1
|
Vanderheyden WM, Lefton M, Flores CC, Owada Y, Gerstner JR. Fabp7 Is Required for Normal Sleep Suppression and Anxiety-Associated Phenotype following Single-Prolonged Stress in Mice. NEUROGLIA (BASEL, SWITZERLAND) 2022; 3:73-83. [PMID: 36909794 PMCID: PMC10001429 DOI: 10.3390/neuroglia3020005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Humans with post-traumatic stress disorder (PTSD) exhibit sleep disturbances that include insomnia, nightmares, and enhanced daytime sleepiness. Sleep disturbances are considered a hallmark feature of PTSD; however, little is known about the cellular and molecular mechanisms regulating trauma-induced sleep disorders. Using a rodent model of PTSD called "Single Prolonged Stress" (SPS) we examined the requirement of the brain-type fatty acid binding protein Fabp7, an astrocyte expressed lipid-signaling molecule, in mediating trauma-induced sleep disturbances. We measured baseline sleep/wake parameters and then exposed Fabp7 knock-out (KO) and wild-type (WT) C57BL/6N genetic background control animals to SPS. Sleep and wake measurements were obtained immediately following the initial trauma exposure of SPS, and again 7 days later. We found that active-phase (dark period) wakefulness was similar in KO and WT at baseline and immediately following SPS; however, it was significantly increased after 7 days. These effects were opposite in the inactive-phase (light period), where KOs exhibited increased wake in baseline and following SPS, but returned to WT levels after 7 days. To examine the effects of Fabp7 on unconditioned anxiety following trauma, we exposed KO and WT mice to the light-dark box test before and after SPS. Prior to SPS, KO and WT mice spent similar amounts of time in the lit compartment. Following SPS, KO mice spent significantly more time in the lit compartment compared to WT mice. These results demonstrate that mutations in an astrocyte-expressed gene (Fabp7) influence changes in stress-dependent sleep disturbances and associated anxiety behavior.
Collapse
Affiliation(s)
- William M. Vanderheyden
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Correspondence: (W.M.V.); (J.R.G.)
| | - Micah Lefton
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Carlos C. Flores
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Seiryo-cho 2-1, Aobaku, Sendai 980-8575, Japan
| | - Jason R. Gerstner
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA
- Correspondence: (W.M.V.); (J.R.G.)
| |
Collapse
|
2
|
Even‐Chen O, Herburg L, Kefalakes E, Urshansky N, Grothe C, Barak S. FGF2 is an endogenous regulator of alcohol reward and consumption. Addict Biol 2022; 27:e13115. [PMID: 34796591 DOI: 10.1111/adb.13115] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 11/30/2022]
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing disorder, characterized by escalating alcohol drinking and loss of control, with very limited available treatments. We recently reported that the expression of fibroblast growth factor 2 (Fgf2) is increased in the striatum of rodents following long-term excessive alcohol drinking and that the systemic or intra-striatal administration of recombinant FGF2 increases alcohol consumption. Here, we set out to determine whether the endogenous FGF2 plays a role in alcohol drinking and reward, by testing the behavioural phenotype of Fgf2 knockout mice. We found that Fgf2 deficiency resulted in decreased alcohol consumption when tested in two-bottle choice procedures with various alcohol concentrations. Importantly, these effects were specific for alcohol, as a natural reward (sucrose) or water consumption was not affected by Fgf2 deficiency. In addition, Fgf2 knockout mice failed to show alcohol-conditioned place preference (CPP) but showed normal fear conditioning, suggesting that deletion of the growth factor reduces alcohol's rewarding properties. Finally, Fgf2 knockout mice took longer to recover from the loss of righting reflex and showed higher blood alcohol concentrations when challenged with an intoxicating alcohol dose, suggesting that their ethanol metabolism might be affected. Together, our results show that the endogenous FGF2 plays a critical role in alcohol drinking and reward and indicate that FGF2 is a positive regulator of alcohol-drinking behaviours. Our findings suggest that FGF2 is a potential biomarker for problem alcohol drinking and is a potential target for pharmacotherapy development for AUD.
Collapse
Affiliation(s)
- Oren Even‐Chen
- School of Psychological Sciences Tel Aviv University Tel Aviv 69978 Israel
| | - Leonie Herburg
- Institute of Neuroanatomy and Cell Biology Hannover Medical School Carl‐Neuberg‐Straße 1 Hanover 30625 Germany
- Center for Systems Neuroscience (ZSN) Hannover Germany
| | - Ekaterini Kefalakes
- Institute of Neuroanatomy and Cell Biology Hannover Medical School Carl‐Neuberg‐Straße 1 Hanover 30625 Germany
- Center for Systems Neuroscience (ZSN) Hannover Germany
| | - Nataly Urshansky
- School of Psychological Sciences Tel Aviv University Tel Aviv 69978 Israel
| | - Claudia Grothe
- Institute of Neuroanatomy and Cell Biology Hannover Medical School Carl‐Neuberg‐Straße 1 Hanover 30625 Germany
- Center for Systems Neuroscience (ZSN) Hannover Germany
| | - Segev Barak
- School of Psychological Sciences Tel Aviv University Tel Aviv 69978 Israel
- Sagol School of Neuroscience Tel Aviv University Tel Aviv 69978 Israel
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences Tel Aviv University Tel Aviv 69978 Israel
| |
Collapse
|
3
|
Jeong M, Bojkovic K, Sagi V, Stankovic KM. Molecular and Clinical Significance of Fibroblast Growth Factor 2 in Development and Regeneration of the Auditory System. Front Mol Neurosci 2022; 14:757441. [PMID: 35002617 PMCID: PMC8733209 DOI: 10.3389/fnmol.2021.757441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/18/2021] [Indexed: 01/25/2023] Open
Abstract
The fibroblast growth factor 2 (FGF2) is a member of the FGF family which is involved in key biological processes including development, cellular proliferation, wound healing, and angiogenesis. Although the utility of the FGF family as therapeutic agents has attracted attention, and FGF2 has been studied in several clinical contexts, there remains an incomplete understanding of the molecular and clinical function of FGF2 in the auditory system. In this review, we highlight the role of FGF2 in inner ear development and hearing protection and present relevant clinical studies for tympanic membrane (TM) repair. We conclude by discussing the future implications of FGF2 as a potential therapeutic agent.
