew L. MacDonald
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Bhargava A, Arnold AP, Bangasser DA, Denton KM, Gupta A, Hilliard Krause LM, Mayer EA, McCarthy M, Miller WL, Raznahan A, Verma R. Considering Sex as a Biological Variable in Basic and Clinical Studies: An Endocrine Society Scientific Statement. Endocr Rev 2021; 42:219-258. [PMID: 33704446 PMCID: PMC8348944 DOI: 10.1210/endrev/bnaa034] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 02/08/2023]
Abstract
In May 2014, the National Institutes of Health (NIH) stated its intent to "require applicants to consider sex as a biological variable (SABV) in the design and analysis of NIH-funded research involving animals and cells." Since then, proposed research plans that include animals routinely state that both sexes/genders will be used; however, in many instances, researchers and reviewers are at a loss about the issue of sex differences. Moreover, the terms sex and gender are used interchangeably by many researchers, further complicating the issue. In addition, the sex or gender of the researcher might influence study outcomes, especially those concerning behavioral studies, in both animals and humans. The act of observation may change the outcome (the "observer effect") and any experimental manipulation, no matter how well-controlled, is subject to it. This is nowhere more applicable than in physiology and behavior. The sex of established cultured cell lines is another issue, in addition to aneuploidy; chromosomal numbers can change as cells are passaged. Additionally, culture medium contains steroids, growth hormone, and insulin that might influence expression of various genes. These issues often are not taken into account, determined, or even considered. Issues pertaining to the "sex" of cultured cells are beyond the scope of this Statement. However, we will discuss the factors that influence sex and gender in both basic research (that using animal models) and clinical research (that involving human subjects), as well as in some areas of science where sex differences are routinely studied. Sex differences in baseline physiology and associated mechanisms form the foundation for understanding sex differences in diseases pathology, treatments, and outcomes. The purpose of this Statement is to highlight lessons learned, caveats, and what to consider when evaluating data pertaining to sex differences, using 3 areas of research as examples; it is not intended to serve as a guideline for research design.
Collapse
Affiliation(s)
- Aditi Bhargava
- Center for Reproductive Sciences, San Francisco, CA, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, CA, USA
| | - Arthur P Arnold
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lucinda M Hilliard Krause
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, CA, USA
| | - Margaret McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Walter L Miller
- Center for Reproductive Sciences, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institutes of Mental Health, Intramural Research Program, Bethesda, MD, USA
| | - Ragini Verma
- Diffusion and Connectomics In Precision Healthcare Research (DiCIPHR) lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Poumeaud F, Mircher C, Smith PJ, Faye PA, Sturtz FG. Deciphering the links between psychological stress, depression, and neurocognitive decline in patients with Down syndrome. Neurobiol Stress 2021; 14:100305. [PMID: 33614867 PMCID: PMC7879042 DOI: 10.1016/j.ynstr.2021.100305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/16/2021] [Accepted: 01/23/2021] [Indexed: 12/27/2022] Open
Abstract
The relationships between psychological stress and cognitive functions are still to be defined despite some recent progress. Clinically, we noticed that patients with Down syndrome (DS) may develop rapid neurocognitive decline and Alzheimer's disease (AD) earlier than expected, often shortly after a traumatic life event (bereavement over the leave of a primary caregiver, an assault, modification of lifestyle, or the loss of parents). Of course, individuals with DS are naturally prone to develop AD, given the triplication of chromosome 21. However, the relatively weak intensity of the stressful event and the rapid pace of cognitive decline after stress in these patients have to be noticed. It seems DS patients react to stress in a similar manner normal persons react to a very intense stress, and thereafter develop a state very much alike post-traumatic stress disorders. Unfortunately, only a few studies have studied stress-induced regression in patients with DS. Thus, we reviewed the biochemical events involved in psychological stress and found some possible links with cognitive impairment and AD. Interestingly, these links could probably be also applied to non-DS persons submitted to an intense stress. We believe these links should be further explored as a better understanding of the relationships between stress and cognition could help in many situations including individuals of the general population.
Collapse
Affiliation(s)
- François Poumeaud
- Univ. Limoges, Peripheral Neuropathies, EA6309, F-87000, Limoges, France
| | - Clotilde Mircher
- Institut Jérôme Lejeune, 37 Rue des Volontaires, F-75015, Paris, France
| | - Peter J. Smith
- University of Chicago, 950 E. 61st Street, SSC Suite 207, Chicago, IL, 60637, USA
| | - Pierre-Antoine Faye
- Univ. Limoges, Peripheral Neuropathies, EA6309, F-87000, Limoges, France
- CHU Limoges, Department of Biochemistry and Molecular Biology, F-87000, Limoges, France
| | - Franck G. Sturtz
- Univ. Limoges, Peripheral Neuropathies, EA6309, F-87000, Limoges, France
- CHU Limoges, Department of Biochemistry and Molecular Biology, F-87000, Limoges, France
| |
Collapse
|
22
|
Mentis AFA, Dardiotis E, Chrousos GP. Apolipoprotein E4 and meningeal lymphatics in Alzheimer disease: a conceptual framework. Mol Psychiatry 2021; 26:1075-1097. [PMID: 32355332 PMCID: PMC7985019 DOI: 10.1038/s41380-020-0731-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 04/01/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
The potential existence and roles of the meningeal lymphatic system in normal and pathological brain function have been a long-standing enigma. Recent evidence suggests that meningeal lymphatic vessels are present in both the mouse and human brain; in mice, they seem to play a role in clearing toxic amyloid-beta peptides, which have been connected with Alzheimer disease (AD). Here, we review the evidence linking the meningeal lymphatic system with human AD. Novel findings suggest that the recently described meningeal lymphatic vessels could be linked to, and possibly drain, the efferent paravascular glial lymphatic (glymphatic) system carrying cerebrospinal fluid, after solute and immune cell exchange with brain interstitial fluid. In so doing, the glymphatic system could contribute to the export of toxic solutes and immune cells from the brain (an exported fluid we wish to describe as glymph, similarly to lymph) to the meningeal lymphatic system; the latter, by being connected with downstream anatomic regions, carries the glymph to the conventional cervical lymphatic vessels and nodes. Thus, abnormal function in the meningeal lymphatic system could, in theory, lead to the accumulation, in the brain, of amyloid-beta, cellular debris, and inflammatory mediators, as well as immune cells, resulting in damage of the brain parenchyma and, in turn, cognitive and other neurologic dysfunctions. In addition, we provide novel insights into APOE4-the leading genetic risk factor for AD-and its relation to the meningeal lymphatic system. In this regard, we have reanalyzed previously published RNA-Seq data to show that induced pluripotent stem cells (iPSCs) carrying the APOE4 allele (either as APOE4 knock-in or stemming from APOE4 patients) express lower levels of (a) genes associated with lymphatic markers, and (b) genes for which well-characterized missense mutations have been linked to peripheral lymphedema. Taking into account this evidence, we propose a new conceptual framework, according to which APOE4 could play a novel role in the premature shrinkage of meningeal lymphatic vessels (meningeal lymphosclerosis), leading to abnormal meningeal lymphatic functions (meningeal lymphedema), and, in turn, reduction in the clearance of amyloid-beta and other macromolecules and inflammatory mediators, as well as immune cells, from the brain, exacerbation of AD manifestations, and progression of the disease. Altogether, these findings and their potential interpretations may herald novel diagnostic tools and therapeutic approaches in patients with AD.