Collapse
Affiliation(s)
- Minjin Jeong
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| | - Katarina Bojkovic
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| | - Varun Sagi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,University of Minnesota Medical School, Minneapolis, MN, United States
| | - Konstantina M Stankovic
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Bryant EM, Richardson R, Graham BM. The Association Between Salivary FGF2 and Physiological and Psychological Components of the Human Stress Response. CHRONIC STRESS 2022; 6:24705470221114787. [PMID: 35874911 PMCID: PMC9297468 DOI: 10.1177/24705470221114787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/26/2022] [Accepted: 07/05/2022] [Indexed: 11/20/2022]
Abstract
Background Fibroblast Growth Factor 2 (FGF2) is a neurotrophic protein that has been implicated as a biomarker for anxiety and depressive disorders, which comprise a significant component of the global burden of disease. Research using rodents has indicated that FGF2 is part of the stress response, but whether this translates to humans has yet to be investigated. In this study, we aimed to explore the potential role of FGF2 in the human stress response by examining its association with physiological and psychological processes during and following the Trier Social Stress Test (TSST). Methods Participants in the active stress experiment (N = 87) underwent the TSST, provided saliva samples to obtain levels of cortisol and FGF2, and reported on post-event rumination related to the TSST task over the following week. Participants in the no-stress experiment (N = 25) provided saliva samples for measurement of FGF2 and cortisol across a corresponding time period. Results Salivary FGF2 levels changed after the TSST and were associated with the pattern of change in salivary cortisol. Cortisol responses in the active stress condition were blunted in females (relative to males), however, sex did not interact with any other effect. FGF2 reactivity (ie, the magnitude of change over time) was not correlated with cortisol reactivity. Lower FGF2 reactivity following the TSST, but not overall FGF2 levels, or cortisol, was associated with higher fear of negative evaluation, repetitive negative thinking and post-event processing, as well as repetitive negative thinking in the week following the TSST. Participants in the no-stress experiment showed a decrease in cortisol, yet no change in their FGF2 levels. Conclusion These findings suggest that FGF2 is involved in the human stress response and higher levels of FGF2 reactivity may be associated with protective cognitive processes following stress exposure.
Collapse
Affiliation(s)
- Emma M. Bryant
- University of New South Wales, School of Psychology, Sydney, NSW 2052, Australia
| | - Rick Richardson
- University of New South Wales, School of Psychology, Sydney, NSW 2052, Australia
| | - Bronwyn M. Graham
- University of New South Wales, School of Psychology, Sydney, NSW 2052, Australia
| |
Collapse
|
5
|
Zhang L, Xu H, Ding N, Li X, Chen X, Chen Z. Beneficial Effects on Brain Micro-Environment by Caloric Restriction in Alleviating Neurodegenerative Diseases and Brain Aging. Front Physiol 2021; 12:715443. [PMID: 34899367 PMCID: PMC8660583 DOI: 10.3389/fphys.2021.715443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022] Open
Abstract
Aging and neurodegenerative diseases are frequently associated with the disruption of the extracellular microenvironment, which includes mesenchyme and body fluid components. Caloric restriction (CR) has been recognized as a lifestyle intervention that can improve long-term health. In addition to preventing metabolic disorders, CR has been shown to improve brain health owing to its enhancing effect on cognitive functions or retarding effect on the progression of neurodegenerative diseases. This article summarizes current findings regarding the neuroprotective effects of CR, which include the modulation of metabolism, autophagy, oxidative stress, and neuroinflammation. This review may offer future perspectives for brain aging interventions.
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Huachong Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Ning Ding
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Medical College, Kunming University of Science and Technology, Kunming, China
| | - Xue Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Xiaoyin Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhuangfei Chen
- Medical College, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
6
|
Dremencov E, Jezova D, Barak S, Gaburjakova J, Gaburjakova M, Kutna V, Ovsepian SV. Trophic factors as potential therapies for treatment of major mental disorders. Neurosci Lett 2021; 764:136194. [PMID: 34433100 DOI: 10.1016/j.neulet.2021.136194] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 08/02/2021] [Accepted: 08/20/2021] [Indexed: 12/20/2022]
Abstract
Notwithstanding major advances in psychotherapeutics, their efficacy and specificity remain limited. The slow onset of beneficial outcomes and numerous adverse effects of widely used medications remain of chief concern, warranting in-depth studies. The majority of frontline therapies are thought to enhance the endogenous monoaminergic drive, to initiate a cascade of molecular events leading to lasting functional and structural plasticity. They also involve alterations in trophic factor signalling, including brain-derived neurotrophic factor (BDNF), VGF (non-acronymic), vascular endothelial growth factor (VEGF), fibroblast growth factor 2 (FGF2), glial cell-derived neurotrophic factor (GDNF), and others. In several major mental disorders, emerging data suggest protective and restorative effects of trophic factors in preclinical models, when applied on their own. Antidepressant outcomes of VGF and FGF2, for instance, were shown in experimental animals, while BDNF and GDNF prove useful in the treatment of addiction, schizophrenia, and autism spectrum disorders. The main challenge with the effective translation of these and other findings in the clinic is the knowledge gap in action mechanisms with potential risks, as well as the lack of effective platforms for validation under clinical settings. Herein, we review the state-of-the-art and advances in the therapeutic use of trophic factors in several major neuropsychiatric disorders.
Collapse
Affiliation(s)
- Eliyahu Dremencov
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Segev Barak
- School of Psychological Sciences and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Jana Gaburjakova
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Marta Gaburjakova
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Viera Kutna
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Saak V Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| |
Collapse
|
7
|
Klimaschewski L, Claus P. Fibroblast Growth Factor Signalling in the Diseased Nervous System. Mol Neurobiol 2021; 58:3884-3902. [PMID: 33860438 PMCID: PMC8280051 DOI: 10.1007/s12035-021-02367-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Fibroblast growth factors (FGFs) act as key signalling molecules in brain development, maintenance, and repair. They influence the intricate relationship between myelinating cells and axons as well as the association of astrocytic and microglial processes with neuronal perikarya and synapses. Advances in molecular genetics and imaging techniques have allowed novel insights into FGF signalling in recent years. Conditional mouse mutants have revealed the functional significance of neuronal and glial FGF receptors, not only in tissue protection, axon regeneration, and glial proliferation but also in instant behavioural changes. This review provides a summary of recent findings regarding the role of FGFs and their receptors in the nervous system and in the pathogenesis of major neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Lars Klimaschewski
- Department of Anatomy, Histology and Embryology, Institute of Neuroanatomy, Medical University of Innsbruck, Innsbruck, Austria.
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
8
|
Łoś K, Waszkiewicz N. Biological Markers in Anxiety Disorders. J Clin Med 2021; 10:1744. [PMID: 33920547 PMCID: PMC8073190 DOI: 10.3390/jcm10081744] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
Anxiety disorders are one of the most commonly reported disorders in psychiatry, causing a high medical and socio-economic burden. Recently, there has been a soaring interest in the biological basis of anxiety disorders, which is reflected in an increasing number of articles related to the topic. Due to the ambiguity of the diagnosis and a large number of underdiagnosed patients, researchers are looking for laboratory tests that could facilitate the diagnosis of anxiety disorders in clinical practice and would allow for the earliest possible implementation of appropriate treatment. Such potential biomarkers may also be useable in monitoring the efficacy of pharmacological therapy for anxiety disorders. Therefore this article reviews the literature of potential biomarkers such as components of saliva, peripheral blood, cerebrospinal fluid (CSF), and neuroimaging studies. There are promising publications in the literature that can be useful. The most valuable and promising markers of saliva are cortisol, lysozyme, and α-amylase (sAA). In the blood, in turn, we can distinguish serotonin, brain-derived serum neurotrophic factor (BDNF), cortisol, and microRNA. Structural changes in the amygdala and hippocampus are promising neuroimaging markers, while in CSF, potential markers include oxytocin and 5-Hydroxyindoleacetic acid (5-HIAA). Unfortunately, research in the field of biomarkers is hampered by insufficient knowledge about the etiopathogenesis of anxiety disorders, the significant heterogeneity of anxiety disorders, frequent comorbidities, and low specificity of biomarkers. The development of appropriate biomarker panels and their assessment using new approaches may have the prospective to overcome the above-mentioned obstacles.
Collapse
Affiliation(s)
- Kacper Łoś
- Department of Psychiatry, Medical University of Bialystok, Plac Brodowicza 1, 16-070 Choroszcz, Poland;
| | | |
Collapse
|
9
|
Chojnowska S, Ptaszyńska-Sarosiek I, Kępka A, Knaś M, Waszkiewicz N. Salivary Biomarkers of Stress, Anxiety and Depression. J Clin Med 2021; 10:jcm10030517. [PMID: 33535653 PMCID: PMC7867141 DOI: 10.3390/jcm10030517] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 01/06/2023] Open
Abstract
Stress, anxiety and depressive disorders are often characterized by the activation of the stress axis, which results in similar symptoms at some point in these disorders. These disorders are closely related to each other—they occur simultaneously or follow one another. The diagnosis of stress, anxiety and depression is not a perfect procedure currently—it is based on patient observation and an interview with the patient and their family. There are no laboratory tests that would dispel the doubts of the doctor making the diagnosis and allow the appropriate treatment to be implemented as soon as possible. Therefore, this study will review the components of saliva that could be helpful in the quick diagnosis of stress, anxiety and/or depression. Such potential salivary biomarkers could also be useful in monitoring the effectiveness of pharmacological treatment prescribed by a psychiatrist. The following are promising salivary biomarkers of stress, anxiety or depression: cortisol, immunoglobulin A (sIgA), lysozyme, melatonin, α-amylase (sAA), chromogranin A (CgA) and fibroblast growth factor 2 (FGF-2). To the best valuable potential salivary markers of stress, we can include cortisol, lysozyme, sAA and CgA. To differentiate depression from stress, salivary cortisol and melatonin can be helpful. Fluctuations in the concentrations of the above-mentioned substances in saliva indicate a particularly strong relationship with typical human psychological problems, such as stress, depression or anxiety.
Collapse
Affiliation(s)
- Sylwia Chojnowska
- Faculty of Health Sciences, Lomza State University of Applied Sciences, Akademicka Street 14, 18-400 Lomza, Poland;
- Correspondence:
| | | | - Alina Kępka
- Department of Biochemistry, Radioimmunology and Experimental Medicine, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Małgorzata Knaś
- Faculty of Health Sciences, Lomza State University of Applied Sciences, Akademicka Street 14, 18-400 Lomza, Poland;
| | - Napoleon Waszkiewicz
- Department of Psychiatry, Medical University of Bialystok, 16-070 Choroszcz, Poland;
| |
Collapse
|
10
|
Liran M, Rahamim N, Ron D, Barak S. Growth Factors and Alcohol Use Disorder. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a039271. [PMID: 31964648 DOI: 10.1101/cshperspect.a039271] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neurotrophic growth factors were originally characterized for their support in neuronal differentiation, outgrowth, and survival during development. However, it has been acknowledged that they also play a vital role in the adult brain. Abnormalities in growth factors have been implicated in a variety of neurological and psychiatric disorders, including alcohol use disorder (AUD). This work focuses on the interaction between alcohol and growth factors. We review literature suggesting that several growth factors play a unique role in the regulation of alcohol consumption, and that breakdown in these growth factor systems is linked to the development of AUD. Specifically, we focus on the brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), and insulin growth factor 1 (IGF-1). We also review the literature on the potential role of midkine (MDK) and pleiotrophin (PTN) and their receptor, anaplastic lymphoma kinase (ALK), in AUD. We show that alcohol alters the expression of these growth factors or their receptors in brain regions previously implicated in addiction, and that manipulations on these growth factors and their downstream signaling can affect alcohol-drinking behaviors in animal models. We conclude that there is a need for translational and clinical research to assess the therapeutic potential of new pharmacotherapies targeting these systems.