Collapse
Affiliation(s)
- Alexios-Fotios A Mentis
- Public Health Laboratories, Hellenic Pasteur Institute, Vas. Sofias Avenue 127, 115 21, Athens, Greece.
- Department of Microbiology, University of Thessaly, Panepistimiou 3, Viopolis, 41 500, Larissa, Greece.
| | - Efthimios Dardiotis
- Department of Neurology, University of Thessaly, Panepistimiou 3, Viopolis, 41 500, Larissa, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Medical School, Aghia Sophia Children's Hospital, Livadias 8, 115 27, Athens, Greece
- UNESCO Chair on Adolescent Health Care, Athens, Greece
| |
Collapse
|
23
|
Alcantara-Gonzalez D, Chartampila E, Criscuolo C, Scharfman HE. Early changes in synaptic and intrinsic properties of dentate gyrus granule cells in a mouse model of Alzheimer's disease neuropathology and atypical effects of the cholinergic antagonist atropine. Neurobiol Dis 2021; 152:105274. [PMID: 33484828 DOI: 10.1016/j.nbd.2021.105274] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022] Open
Abstract
It has been reported that hyperexcitability occurs in a subset of patients with Alzheimer's disease (AD) and hyperexcitability could contribute to the disease. Several studies have suggested that the hippocampal dentate gyrus (DG) may be an important area where hyperexcitability occurs. Therefore, we tested the hypothesis that the principal DG cell type, granule cells (GCs), would exhibit changes at the single-cell level which would be consistent with hyperexcitability and might help explain it. We used the Tg2576 mouse, where it has been shown that hyperexcitability is robust at 2-3 months of age. GCs from 2 to 3-month-old Tg2576 mice were compared to age-matched wild type (WT) mice. Effects of muscarinic cholinergic antagonism were tested because previously we found that Tg2576 mice exhibited hyperexcitability in vivo that was reduced by the muscarinic cholinergic antagonist atropine, counter to the dogma that in AD one needs to boost cholinergic function. The results showed that GCs from Tg2576 mice exhibited increased frequency of spontaneous excitatory postsynaptic potentials/currents (sEPSP/Cs) and reduced frequency of spontaneous inhibitory synaptic events (sIPSCs) relative to WT, increasing the excitation:inhibition (E:I) ratio. There was an inward NMDA receptor-dependent current that we defined here as a novel synaptic current (nsC) in Tg2576 mice because it was very weak in WT mice. Intrinsic properties were distinct in Tg2576 GCs relative to WT. In summary, GCs of the Tg2576 mouse exhibit early electrophysiological alterations that are consistent with increased synaptic excitation, reduced inhibition, and muscarinic cholinergic dysregulation. The data support previous suggestions that the DG contributes to hyperexcitability and there is cholinergic dysfunction early in life in AD mouse models.
Collapse
Affiliation(s)
- David Alcantara-Gonzalez
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Elissavet Chartampila
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Chiara Criscuolo
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | - Helen E Scharfman
- Center for Dementia Research, the Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; Department of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York, NY 10016, USA; Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
24
|
Reproductive status impact on tau phosphorylation induced by chronic stress. Neurobiol Stress 2020; 13:100241. [PMID: 33344697 PMCID: PMC7739034 DOI: 10.1016/j.ynstr.2020.100241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 01/20/2023] Open
Abstract
Sex and exposure to chronic stress have been identified as risk factors for developing Alzheimer's disease (AD). Although AD has been demonstrated to be more prevalent in females, sex is often overlooked in research studies, likely due to the complexity of the hormonal status. In female rats, the reproductive status can modulate the well-known increase in tau phosphorylation (pTau) caused by the exposure to acute physical and psychological stressors. To test the hypothesis that reproductive status can impact hippocampal pTau induced by chronic stress, cohorts of virgin, lactating (4–5 days pp), and post-maternal (1-month post-weaned) rats were subjected to a daily 30-min episode of restraint stress for 14 days and were sacrificed either 20 min or 24 h after their last stress/handling episode. Western blot analysis of two well-characterized AD-relevant pTau epitopes (AT8 and PHF-1) and upstream pTau mechanisms (e.g. GSK3β) analysis, showed that stressed post-maternal rats have increased pTau in comparison to stressed lactating rats 20 min after their last stress episode. Furthermore, an increase in pTau was also seen 24 h after the last stress episode in stressed post-maternal rats in comparison to their non-stressed controls in the detergent-soluble fraction. GSK3 analysis showed an increase in total levels of GSK3β in virgin rats and an increase of inactive levels of GSK3β in post-maternal rats, which suggests a different stress response in pTau after the rat has gone through the maternal experience. Interestingly, post-maternal rats also presented the more variability in their estrous cycles in response to stress. Besides no differences in pTau, non-stressed lactating rats showed an increase in inactive GSK3β 24 h after the last handling episode. Immunohistochemical detection of the PHF-1 epitope revealed increased pTau in the CA4/hilar subfield of the hippocampus of virgin and post-maternal rats exposed to chronic stress shortly after their last stress episode. Overall, lactating rats remained unresponsive to chronic restraint stress. These results suggest increased sensitivity of the virgin and post-maternal rats to hippocampal stress-induced pTau with chronic restraint stress compared to lactating rats. Because no differences were detected in response to stress by lactating rats and an exaggerated response was observed in post-maternal rats, current results support the hypothesis that lactation affects tau processing in the brain of the female. pTau increases in the hippocampus of stressed virgin and especially post-maternal rats but not in that of lactating dams. The hippocampal area CA4 of virgin and post-maternal rats is most affected by the chronic restraint stress. GSK3β overall levels and activity are modified by the reproductive condition and stress. Reproductive experience modifies pTau induced by chronic stress.
Collapse
|
25
|
Abstract
The central noradrenergic system comprises multiple brainstem nuclei whose cells synthesize and release the catecholamine transmitter norepinephrine (NE). The largest of these nuclei is the pontine locus coeruleus (LC), which innervates the vast majority of the forebrain. NE interacts with a number of pre- and postsynaptically expressed G protein-coupled receptors to affect a wide array of functions, including sensory signal processing, waking and arousal, stress responsiveness, mood, attention, and memory. Given the myriad functions ascribed to the locus coeruleus-noradrenergic (LC-NE) system, it is unsurprising that it is implicated in many disease states, including various mood, cognitive, neuropsychiatric, and neurodegenerative diseases. The LC-NE system is also notably sexually dimorphic with regard to its morphologic and anatomical features as well as how it responds to the peptide transmitter corticotropin releasing hormone (CRH), a major mediator of the central stress response. The sex-biased morphology and signaling that is observed in the LC could then be considered a potential contributor to the differential prevalence of various diseases between men and women. This chapter summarizes the primary differences between the male and female LC, based primarily on preclinical observations and how these disparities may relate to differential diagnoses of several diseases between men and women.