Collapse
Affiliation(s)
- Mirit Liran
- Department of Neurobiology, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Nofar Rahamim
- Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Dorit Ron
- Department of Neurology, University of California, 675 Nelson Rising Lane, San Francisco, California 94143-0663, USA
| | - Segev Barak
- Department of Neurobiology, Tel Aviv University, 69978 Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.,School of Psychological Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| |
Collapse
|
11
|
Polysialylation and disease. Mol Aspects Med 2020; 79:100892. [PMID: 32863045 DOI: 10.1016/j.mam.2020.100892] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/31/2022]
Abstract
Polysialic acid (polySia, PSA) is a unique constituent of the glycocalyx on the surface of bacterial and vertebrate cells. In vertebrates, its biosynthesis is highly regulated, not only in quantity and quality, but also in time and location, which allows polySia to be involved in various important biological phenomena. Therefore, impairments in the expression and structure of polySia sometimes relate to diseases, such as schizophrenia, bipolar disorder, and cancer. Some bacteria express polySia as a tool for protecting themselves from the host immune system during invasion. PolySia is proven to be a biosafe material; polySia, as well as polySia-recognizing molecules, are key therapeutic agents. This review first comprehensive outlines the occurrence, features, biosynthesis, and functions of polySia and subsequently focuses on the related diseases.
Collapse
|
12
|
Persistence of Fear Memory Depends on a Delayed Elevation of BAF53b and FGF1 Expression in the Lateral Amygdala. J Neurosci 2020; 40:7133-7141. [PMID: 32817243 DOI: 10.1523/jneurosci.0679-20.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/24/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022] Open
Abstract
Endurance represents a highly adaptive function of fear memory and a major cause of maladaptive fear- and anxiety-related mental disorders. However, less is known about the mechanisms underlying the persistence of fear memory. The epigenetic gene regulation recently emerged as an important mechanism for memory persistence. In the previous study, we found that BAF53b, a neuron-specific subunit of BAF chromatin remodeling complex, is induced after auditory cued fear conditioning in the lateral amygdala (LA) and is crucial for recent fear memory formation. In this study using mice of both sexes, we report a delayed induction of BAF53b in the LA 48 h after auditory fear conditioning and its critical role for the persistence of established fear memory. To specifically block the delayed but not the early induced BAF53b function, we used a postlearning knock-down method based on RNAi. The transient knockdown of Baf53b using siRNA in the lateral amygdala 24 h after cued fear conditioning led to specific impairment of remote but not recent memory retrieval. RNA-sequencing analyses identified fibroblast growth factor 1 (FGF1) as a candidate downstream effector. Consistently, postlearning administration of FGF1 peptide rescued memory persistence in Baf53b knock-down mice. These results demonstrate the crucial role of BAF53b and FGF1 in persistent retention of fear memory, giving insights into how fear memory persistently stored through consolidation processes and suggest candidate target for treating mental disorders related to traumatic memory.SIGNIFICANCE STATEMENT It is still unclear how once consolidated memory persists over time. In this study, we report the delayed induction of nucleosome remodeling factor BAF53b in the lateral nucleus of amygdala after fear learning and its crucial role for persistence of established memory beyond 24 h after learning. Our data link the regulation of BAF53b and fibroblast growth factor 1 expression in the amygdala to fear memory persistence. Results from this study open a new direction to understand the time-dependent continuous consolidation processes potentially by a nucleosome-remodeling mechanism enabling long-lasting memory formation and give insights into how to treat mental disorders caused by enduring traumatic memory.
Collapse
|
13
|
Humer E, Pieh C, Probst T. Metabolomic Biomarkers in Anxiety Disorders. Int J Mol Sci 2020; 21:E4784. [PMID: 32640734 PMCID: PMC7369790 DOI: 10.3390/ijms21134784] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/04/2020] [Accepted: 07/05/2020] [Indexed: 12/24/2022] Open
Abstract
Anxiety disorders range among the most prevalent psychiatric disorders and belong to the leading disorders in the study of the total global burden of disease. Anxiety disorders are complex conditions, with not fully understood etiological mechanisms. Numerous factors, including psychological, genetic, biological, and chemical factors, are thought to be involved in their etiology. Although the diagnosis of anxiety disorders is constantly evolving, diagnostic manuals rely on symptom lists, not on objective biomarkers and treatment effects are small to moderate. The underlying biological factors that drive anxiety disorders may be better suited to serve as biomarkers for guiding personalized medicine, as they are objective and can be measured externally. Therefore, the incorporation of novel biomarkers into current clinical methods might help to generate a classification system for anxiety disorders that can be linked to the underlying dysfunctional pathways. The study of metabolites (metabolomics) in a large-scale manner shows potential for disease diagnosis, for stratification of patients in a heterogeneous patient population, for monitoring therapeutic efficacy and disease progression, and for defining therapeutic targets. All of these are important properties for anxiety disorders, which is a multifactorial condition not involving a single-gene mutation. This review summarizes recent investigations on metabolomics studies in anxiety disorders.