Collapse
Affiliation(s)
- Neal Joshi
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - Daniel Chandler
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States.
| |
Collapse
|
26
|
Wulaer B, Kunisawa K, Hada K, Jaya Suento W, Kubota H, Iida T, Kosuge A, Nagai T, Yamada K, Nitta A, Yamamoto Y, Saito K, Mouri A, Nabeshima T. Shati/Nat8l deficiency disrupts adult neurogenesis and causes attentional impairment through dopaminergic neuronal dysfunction in the dentate gyrus. J Neurochem 2020; 157:642-655. [PMID: 32275776 DOI: 10.1111/jnc.15022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/18/2022]
Abstract
Successful completion of daily activities relies on the ability to select the relevant features of the environment for memory and recall. Disruption to these processes can lead to various disorders, such as attention-deficit hyperactivity disorder (ADHD). Dopamine is a neurotransmitter implicated in the regulation of several processes, including attention. In addition to the higher-order brain function, dopamine is implicated in the regulation of adult neurogenesis. Previously, we generated mice lacking Shati, an N-acetyltransferase-8-like protein on a C57BL/6J genetic background (Shati/Nat8l-/- ). These mice showed a series of changes in the dopamine system and ADHD-like behavioral phenotypes. Therefore, we hypothesized that deficiency of Shati/Nat8l would affect neurogenesis and attentional behavior in mice. We found aberrant morphology of neurons and impaired neurogenesis in the dentate gyrus of Shati/Nat8l-/- mice. Additionally, research has suggested that impaired neurogenesis might be because of the reduction of dopamine in the hippocampus. Galantamine (GAL) attenuated the attentional impairment observed in the object-based attention test via increasing the dopamine release in the hippocampus of Shati/Nat8l-/- mice. The α7 nicotinic acetylcholine receptor antagonist, methyllycaconitine, and dopamine D1 receptor antagonist, SCH23390, blocked the ameliorating effect of GAL on attentional impairment in Shati/Nat8l-/- mice. These results suggest that the ameliorating effect of GAL on Shati/Nat8l-/- attentional impairment is associated with activation of D1 receptors following increased dopamine release in the hippocampus via α7 nicotinic acetylcholine receptor. In summary, Shati/Nat8l is important in both morphogenesis and neurogenesis in the dentate gyrus and attention, possible via modulation of dopaminergic transmission. Cover Image for this issue: https://doi.org/10.1111/jnc.15061.
Collapse
Affiliation(s)
- Bolati Wulaer
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Kazuhiro Hada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Willy Jaya Suento
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Department of Psychiatry, Hasanuddin University, South Sulawesi, Indonesia
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Tsubasa Iida
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Aika Kosuge
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Aichi, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Atsumi Nitta
- Department of Pharmaceutical Therapy and Neuropharmacology, Graduate School of Pharmaceutical Sciences, University of Toyama, Toyama, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Yasuko Yamamoto
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Kuniaki Saito
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| |
Collapse
|
27
|
Peña-Bautista C, Casas-Fernández E, Vento M, Baquero M, Cháfer-Pericás C. Stress and neurodegeneration. Clin Chim Acta 2020; 503:163-168. [PMID: 31987795 DOI: 10.1016/j.cca.2020.01.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases are a great concern because of aging worldwide population. Despite substantial effort to advance our understanding of the etiology and potential treatment of neurodegeneration, there remains a paucity of information with respect to this complex disease process. Interestingly, stress has been implicated among the potential mechanisms implicated in neurodegenerative pathology. Given the increase in chronic stress in modern society, this premise warrants further investigation. The aim of this review is to evaluate the influence of stress on neurodegeneration, the effect of neurodegenerative diseases diagnosis on stress, and therapeutic strategies for neurodegenerative diseases with a special focus on stress reduction. Neurodegenerative disorders such as Alzheimer's, Parkinson's and Huntington's Disease showed an acceleration in disease progression and a worsening of symptoms under stress. Some therapies (e.g., yoga, meditation) focused on reducing stress showed beneficial effects against neurodegeneration. Nevertheless, more studies are necessary in order to completely understand the implications of stress in neurodegeneration and the usefulness of stress reduction in the treatment thereof.
Collapse
Affiliation(s)
| | | | - Máximo Vento
- Health Research Institute La Fe, Valencia, Spain
| | - Miguel Baquero
- Division of Neurology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | | |
Collapse
|
28
|
Uribe KP, Correa VL, Pinales BE, Flores RJ, Cruz B, Shan Z, Bruijnzeel AW, Khan AM, O'Dell LE. Overexpression of corticotropin-releasing factor in the nucleus accumbens enhances the reinforcing effects of nicotine in intact female versus male and ovariectomized female rats. Neuropsychopharmacology 2020; 45:394-403. [PMID: 31614362 PMCID: PMC6901467 DOI: 10.1038/s41386-019-0543-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/01/2019] [Accepted: 10/07/2019] [Indexed: 12/18/2022]
Abstract
This study assessed the role of stress systems in the nucleus accumbens (NAc) in promoting sex differences in the reinforcing effects of nicotine. Intravenous self-administration (IVSA) of various doses of nicotine was compared following overexpression of corticotropin-releasing factor (CRF) in the NAc of female and male rats. Ovariectomized (OVX) females were also included to assess the role of ovarian hormones in promoting nicotine reinforcement. Rats received intra-NAc administration of an adeno-associated vector that overexpressed CRF (AAV2/5-CRF) or green fluorescent protein (AAV2/5-GFP). All rats were then given extended access (23 h/day) to an inactive and an active lever that delivered nicotine. Separate groups of rats received intra-NAc AAV2/5-CRF and saline IVSA. Rats were also allowed to nose-poke for food and water during IVSA testing. At the end of the study, the NAc was dissected and rt-qPCR methods were used to estimate CRF overexpression and changes in CRF receptors (CRFr1, CRFr2) and the CRF receptor internalizing protein, β-arrestin2 (Arrb2). Overexpression of CRF in the NAc increased nicotine IVSA to a larger extent in intact female versus male and OVX females. Food intake was increased to a larger extent in intact and OVX females as compared to males. The increase in CRF gene expression was similar across all groups; however, in females, overexpression of CRF resulted in a larger increase in CRFr1 and CRFr2 relative to males. In males, overexpression of CRF produced a larger increase in Arrb2 than females, suggesting greater CRF receptor internalization. Our results suggest that stress systems in the NAc promote the reinforcing effectiveness of nicotine in female rats in an ovarian hormone-dependent manner.
Collapse
Affiliation(s)
- Kevin P Uribe
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Victor L Correa
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Briana E Pinales
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Rodolfo J Flores
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Bryan Cruz
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI, 49931, USA
| | | | - Arshad M Khan
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Laura E O'Dell
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, 79968, USA.
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
29
|
Dominguez S, Rodriguez G, Fazelinia H, Ding H, Spruce L, Seeholzer SH, Dong H. Sex Differences in the Phosphoproteomic Profiles of APP/PS1 Mice after Chronic Unpredictable Mild Stress. J Alzheimers Dis 2020; 74:1131-1142. [PMID: 32144982 PMCID: PMC9843707 DOI: 10.3233/jad-191009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Approximately two-thirds of those suffering with Alzheimer's disease (AD) are women, however, the biological mechanisms underlying this sex divergence of AD prevalence remain unknown. Previous research has shown sex-specific biochemical differences that bias female mice toward pro-AD signaling on the phosphoproteomic level via corticotropin releasing factor (CRF) receptor 1 activation after CRF overexpression. Here we aimed to determine if chronic stress would induce a similar response in AD mouse models. We stressed 4-month-old APP/PS1 mice using a chronic unpredictable mild stress (CUMS) paradigm for up to 1 month. Following CUMS and behavioral assessments, we quantified whole protein and phosphoprotein levels in the cortex of stressed and non-stressed APP/PS1 mice using mass spectrometry-based proteomics. While there were no statistically significant differences at the total protein and peptide abundance levels, we found 909 and 841 statistically significant phosphopeptides between stressed and unstressed females and males, respectively, using a false discovery rate of 5%. Of these significant phosphopeptides, only 301 were the same in males and females. These results indicate that while both males and females undergo protein phosphorylation changes following stress, the peptides that are phosphorylated differ between sexes. We then used Metacore analysis to determine which biological pathways were affected. We found that several pathways were changed differently between male and female mice including NMDA receptor trafficking, cytoskeleton organization, and tau pathology. The differing biological pathways affected between males and females in response to chronic stress may help us to better understand why women are at a higher risk of AD.
Collapse
Affiliation(s)
- Sky Dominguez
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hossein Fazelinia
- Proteomics Core, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hua Ding
- Proteomics Core, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lynn Spruce
- Proteomics Core, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Steven H. Seeholzer
- Proteomics Core, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA,Correspondence to: Hongxin Dong, MD, PhD, Feinberg School of Medicine, Department of Psychiatry and Behavioral Sciences, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| |
Collapse
|
30
|
Wouters E, Walraed J, Robertson MJ, Meyrath M, Szpakowska M, Chevigné A, Skiniotis G, Stove C. Assessment of Biased Agonism among Distinct Synthetic Cannabinoid Receptor Agonist Scaffolds. ACS Pharmacol Transl Sci 2019; 3:285-295. [PMID: 32296768 DOI: 10.1021/acsptsci.9b00069] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Indexed: 12/13/2022]
Abstract
Cannabinoid receptor 1 (CB1) is a key drug target for a number of diseases, including metabolic syndromes and neuropathic pain. Most of the typical cannabinoid ligands provoke psychotropic side effects that impair their therapeutic utility. As of today, it is not yet clearly known which structural features of cannabinoid ligands determine a preference toward specific signaling pathways. Distinct bioassays are typically used to elucidate signaling preferences. However, these are often based on different cell lines and use different principles and/or read-outs, which makes straightforward assessment of "ligand bias" difficult. Within this context, this study is the first to investigate ligand bias among synthetic cannabinoid receptor agonists (SCRAs) in as closely analogous conditions as possible, by applying a new functional complementation-based assay panel to assess the recruitment of Gαi protein or β-arrestin2 to CB1. In a panel of 21 SCRAs, chosen to cover a broad diversity in chemical structures, distinct, although often subtle, preferences toward specific signaling pathways were observed. Relative to CP55940, here considered as a "balanced" reference agonist, most of the selected SCRAs (e.g., 5F-APINACA, CUMYL-PEGACLONE, among others) displayed preferred signaling through the β-arrestin2 pathway, whereas MMB-CHMICA could serve as a potential "balanced" agonist. Interestingly, EG-018 was the only SCRA showing a significant (10-fold) preference toward G protein over β-arrestin2 recruitment. While it is currently unclear what this exactly means in terms of abuse potential and/or toxicity, the approach proposed here may allow construction of a knowledge base that in the end may allow better insight into the structure-"functional" activity relationship of these compounds. This may aid the development of new therapeutics with less unwanted psychoactive effects.
Collapse
Affiliation(s)
- Elise Wouters
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Jolien Walraed
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Michael Joseph Robertson
- Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, 94305 California, United States.,Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, 94305 California, United States
| | - Max Meyrath
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen 1445, Luxembourg
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen 1445, Luxembourg
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Strassen 1445, Luxembourg
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, 94305 California, United States.,Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, 94305 California, United States
| | - Christophe Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| |
Collapse
|
31
|
Cao M, Li H, Zhao J, Cui J, Hu G. Identification of age- and gender-associated long noncoding RNAs in the human brain with Alzheimer's disease. Neurobiol Aging 2019; 81:116-126. [PMID: 31280115 PMCID: PMC6732230 DOI: 10.1016/j.neurobiolaging.2019.05.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 05/10/2019] [Accepted: 05/30/2019] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is an age- and gender-associated brain disorder. Long noncoding RNAs (lncRNAs) have emerged as key regulators of brain development, homeostasis, and pathologies. Here, we used gene array data sets and bioinformatics analysis to identify differentially expressed age- and gender-associated lncRNAs in human AD brains. We found that the expressions of 16 age-associated and 13 gender-associated lncRNAs were dysregulated in AD brains. Notably, the expressions of age-associated lncRNAs-SNHG19 and LINC00672-were significantly correlated with Braak stage of AD, positively and negatively, respectively, whereas the expressions of gender-associated lncRNAs-RNF144A-AS1, LY86-AS1, and LINC00639-were negatively correlated with Braak stage of AD. Functional analysis suggests that the pathways involved in neurodegenerative diseases, synaptic vesicle cycle, and endocytosis were overly represented within age- and gender-associated lncRNA-correlating genes. The identification of age- and gender-associated lncRNAs and their differential expressions in the human AD brain provide potential targets for further experimental validation and mechanistic investigation, which could, in turn, pave the way for developing age- and gender-specific prevention and adjunctive therapeutic options for patients with AD.
Collapse
Affiliation(s)
- Mei Cao
- Core Laboratory, School of Medicine, Sichuan Provincial People's Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Huaqing Li
- Department of Computer Science and Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jian Zhao
- Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu, People's Republic of China.
| | - Juan Cui
- Department of Computer Science and Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA.
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
32
|
Lauer AM, Schrode KM. Sex bias in basic and preclinical noise-induced hearing loss research. Noise Health 2019; 19:207-212. [PMID: 28937014 PMCID: PMC5644379 DOI: 10.4103/nah.nah_12_17] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Introduction: Sex differences in brain biochemistry, physiology, structure, and function have been gaining increasing attention in the scientific community. Males and females can have different responses to medications, diseases, and environmental variables. A small number of the approximately 7500 studies of noise-induced hearing loss (NIHL) have identified sex differences, but the mechanisms and characterization of these differences have not been thoroughly studied. The National Institutes of Health (NIH) issued a mandate in 2015 to include sex as a biological variable in all NIH-funded research beginning in January 2016. Materials and Methods: In the present study, the representation of sex as a biological variable in preclinical and basic studies of NIHL was quantified for a 5-year period from January 2011 to December 2015 prior to the implementation of the NIH mandate. Results: The analysis of 210 basic and preclinical studies showed that when sex is specified, experiments are predominantly performed on male animals. Discussion: This bias is present in studies completed in the United States and foreign institutions, and the proportion of studies using only male participants has actually increased over the 5-year period examined. Conclusion: These results underscore the need to invest resources in studying NIHL in both sexes to better understand how sex shapes the outcomes and to optimize treatment and prevention strategies.