Collapse
Affiliation(s)
- Elke Humer
- Department for Psychotherapy and Biopsychosocial Health, Danube University Krems, 3500 Krems, Austria; (C.P.); (T.P.)
| | | | | |
Collapse
|
14
|
Rivell A, Mattson MP. Intergenerational Metabolic Syndrome and Neuronal Network Hyperexcitability in Autism. Trends Neurosci 2019; 42:709-726. [PMID: 31495451 PMCID: PMC6779523 DOI: 10.1016/j.tins.2019.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/17/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022]
Abstract
We review evidence that suggests a role for excessive consumption of energy-dense foods, particularly fructose, and consequent obesity and insulin resistance (metabolic syndrome) in the recent increase in prevalence of autism spectrum disorders (ASD). Maternal insulin resistance, obesity, and diabetes may predispose offspring to ASD by mechanisms involving chronic activation of anabolic cellular pathways and a lack of metabolic switching to ketosis resulting in a deficit in GABAergic signaling and neuronal network hyperexcitability. Metabolic reprogramming by epigenetic DNA and chromatin modifications may contribute to alterations in gene expression that result in ASD. These mechanistic insights suggest that interventions that improve metabolic health such as intermittent fasting and exercise may ameliorate developmental neuronal network abnormalities and consequent behavioral manifestations in ASD.
Collapse
Affiliation(s)
- Aileen Rivell
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Graham BM, Richardson R. Fibroblast growth factor-2 enhancement of extinction recall depends on the success of within-session extinction training in rats: a re-analysis. Psychopharmacology (Berl) 2019; 236:227-238. [PMID: 30215215 DOI: 10.1007/s00213-018-5032-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/05/2018] [Indexed: 01/12/2023]
Abstract
RATIONALE One approach to improving exposure therapy for anxiety disorders has focused on developing pharmacological adjuncts to enhance extinction, but these efforts have produced modest success in clinical trials. Understanding the factors that predict the efficacy of adjuncts will help to develop personalized treatments for anxiety. OBJECTIVES We assessed whether individual differences in within-session extinction (fear reduction during extinction training) predict the extent to which the neurotrophin fibroblast growth factor-2 (FGF2) enhances extinction recall in rats. METHODS We re-analyzed data from five experiments that involved administering FGF2 immediately after extinction training; extinction recall was assessed the following day. RESULTS Regression analyses revealed that fear responses at the end, but not the start, of extinction training predicted extinction recall in FGF2- but not vehicle-treated rats. Comparisons between FGF2- and vehicle-treated rats that exhibited better or worse extinction recall (determined by a median split in freezing during extinction recall) confirmed that FGF2-treated rats exhibiting better extinction recall had significantly lower freezing at the end of extinction training relative to FGF2-treated rats exhibiting poorer extinction recall. In contrast, vehicle-treated rats did not differ in within-session extinction based on their performance at extinction recall. Finally, even when classified as having poorer extinction recall, FGF2-treated rats had stronger extinction recall than vehicle-treated rats. CONCLUSIONS These results suggest that FGF2 may be most effective amongst rats that exhibit the lowest fear responses at the end of extinction training. Furthermore, FGF2 does not appear to exacerbate fear in rats that exhibit minimal fear reduction during extinction training.
Collapse
Affiliation(s)
- Bronwyn M Graham
- School of Psychology, The University of New South Wales, Sydney, 2052, Australia.
| | - Rick Richardson
- School of Psychology, The University of New South Wales, Sydney, 2052, Australia
| |
Collapse
|
16
|
Abstract
Sialic acid (Sia) is involved in many biological activities and commonly occurs as a monosialyl residue at the nonreducing terminal end of glycoconjugates. The loss of activity of UDP-GlcNAc2-epimerase/ManNAc kinase, which is a key enzyme in Sia biosynthesis, is lethal to the embryo, which clearly indicates the importance of Sia in embryogenesis. Occasionally, oligo/polymeric Sia structures such as disialic acid (diSia), oligosialic acid (oligoSia), and polysialic acid (polySia) occur in glycoconjugates. In particular, polySia, a well-known epitope that commonly occurs in neuroinvasive bacteria and vertebrate brains, is one of the most well-known and biologically/neurologically important glycotopes in vertebrates. The biological effects of polySia, especially on neural cell-adhesion molecules, have been well studied, and in-depth knowledge regarding polySia has been accumulated. In addition, the importance of diSia and oligoSia epitopes has been reported. In this chapter, the recent advances in the study of diSia, oligoSia, and polySia residues in glycoproteins in neurology, and their history, definition, occurrence, analytical methods, biosynthesis, and biological functions evaluated by phenotypes of gene-targeted mice, biochemical features, and related diseases are described.
Collapse
|
17
|
Even-Chen O, Barak S. The role of fibroblast growth factor 2 in drug addiction. Eur J Neurosci 2018; 50:2552-2561. [PMID: 30144335 DOI: 10.1111/ejn.14133] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/08/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022]
Abstract
Fibroblast growth factor 2 (FGF2) is a member of the FGF-family, which consists of 22 members, with four known FGF receptors (five in humans). Over the last 30 years, FGF2 has been extensively studied for its role in cell proliferation, differentiation, growth, survival and angiogenesis during development, as well as for its role in adult neurogenesis and regenerative plasticity. Over the past decade, FGF2 has been implicated in learning and memory, as well as in several neuropsychiatric disorders, including anxiety, stress, depression and drug addiction. In this review, we present accumulating evidence indicating the involvement of FGF2 in neuroadaptations caused by drugs of abuse, namely, amphetamine, cocaine, nicotine and alcohol. Moreover, evidence suggests that FGF2 is a positive regulator of alcohol and drug-related behaviors. Thus, although additional studies are yet required, we suggest that reducing FGF2 activity may provide a novel therapeutic approach for substance use disorders.