Collapse
Affiliation(s)
- Amanda Marie Lauer
- Department of Otolaryngology-HNS and Center for Hearing and Balance, Johns Hopkins University, Baltimore, MD, United States
| | - Katrina Marie Schrode
- Department of Otolaryngology-HNS and Center for Hearing and Balance, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
33
|
Hupalo S, Bryce CA, Bangasser DA, Berridge CW, Valentino RJ, Floresco SB. Corticotropin-Releasing Factor (CRF) circuit modulation of cognition and motivation. Neurosci Biobehav Rev 2019; 103:50-59. [PMID: 31212019 DOI: 10.1016/j.neubiorev.2019.06.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 04/08/2019] [Accepted: 06/11/2019] [Indexed: 01/04/2023]
Abstract
The neuropeptide, corticotropin-releasing factor (CRF), is a key modulator of physiological, endocrine, and behavioral responses during stress. Dysfunction of the CRF system has been observed in stress-related affective disorders including post-traumatic stress disorder, depression, and anxiety. Beyond affective symptoms, these disorders are also characterized by impaired cognition, for which current pharmacological treatments are lacking. Thus, there is a need for pro-cognitive treatments to improve quality of life for individuals suffering from mental illness. In this review, we highlight research demonstrating that CRF elicits potent modulatory effects on higher-order cognition via actions within the prefrontal cortex and subcortical monoaminergic and cholinergic systems. Additionally, we identify questions for future preclinical research on this topic, such as the need to investigate sex differences in the cognitive and microcircuit actions of CRF, and whether CRF may represent a pharmacological target to treat cognitive dysfunction. Addressing these questions will provide new insight into pathophysiology underlying cognitive dysfunction and may lead to improved treatments for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sofiya Hupalo
- Integrative Neuroscience Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, United States.
| | - Courtney A Bryce
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Debra A Bangasser
- Psychology Department and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Craig W Berridge
- Department of Psychology, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Rita J Valentino
- National Institute on Drug Abuse, Bethesda, MD 20892, United States
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
34
|
Dioli C, Patrício P, Sousa N, Kokras N, Dalla C, Guerreiro S, Santos-Silva MA, Rego AC, Pinto L, Ferreiro E, Sotiropoulos I. Chronic stress triggers divergent dendritic alterations in immature neurons of the adult hippocampus, depending on their ultimate terminal fields. Transl Psychiatry 2019; 9:143. [PMID: 31028242 PMCID: PMC6486609 DOI: 10.1038/s41398-019-0477-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/02/2019] [Accepted: 03/08/2019] [Indexed: 12/18/2022] Open
Abstract
Chronic stress, a suggested precipitant of brain pathologies, such as depression and Alzheimer's disease, is known to impact on brain plasticity by causing neuronal remodeling as well as neurogenesis suppression in the adult hippocampus. Although many studies show that stressful conditions reduce the number of newborn neurons in the adult dentate gyrus (DG), little is known about whether and how stress impacts on dendritic development and structural maturation of these newborn neurons. We, herein, demonstrate that chronic stress impacts differentially on doublecortin (DCX)-positive immature neurons in distinct phases of maturation. Specifically, the density of the DCX-positive immature neurons whose dendritic tree reaches the inner molecular layer (IML) of DG is reduced in stressed animals, whereas their dendritic complexity is increased. On the contrary, no change on the density of DCX-positive neurons whose dendritic tree extends to the medial/outer molecular layer (M/OML) of the DG is found under stress conditions, whereas the dendritic complexity of these cells is diminished. In addition, DCX+ cells displayed a more complex and longer arbor in the dendritic compartments located in the granular cell layer of the DG under stress conditions; on the contrary, their dendritic segments localized into the M/OML were shorter and less complex. These findings suggest that the neuroplastic effects of chronic stress on dendritic maturation and complexity of DCX+ immature neurons vary based on the different maturation stage of DCX-positive cells and the different DG sublayer, highlighting the complex and dynamic stress-driven neuroplasticity of immature neurons in the adult hippocampus.
Collapse
Affiliation(s)
- Chrysoula Dioli
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Patrícia Patrício
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Nuno Sousa
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Nikolaos Kokras
- 0000 0001 2155 0800grid.5216.0First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece ,0000 0001 2155 0800grid.5216.0Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- 0000 0001 2155 0800grid.5216.0Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sara Guerreiro
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Miguel A. Santos-Silva
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Ana Cristina Rego
- 0000 0000 9511 4342grid.8051.cCenter for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute of Biochemistry, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Luísa Pinto
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Elisabete Ferreiro
- 0000 0000 9511 4342grid.8051.cCenter for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute for Interdisciplinary Research of the University of Coimbra (IIIUC), Coimbra, Portugal
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's - PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.
| |
Collapse
|
35
|
Affiliation(s)
- Hongxin Dong
- Department Psychiatry and Behavioral Sciences Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Department Psychiatry and Behavioral Sciences Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - John G Csernansky
- Department Psychiatry and Behavioral Sciences Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
36
|
Sase AS, Lombroso SI, Santhumayor BA, Wood RR, Lim CJ, Neve RL, Heller EA. Sex-Specific Regulation of Fear Memory by Targeted Epigenetic Editing of Cdk5. Biol Psychiatry 2019; 85:623-634. [PMID: 30661667 DOI: 10.1016/j.biopsych.2018.11.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/29/2018] [Accepted: 11/15/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Sex differences in the expression and prevalence of trauma- and stress-related disorders have led to a growing interest in the sex-specific molecular and epigenetic mechanisms underlying these diseases. Cyclin-dependent kinase 5 (CDK5) is known to underlie both fear memory and stress behavior in male mice. Given our recent finding that targeted histone acetylation of Cdk5 regulates stress responsivity in male mice, we hypothesized that such a mechanism may be functionally relevant in female mice as well. METHODS We applied epigenetic editing of Cdk5 in the hippocampus and examined the regulation of fear memory retrieval in male and female mice. Viral expression of zinc finger proteins targeting histone acetylation to the Cdk5 promoter was paired with a quantification of learning and memory of contextual fear conditioning, expression of CDK5, and enrichment of histone modifications of the Cdk5 gene. RESULTS We found that male mice exhibit stronger long-term memory retrieval than do female mice, and this finding was associated with male-specific epigenetic activation of hippocampal Cdk5 expression. Sex differences in behavior and epigenetic regulation of Cdk5 occurred after long-term, but not short-term, fear memory retrieval. Finally, targeted histone acetylation of hippocampal Cdk5 promoter attenuated fear memory retrieval and increased tau phosphorylation in female but not male mice. CONCLUSIONS Epigenetic editing uncovered a female-specific role of Cdk5 activation in attenuating fear memory retrieval. This finding may be attributed to CDK5 mediated hyperphosphorylation of tau only in the female hippocampus. Sex-specific epigenetic regulation of Cdk5 may reflect differences in the effect of CDK5 on downstream target proteins that regulate memory.