Collapse
Affiliation(s)
- Oren Even-Chen
- School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, 69978, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
18
|
McCullough KM, Daskalakis NP, Gafford G, Morrison FG, Ressler KJ. Cell-type-specific interrogation of CeA Drd2 neurons to identify targets for pharmacological modulation of fear extinction. Transl Psychiatry 2018; 8:164. [PMID: 30135420 PMCID: PMC6105686 DOI: 10.1038/s41398-018-0190-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/23/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022] Open
Abstract
Behavioral and molecular characterization of cell-type-specific populations governing fear learning and behavior is a promising avenue for the rational identification of potential therapeutics for fear-related disorders. Examining cell-type-specific changes in neuronal translation following fear learning allows for targeted pharmacological intervention during fear extinction learning, mirroring possible treatment strategies in humans. Here we identify the central amygdala (CeA) Drd2-expressing population as a novel fear-supporting neuronal population that is molecularly distinct from other, previously identified, fear-supporting CeA populations. Sequencing of actively translating transcripts of Drd2 neurons using translating ribosome affinity purification (TRAP) technology identifies mRNAs that are differentially regulated following fear learning. Differentially expressed transcripts with potentially targetable gene products include Npy5r, Rxrg, Adora2a, Sst5r, Fgf3, Erbb4, Fkbp14, Dlk1, and Ssh3. Direct pharmacological manipulation of NPY5R, RXR, and ADORA2A confirms the importance of this cell population and these cell-type-specific receptors in fear behavior. Furthermore, these findings validate the use of functionally identified specific cell populations to predict novel pharmacological targets for the modulation of emotional learning.
Collapse
Affiliation(s)
- Kenneth M McCullough
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, and Behavioral Sciences, Behavioral Neuroscience, Emory University, Atlanta, GA, USA
| | - Nikolaos P Daskalakis
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Georgette Gafford
- Department of Psychiatry, and Behavioral Sciences, Behavioral Neuroscience, Emory University, Atlanta, GA, USA
| | - Filomene G Morrison
- Department of Psychiatry, and Behavioral Sciences, Behavioral Neuroscience, Emory University, Atlanta, GA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Behavioral Science Division, National Center for PTSD, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Kerry J Ressler
- Division of Depression and Anxiety Disorders, McLean Hospital, Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, and Behavioral Sciences, Behavioral Neuroscience, Emory University, Atlanta, GA, USA.
| |
Collapse
|
19
|
Mattson MP, Moehl K, Ghena N, Schmaedick M, Cheng A. Intermittent metabolic switching, neuroplasticity and brain health. Nat Rev Neurosci 2018; 19:63-80. [PMID: 29321682 PMCID: PMC5913738 DOI: 10.1038/nrn.2017.156] [Citation(s) in RCA: 314] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During evolution, individuals whose brains and bodies functioned well in a fasted state were successful in acquiring food, enabling their survival and reproduction. With fasting and extended exercise, liver glycogen stores are depleted and ketones are produced from adipose-cell-derived fatty acids. This metabolic switch in cellular fuel source is accompanied by cellular and molecular adaptations of neural networks in the brain that enhance their functionality and bolster their resistance to stress, injury and disease. Here, we consider how intermittent metabolic switching, repeating cycles of a metabolic challenge that induces ketosis (fasting and/or exercise) followed by a recovery period (eating, resting and sleeping), may optimize brain function and resilience throughout the lifespan, with a focus on the neuronal circuits involved in cognition and mood. Such metabolic switching impacts multiple signalling pathways that promote neuroplasticity and resistance of the brain to injury and disease.
Collapse
Affiliation(s)
- Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Keelin Moehl
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224, USA
| | - Nathaniel Ghena
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224, USA
| | - Maggie Schmaedick
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224, USA
| | - Aiwu Cheng
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224, USA
| |
Collapse
|
20
|
Graham BM. Fibroblast Growth Factor-2: A Promising Biomarker for Anxiety and Trauma Disorders. J Exp Neurosci 2017; 11:1179069517749589. [PMID: 29308016 PMCID: PMC5751900 DOI: 10.1177/1179069517749589] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 01/28/2023] Open
Abstract
Anxiety and trauma disorders are a significant source of global burden. Although it is clear that there is great heterogeneity in humans' response to trauma and stress, most research on fear and anxiety has focused on the "average" animal. Increased understanding of the sources of individual differences in fear reactions may lead to more refined means of predicting who is at risk for the development of anxiety disorders so that early preventative interventions can be implemented. This commentary highlights recent cross-species work (in rats and humans) indicating that the neurotrophin fibroblast growth factor-2 (FGF2) holds promise as a potential biomarker for anxiety disorder vulnerability. Both central (hippocampal) and peripheral (serum and saliva) markers of FGF2 correlate negatively with fear expression after an aversive conditioning experience. Here, 2 broad accounts of the potential mechanism of vulnerability captured by measures of FGF2 are outlined. In particular, it is suggested that basal differences in FGF2 (across different tissue types) may provide a general index of one's regenerative capacity; alternatively, differences in FGF2 reactivity (in specific tissue types) may be indicative of one's coping capacity in response to stress.
Collapse
Affiliation(s)
- Bronwyn M Graham
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
21
|
Prater KE. Individual Differences Make a Marker in Fear Research. Biol Psychiatry 2017; 82:e65-e66. [PMID: 28935099 PMCID: PMC5972024 DOI: 10.1016/j.biopsych.2017.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 07/25/2017] [Indexed: 11/19/2022]
|
22
|
Prater KE, Aurbach EL, Larcinese HK, Smith TN, Turner CA, Blandino P, Watson SJ, Maren S, Akil H. Selectively Bred Rats Provide a Unique Model of Vulnerability to PTSD-Like Behavior and Respond Differentially to FGF2 Augmentation Early in Life. Neuropsychopharmacology 2017; 42:1706-1714. [PMID: 28205604 PMCID: PMC5518903 DOI: 10.1038/npp.2017.37] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 02/09/2017] [Accepted: 02/11/2017] [Indexed: 11/10/2022]
Abstract
Individuals respond differently to traumatic experiences, including their propensity to develop posttraumatic stress disorder (PTSD). Understanding individual differences in PTSD vulnerability will allow the development of improved prevention and treatment options. Here we characterized fear conditioning and extinction in rats selectively bred for differences in their locomotor response to a novel environment. Selectively bred high-responder (bHR) and low-responder (bLR) male rats are known to differ in their emotional reactivity on a range of measures of spontaneous anxiety- and depressive-like behaviors. We demonstrate that bHRs have facilitated extinction learning and retention compared with outbred Sprague Dawley rats, whereas bLRs show reduced extinction learning and retention. This indicates that bLRs are more vulnerable to PTSD-like behavior. Fibroblast growth factor 2 (FGF2) has previously been implicated in the development of these behavioral phenotypes and facilitates extinction learning in outbred animals, therefore we examined the effects of early-life FGF2 on bHR and bLR behavior. FGF2 administered on the day after birth facilitated extinction learning and retention in bHRs, but not in bLRs or control rats, during adulthood. This indicates that, under the current fear conditioning paradigm, early-life FGF2 has protective effects only in resilient animals. This stands in contrast to FGF2's ability to protect vulnerable animals in milder tests of anxiety. These results provide a unique animal model of individual differences in PTSD-like behavior, allowing the study of genetic, developmental, and environmental factors in its expression.
Collapse
Affiliation(s)
- Katherine E Prater
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA,Molecular and Behavioral Neuroscience Institute, University of Michigan, 2008 MBNI Building, 205 Zina Pitcher Place, Ann Arbor, MI 48109-5720, USA, Tel: 734 764 6999, Fax: 734 647 4130, E-mail:
| | - Elyse L Aurbach
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Hanna K Larcinese
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Taylor N Smith
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Cortney A Turner
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Peter Blandino
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Stanley J Watson
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Stephen Maren
- Department of Psychology, Texas A&M University, College Station TX, USA
| | - Huda Akil
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Harmatz ES, Stone L, Lim SH, Lee G, McGrath A, Gisabella B, Peng X, Kosoy E, Yao J, Liu E, Machado NJ, Weiner VS, Slocum W, Cunha RA, Goosens KA. Central Ghrelin Resistance Permits the Overconsolidation of Fear Memory. Biol Psychiatry 2017; 81:1003-1013. [PMID: 28010876 PMCID: PMC5447505 DOI: 10.1016/j.biopsych.2016.11.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 10/25/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND There are many contradictory findings about the role of the hormone ghrelin in aversive processing, with studies suggesting that ghrelin signaling can both inhibit and enhance aversion. Here, we characterize and reconcile the paradoxical role of ghrelin in the acquisition of fearful memories. METHODS We used enzyme-linked immunosorbent assay to measure endogenous acyl-ghrelin and corticosterone at time points surrounding auditory fear learning. We used pharmacological (systemic and intra-amygdala) manipulations of ghrelin signaling and examined several aversive and appetitive behaviors. We also used biotin-labeled ghrelin to visualize ghrelin binding sites in coronal brain sections of amygdala. All work was performed in rats. RESULTS In unstressed rodents, endogenous peripheral acyl-ghrelin robustly inhibits fear memory consolidation through actions in the amygdala and accounts for virtually all interindividual variability in long-term fear memory strength. Higher levels of endogenous ghrelin after fear learning were associated with weaker long-term fear memories, and pharmacological agonism of the ghrelin receptor during the memory consolidation period reduced fear memory strength. These fear-inhibitory effects cannot be explained by changes in appetitive behavior. In contrast, we show that chronic stress, which increases both circulating endogenous acyl-ghrelin and fear memory formation, promotes profound loss of ghrelin binding sites in the amygdala and behavioral insensitivity to ghrelin receptor agonism. CONCLUSIONS These studies provide a new link between stress, a novel type of metabolic resistance, and vulnerability to excessive fear memory formation and reveal that ghrelin can regulate negative emotionality in unstressed animals without altering appetite.
Collapse
Affiliation(s)
- Elia S Harmatz
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Lauren Stone
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Seh Hong Lim
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge; Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts
| | - Graham Lee
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Anna McGrath
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Barbara Gisabella
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Xiaoyu Peng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Eliza Kosoy
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Junmei Yao
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Elizabeth Liu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Nuno J Machado
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Veronica S Weiner
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Warren Slocum
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge
| | - Rodrigo A Cunha
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ki A Goosens
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge.
| |
Collapse
|
24
|
King G, Scott E, Graham BM, Richardson R. Individual differences in fear extinction and anxiety-like behavior. ACTA ACUST UNITED AC 2017; 24:182-190. [PMID: 28416629 PMCID: PMC5397683 DOI: 10.1101/lm.045021.117] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/03/2017] [Indexed: 11/25/2022]
Abstract
There is growing appreciation for the substantial individual differences in the acquisition and inhibition of aversive associations, and the insights this might give into identifying individuals particularly vulnerable to stress and psychopathology. We examined whether animals that differed in rate of extinction (i.e., Fast versus Slow) were different in their response to an acute stress in adulthood or following a chronic stress that occurred either early or later in life. We found that Slow Extinguishers had significantly poorer extinction retention than Fast Extinguishers, but an acute stressor did not differentially affect anxiety-like behavior in the two groups. Further, while exposure to chronic stress in adulthood did not impact on the extinction phenotypes or anxiety-like behavior, exposure to chronic stress early in life affected both extinction retention and anxiety-like behavior. These findings have implications for the development of a more nuanced approach to identifying those most at risk of anxiety disorders.