Collapse
Affiliation(s)
- Ajinkya S Sase
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sonia I Lombroso
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brandon A Santhumayor
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rozalyn R Wood
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carissa J Lim
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, Boston, Massachusetts
| | - Elizabeth A Heller
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania; Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
37
|
Meta-analysis of expression and methylation signatures indicates a stress-related epigenetic mechanism in multiple neuropsychiatric disorders. Transl Psychiatry 2019; 9:32. [PMID: 30670680 PMCID: PMC6342918 DOI: 10.1038/s41398-018-0358-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/12/2018] [Accepted: 12/09/2018] [Indexed: 02/02/2023] Open
Abstract
Similar environmental risk factors have been implicated in different neuropsychiatric disorders (including major psychiatric and neurodegenerative diseases), indicating the existence of common epigenetic mechanisms underlying the pathogenesis shared by different illnesses. To investigate such commonality, we applied an unsupervised computational approach identifying several consensus co-expression and co-methylation signatures from a data cohort of postmortem prefrontal cortex (PFC) samples from individuals with six different neuropsychiatric disorders-schizophrenia, bipolar disorder, major depression, alcoholism, Alzheimer's and Parkinson's-as well as healthy controls. Among our results, we identified a pair of strongly interrelated co-expression and co-methylation (E-M) signatures showing consistent and significant disease association in multiple types of disorders. This E-M signature was enriched for interneuron markers, and we further demonstrated that it is unlikely for this enrichment to be due to varying subpopulation abundance of normal interneurons across samples. Moreover, gene set enrichment analysis revealed overrepresentation of stress-related biological processes in this E-M signature. Our integrative analysis of expression and methylation profiles, therefore, suggests a stress-related epigenetic mechanism in the brain, which could be associated with the pathogenesis of multiple neuropsychiatric diseases.
Collapse
|
38
|
Medeiros ADM, Silva RH. Sex Differences in Alzheimer’s Disease: Where Do We Stand? J Alzheimers Dis 2019; 67:35-60. [DOI: 10.3233/jad-180213] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- André de Macêdo Medeiros
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Center of Health and Biological Sciences, Universidade Federal Rural do Semiárido, Mossoró, Brazil
| | - Regina Helena Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
39
|
Sex differences in stress reactivity in arousal and attention systems. Neuropsychopharmacology 2019; 44:129-139. [PMID: 30022063 PMCID: PMC6235989 DOI: 10.1038/s41386-018-0137-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/21/2018] [Accepted: 06/15/2018] [Indexed: 01/04/2023]
Abstract
Women are more likely than men to suffer from psychiatric disorders with hyperarousal symptoms, including posttraumatic stress disorder (PTSD) and major depression. In contrast, women are less likely than men to be diagnosed with schizophrenia and attention deficit hyperactivity disorder (ADHD), which share attentional impairments as a feature. Stressful events exacerbate symptoms of the aforementioned disorders. Thus, researchers are examining whether sex differences in stress responses bias women and men towards different psychopathology. Here we review the preclinical literature suggesting that, compared to males, females are more vulnerable to stress-induced hyperarousal, while they are more resilient to stress-induced attention deficits. Specifically described are sex differences in receptors for the stress neuropeptide, corticotropin-releasing factor (CRF), that render the locus coeruleus arousal system of females more vulnerable to stress and less adaptable to CRF hypersecretion, a condition found in patients with PTSD and depression. Studies on the protective effects of ovarian hormones against CRF-induced deficits in sustained attention are also detailed. Importantly, we highlight how comparing males and females in preclinical studies can lead to the development of novel therapeutics to improve treatments for psychiatric disorders in both women and men.
Collapse
|
40
|
CRF modulation of central monoaminergic function: Implications for sex differences in alcohol drinking and anxiety. Alcohol 2018; 72:33-47. [PMID: 30217435 DOI: 10.1016/j.alcohol.2018.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/03/2018] [Accepted: 01/19/2018] [Indexed: 01/06/2023]
Abstract
Decades of research have described the importance of corticotropin-releasing factor (CRF) signaling in alcohol addiction, as well as in commonly co-expressed neuropsychiatric diseases, including anxiety and mood disorders. However, CRF signaling can also acutely regulate binge alcohol consumption, anxiety, and affect in non-dependent animals, possibly via modulation of central monoaminergic signaling. We hypothesize that basal CRF tone is particularly high in animals and humans with an inherent propensity for high anxiety and alcohol consumption, and thus these individuals are at increased risk for the development of alcohol use disorder and comorbid neuropsychiatric diseases. The current review focuses on extrahypothalamic CRF circuits, particularly those stemming from the bed nucleus of the stria terminalis (BNST), found to play a role in basal phenotypes, and examines whether the intrinsic hyperactivity of these circuits is sufficient to escalate the expression of these behaviors and steepen the trajectory of development of disease states. We focus our efforts on describing CRF modulation of biogenic amine neuron populations that have widespread projections to the forebrain to modulate behaviors, including alcohol and drug intake, stress reactivity, and anxiety. Further, we review the known sex differences and estradiol modulation of these neuron populations and CRF signaling at their synapses to address the question of whether females are more susceptible to the development of comorbid addiction and stress-related neuropsychiatric diseases because of hyperactive extrahypothalamic CRF circuits compared to males.
Collapse
|
41
|
Newcombe EA, Camats-Perna J, Silva ML, Valmas N, Huat TJ, Medeiros R. Inflammation: the link between comorbidities, genetics, and Alzheimer's disease. J Neuroinflammation 2018; 15:276. [PMID: 30249283 PMCID: PMC6154824 DOI: 10.1186/s12974-018-1313-3] [Citation(s) in RCA: 356] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder, most cases of which lack a clear causative event. This has made the disease difficult to characterize and, thus, diagnose. Although some cases are genetically linked, there are many diseases and lifestyle factors that can lead to an increased risk of developing AD, including traumatic brain injury, diabetes, hypertension, obesity, and other metabolic syndromes, in addition to aging. Identifying common factors and trends between these conditions could enhance our understanding of AD and lead to the development of more effective treatments. Although the immune system is one of the body’s key defense mechanisms, chronic inflammation has been increasingly linked with several age-related diseases. Moreover, it is now well accepted that chronic inflammation has an important role in the onset and progression of AD. In this review, the different inflammatory signals associated with AD and its risk factors will be outlined to demonstrate how chronic inflammation may be influencing individual susceptibility to AD. Our goal is to bring attention to potential shared signals presented by the immune system during different conditions that could lead to the development of successful treatments.
Collapse
Affiliation(s)
- Estella A Newcombe
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.
| | - Judith Camats-Perna
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia
| | - Mallone L Silva
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia
| | - Nicholas Valmas
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Tee Jong Huat
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.,Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Rodrigo Medeiros
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.
| |
Collapse
|
42
|
McAlinn HR, Reich B, Contoreggi NH, Kamakura RP, Dyer AG, McEwen BS, Waters EM, Milner TA. Sex Differences in the Subcellular Distribution of Corticotropin-Releasing Factor Receptor 1 in the Rat Hippocampus following Chronic Immobilization Stress. Neuroscience 2018; 383:98-113. [PMID: 29753863 PMCID: PMC5994383 DOI: 10.1016/j.neuroscience.2018.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/01/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
Abstract
Corticotropin-releasing factor receptors (CRFR1) contribute to stress-induced adaptations in hippocampal structure and function that can affect learning and memory processes. Our prior studies showed that female rats with elevated estrogens compared to males have more plasmalemmal CRFR1 in CA1 pyramidal cells, suggesting a greater sensitivity to stress. Here, we examined the distribution of hippocampal CRFR1 following chronic immobilization stress (CIS) in female and male rats using immuno-electron microscopy. Without stress, total CRFR1 dendritic levels were higher in females in CA1 and in males in the hilus; moreover, plasmalemmal CRFR1 was elevated in pyramidal cell dendrites in CA1 in females and in CA3 in males. Following CIS, near-plasmalemmal CRFR1 increased in CA1 pyramidal cell dendrites in males but not to levels of control or CIS females. In CA3 and the hilus, CIS decreased cytoplasmic and total CRFR1 in dendrites in males only. These results suggest that in naive rats, CRF could induce a greater activation of CA1 pyramidal cells in females than males. Moreover, after CIS, which leads to even greater sex differences in CRFR1 by trafficking it to different subcellular compartments, CRF could enhance activation of CA1 pyramidal cells in males but to a lesser extent than either unstressed or CIS females. Additionally, CA3 pyramidal cells and inhibitory interneurons in males have heightened sensitivity to CRF, regardless of stress state. These sex differences in CRFR1 distribution and trafficking in the hippocampus may contribute to reported sex differences in hippocampus-dependent learning processes in baseline conditions and following chronic stress.
Collapse
Affiliation(s)
- Helena R McAlinn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Batsheva Reich
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Andreina G Dyer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Elizabeth M Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
43
|
Precision medicine and drug development in Alzheimer's disease: the importance of sexual dimorphism and patient stratification. Front Neuroendocrinol 2018; 50:31-51. [PMID: 29902481 DOI: 10.1016/j.yfrne.2018.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/29/2018] [Accepted: 06/07/2018] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (ND) are among the leading causes of disability and mortality. Considerable sex differences exist in the occurrence of the various manifestations leading to cognitive decline. Alzheimer's disease (AD) exhibits substantial sexual dimorphisms and disproportionately affects women. Women have a higher life expectancy compared to men and, consequently, have more lifespan to develop AD. The emerging precision medicine and pharmacology concepts - taking into account the individual genetic and biological variability relevant for disease risk, prevention, detection, diagnosis, and treatment - are expected to substantially enhance our knowledge and management of AD. Stratifying the affected individuals by sex and gender is an important basic step towards personalization of scientific research, drug development, and care. We hypothesize that sex and gender differences, extending from genetic to psychosocial domains, are highly relevant for the understanding of AD pathophysiology, and for the conceptualization of basic/translational research and for clinical therapy trial design.
Collapse
|
44
|
Wickens MM, Bangasser DA, Briand LA. Sex Differences in Psychiatric Disease: A Focus on the Glutamate System. Front Mol Neurosci 2018; 11:197. [PMID: 29922129 PMCID: PMC5996114 DOI: 10.3389/fnmol.2018.00197] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/18/2018] [Indexed: 12/21/2022] Open
Abstract
Alterations in glutamate, the primary excitatory neurotransmitter in the brain, are implicated in several psychiatric diseases. Many of these psychiatric diseases display epidemiological sex differences, with either males or females exhibiting different symptoms or disease prevalence. However, little work has considered the interaction of disrupted glutamatergic transmission and sex on disease states. This review describes the clinical and preclinical evidence for these sex differences with a focus on two conditions that are more prevalent in women: Alzheimer's disease and major depressive disorder, and three conditions that are more prevalent in men: schizophrenia, autism spectrum disorder, and attention deficit hyperactivity disorder. These studies reveal sex differences at multiple levels in the glutamate system including metabolic markers, receptor levels, genetic interactions, and therapeutic responses to glutamatergic drugs. Our survey of the current literature revealed a considerable need for more evaluations of sex differences in future studies examining the role of the glutamate system in psychiatric disease. Gaining a more thorough understanding of how sex differences in the glutamate system contribute to psychiatric disease could provide novel avenues for the development of sex-specific pharmacotherapies.
Collapse
Affiliation(s)
- Megan M Wickens
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States.,Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Lisa A Briand
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States.,Neuroscience Program, Temple University, Philadelphia, PA, United States
| |
Collapse
|
45
|
Di Segni M, Andolina D, Ventura R. Long-term effects of early environment on the brain: Lesson from rodent models. Semin Cell Dev Biol 2018; 77:81-92. [DOI: 10.1016/j.semcdb.2017.09.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/20/2017] [Accepted: 09/29/2017] [Indexed: 12/21/2022]
|
46
|
Stress as risk factor for Alzheimer's disease. Pharmacol Res 2018; 132:130-134. [PMID: 29689315 DOI: 10.1016/j.phrs.2018.04.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 11/22/2022]
Abstract
Prolonged stress predisposes susceptible individuals to a number of physiological disorders including cardiovascular disease, obesity and gastrointestinal disorders, as well as psychiatric and neurodegenerative disorders. Preclinical studies have suggested that manipulation of the glucocorticoid milieu can trigger cellular, molecular and behavioral derangement resembling the hallmarks of Alzheimer's Disease (AD). For example, stress or glucocorticoid administration can increase amyloid ß precursor protein and tau phosphorylation which are involved in synaptic dysfunction and neuronal death associated with AD. Although since AD was first described in 1906 at a conference in Tubingen, Germany by Alois Alzheimer our knowledge of neuropathological and neurochemical alterations of AD has been impressively increased, at present, pharmacotherapy is symptomatic at best and has no influence on the progression of the disorder. It is generally believed that most of the drugs developed as disease modifiers have failed in clinical trials because treatment started too late, i.e., after the clinical onset of AD. Because AD pathology begins several years prior to the clinical diagnosis, it is imperative to identify subjects at high risk to develop the disorder. Consequently, the search for putative risk factors has gained importance. ApoE4, diabetes/metabolic syndrome, cardiovascular disorders, and a low cognitive reserve are established risk factors for AD. The focus of this review is on stress and glucocorticoids as potential factors increasing the risk to develop AD.
Collapse
|
47
|
Fisher DW, Bennett DA, Dong H. Sexual dimorphism in predisposition to Alzheimer's disease. Neurobiol Aging 2018; 70:308-324. [PMID: 29754747 DOI: 10.1016/j.neurobiolaging.2018.04.004] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022]
Abstract
Clinical studies indicate that Alzheimer's disease (AD) disproportionately affects women in both disease prevalence and rate of symptom progression, but the mechanisms underlying this sexual divergence are unknown. Although some have suggested this difference in risk is a reflection of the known differences in longevity between men and women, mounting clinical and preclinical evidence supports women also having intrinsic susceptibilities toward the disease. Although a number of potential risk factors have been hypothesized to mediate these differences, none have been definitively verified. In this review, we first summarize the epidemiologic studies of prevalence and incidence of AD among the sexes. Next, we discuss the most likely risk factors to date that interact with biological sex, including (1) genetic factors, (2) sex hormones (3) deviations in brain structure, (4) inflammation and microglia, and (5) and psychosocial stress responses. Overall, though differences in life span are likely to account for part of the divide between the sexes in AD prevalence, the abundance of preclinical and clinical evidence presented here suggests an increase in intrinsic AD risk for women. Therefore, future studies focusing on the underlying biological mechanisms for this phenomenon are needed to better understand AD pathogenesis in both sexes, with the eventual goal of sex-specific prevention and treatment strategies.
Collapse
Affiliation(s)
- Daniel W Fisher
- Departments of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, USA
| | - Hongxin Dong
- Departments of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
48
|
Dong H, Keegan JM, Hong E, Gallardo C, Montalvo-Ortiz J, Wang B, Rice KC, Csernansky J. Corticotrophin releasing factor receptor 1 antagonists prevent chronic stress-induced behavioral changes and synapse loss in aged rats. Psychoneuroendocrinology 2018; 90:92-101. [PMID: 29477954 PMCID: PMC5864558 DOI: 10.1016/j.psyneuen.2018.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/25/2018] [Accepted: 02/14/2018] [Indexed: 11/24/2022]
Abstract
Mounting evidence suggests that chronic stress can alter brain structure and function and promote the development of neuropsychiatric disorders, such as depression and Alzheimer's disease. Although the results of several studies have indicated that aged brains are more vulnerable to chronic stress, it remains unknown whether antagonists of a key stress regulator, the corticotrophin releasing factor receptor 1 (CRF1), can prevent stress-induced anxiety and memory deficits in animal models. In this study, we evaluated the potential benefits of two CRF1 antagonists, R121919 and antalarmin, for preventing stress-induced anxiety-related behavioral and memory deficits and neurodegeneration in aged rats. We stressed rats using isolation-restraint for 3 months starting from the 18 months of age. Subsets of animals were administrated either R121919 or antalarmin through food chow for 3 months, followed by a series of behavioral, biochemical and morphological analyses. We found that stressed aged rats displayed body weight losses and increased corticosterone levels, as well as anxiety-related behaviors and memory deficits. Additionally, chronic stress induced a loss of cortical dendritic spines and synapses. However, R121919 and antalarmin both prevented stress-induced behavioral changes including anxiety-related behaviors and memory deficits and prevented synapse loss, perhaps through reversing HPA axis dysfunction. These results suggest that CRF1 antagonists may hold promise as a potential therapy for preventing stress-induced anxiety and memory deficits in aged individuals.
Collapse
Affiliation(s)
- Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| | - Jack M Keegan
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Ellie Hong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Christopher Gallardo
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Janitza Montalvo-Ortiz
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Becky Wang
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Kenner C. Rice
- National Institute on Drug Abuse, and National Institute Alcohol Abuse and Alcoholism Intramural Research Program, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - John Csernansky
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| |
Collapse
|
49
|
Bangasser DA, Wiersielis KR. Sex differences in stress responses: a critical role for corticotropin-releasing factor. Hormones (Athens) 2018; 17:5-13. [PMID: 29858858 DOI: 10.1007/s42000-018-0002-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 11/03/2017] [Indexed: 12/19/2022]
Abstract
Rates of post-traumatic stress disorder, panic disorder, and major depression are higher in women than in men. Another shared feature of these disorders is that dysregulation of the stress neuropeptide, corticotropin-releasing factor (CRF), is thought to contribute to their pathophysiology. Therefore, sex differences in responses to CRF could contribute to this sex bias in disease prevalence. Here, we review emerging data from non-human animal models that reveal extensive sex differences in CRF functions ranging from its presynaptic regulation to its postsynaptic efficacy. Specifically, detailed are sex differences in the regulation of CRF-containing neurons and the amount of CRF that they produce. We also describe sex differences in CRF receptor expression, distribution, trafficking, and signaling. Finally, we highlight sex differences in the processes that mitigate the effects of CRF. In most cases, the identified sex differences can lead to increased stress sensitivity in females. Thus, the relevance of these differences for the increased risk of depression and anxiety disorders in women compared to men is also discussed.
Collapse
Affiliation(s)
- Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, 1701 North 13th Street, 873 Weiss Hall, Philadelphia, PA, 19122, USA.
| | - Kimberly R Wiersielis
- Department of Psychology and Neuroscience Program, Temple University, 1701 North 13th Street, 873 Weiss Hall, Philadelphia, PA, 19122, USA
| |
Collapse
|
50
|
Yuede CM, Timson BF, Hettinger JC, Yuede KM, Edwards HM, Lawson JE, Zimmerman SD, Cirrito JR. Interactions between stress and physical activity on Alzheimer's disease pathology. Neurobiol Stress 2018; 8:158-171. [PMID: 29888311 PMCID: PMC5991353 DOI: 10.1016/j.ynstr.2018.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 02/20/2018] [Indexed: 12/14/2022] Open
Abstract
Physical activity and stress are both environmental modifiers of Alzheimer's disease (AD) risk. Animal studies of physical activity in AD models have largely reported positive results, however benefits are not always observed in either cognitive or pathological outcomes and inconsistencies among findings remain. Studies using forced exercise may increase stress and mitigate some of the benefit of physical activity in AD models, while voluntary exercise regimens may not achieve optimal intensity to provide robust benefit. We evaluated the findings of studies of voluntary and forced exercise regimens in AD mouse models to determine the influence of stress, or the intensity of exercise needed to outweigh the negative effects of stress on AD measures. In addition, we show that chronic physical activity in a mouse model of AD can prevent the effects of acute restraint stress on Aβ levels in the hippocampus. Stress and physical activity have many overlapping and divergent effects on the body and some of the possible mechanisms through which physical activity may protect against stress-induced risk factors for AD are discussed. While the physiological effects of acute stress and acute exercise overlap, chronic effects of physical activity appear to directly oppose the effects of chronic stress on risk factors for AD. Further study is needed to identify optimal parameters for intensity, duration and frequency of physical activity to counterbalance effects of stress on the development and progression of AD.
Collapse
Affiliation(s)
- Carla M Yuede
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.,Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Benjamin F Timson
- Biomedical Science Department, Missouri State University, Springfield, MO, USA
| | - Jane C Hettinger
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Kayla M Yuede
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Hannah M Edwards
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Justin E Lawson
- Biomedical Science Department, Missouri State University, Springfield, MO, USA
| | - Scott D Zimmerman
- Biomedical Science Department, Missouri State University, Springfield, MO, USA
| | - John R Cirrito
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| |
Collapse
|