Collapse
Affiliation(s)
- Gabrielle King
- School of Psychology, The University of New South Wales, UNSW, Sydney, 2052, Australia
| | - Elliot Scott
- School of Psychology, The University of New South Wales, UNSW, Sydney, 2052, Australia
| | - Bronwyn M Graham
- School of Psychology, The University of New South Wales, UNSW, Sydney, 2052, Australia
| | - Rick Richardson
- School of Psychology, The University of New South Wales, UNSW, Sydney, 2052, Australia
| |
Collapse
|
25
|
Zhang T, Zhou S, Hu L, Peng B, Liu Y, Luo X, Song Y, Liu X, Deng Y. Polysialic acid-modifying liposomes for efficient delivery of epirubicin, in-vitro characterization and in-vivo evaluation. Int J Pharm 2016; 515:449-459. [DOI: 10.1016/j.ijpharm.2016.10.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 10/06/2016] [Accepted: 10/23/2016] [Indexed: 12/22/2022]
|
26
|
Van ND, Falk CS, Sandmann L, Vondran FWR, Helfritz F, Wedemeyer H, Manns MP, Ciesek S, von Hahn T. Modulation of HCV reinfection after orthotopic liver transplantation by fibroblast growth factor-2 and other non-interferon mediators. Gut 2016; 65:1015-23. [PMID: 25800783 PMCID: PMC4893125 DOI: 10.1136/gutjnl-2014-308003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 02/12/2015] [Indexed: 01/11/2023]
Abstract
OBJECTIVE In HCV infected individuals graft infection occurs shortly after orthotopic liver transplantation (OLT). We aimed to describe the composition of the inflammatory response at this time, how it affects the HCV replication cycle and identify novel proviral and antiviral factors. DESIGN We used a Luminex assay to quantify 50 inflammatory mediators in sera before and shortly after OLT. In vitro grown HCV based on the JFH-1 isolate were used to characterise the effects of patient sera and individual mediators on HCV. RESULTS Although the mediator composition is highly variable between individuals, sera drawn immediately post-OLT significantly enhance HCV infectivity compared with control sera from before OLT in about half of the cases. Among 27 non-interferon inflammatory mediators fibroblast growth factor (FGF)-2 stood out as it enhanced HCV RNA replication and release of infectious particles. The effect was concentration-dependent and detectable in dividing and non-dividing cells. Moreover, pharmacological inhibition of FGF-2 receptor signalling abrogated the enhancing effect of FGF-2 and inhibited HCV replication in the absence of serum FGF-2 suggesting that HCV replication is dependent on basal activation of the FGF-2 triggered signalling pathway. Finally, in individuals with chronic HCV infection with high viral load, serum FGF-2 was significantly higher compared with those with low viral load. CONCLUSIONS Although no single mediator may account for this effect, serum shortly post-OLT enhances HCV infection. FGF-2 is a novel endogenous driver of HCV replication and a potential therapeutic target.
Collapse
Affiliation(s)
- Nguyen Dinh Van
- Department of Gastroenterology, Hepatology and Endocrinology, Medizinische Hochschule Hannover, Hannover, Germany,German Center for Infection Research (DZIF), Hannover, Germany,Institute for Molecular Biology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Christine S Falk
- Institute of Transplant Immunology, IFB-Tx, Medizinische Hochschule Hannover, Hannover, Germany
| | - Lisa Sandmann
- Department of Gastroenterology, Hepatology and Endocrinology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Florian W R Vondran
- German Center for Infection Research (DZIF), Hannover, Germany,Department of General, Visceral and Transplantation Surgery, Medizinische Hochschule Hannover, Hannover, Germany
| | - Fabian Helfritz
- German Center for Infection Research (DZIF), Hannover, Germany,Department of General, Visceral and Transplantation Surgery, Medizinische Hochschule Hannover, Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Medizinische Hochschule Hannover, Hannover, Germany,German Center for Infection Research (DZIF), Hannover, Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Medizinische Hochschule Hannover, Hannover, Germany,German Center for Infection Research (DZIF), Hannover, Germany
| | - Sandra Ciesek
- Department of Gastroenterology, Hepatology and Endocrinology, Medizinische Hochschule Hannover, Hannover, Germany,German Center for Infection Research (DZIF), Hannover, Germany
| | - Thomas von Hahn
- Department of Gastroenterology, Hepatology and Endocrinology, Medizinische Hochschule Hannover, Hannover, Germany,German Center for Infection Research (DZIF), Hannover, Germany,Institute for Molecular Biology, Medizinische Hochschule Hannover, Hannover, Germany
| |
Collapse
|
27
|
Veniaminova EA, Zubareva OE. The changes in exploratory behavior and Fgf2 gene expression in cells of the rat brain after the early postnatal administration of bacterial lipopolysaccharide. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415020105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Singewald N, Schmuckermair C, Whittle N, Holmes A, Ressler KJ. Pharmacology of cognitive enhancers for exposure-based therapy of fear, anxiety and trauma-related disorders. Pharmacol Ther 2014; 149:150-90. [PMID: 25550231 PMCID: PMC4380664 DOI: 10.1016/j.pharmthera.2014.12.004] [Citation(s) in RCA: 275] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 12/24/2014] [Indexed: 12/20/2022]
Abstract
Pathological fear and anxiety are highly debilitating and, despite considerable advances in psychotherapy and pharmacotherapy they remain insufficiently treated in many patients with PTSD, phobias, panic and other anxiety disorders. Increasing preclinical and clinical evidence indicates that pharmacological treatments including cognitive enhancers, when given as adjuncts to psychotherapeutic approaches [cognitive behavioral therapy including extinction-based exposure therapy] enhance treatment efficacy, while using anxiolytics such as benzodiazepines as adjuncts can undermine long-term treatment success. The purpose of this review is to outline the literature showing how pharmacological interventions targeting neurotransmitter systems including serotonin, dopamine, noradrenaline, histamine, glutamate, GABA, cannabinoids, neuropeptides (oxytocin, neuropeptides Y and S, opioids) and other targets (neurotrophins BDNF and FGF2, glucocorticoids, L-type-calcium channels, epigenetic modifications) as well as their downstream signaling pathways, can augment fear extinction and strengthen extinction memory persistently in preclinical models. Particularly promising approaches are discussed in regard to their effects on specific aspects of fear extinction namely, acquisition, consolidation and retrieval, including long-term protection from return of fear (relapse) phenomena like spontaneous recovery, reinstatement and renewal of fear. We also highlight the promising translational value of the preclinial research and the clinical potential of targeting certain neurochemical systems with, for example d-cycloserine, yohimbine, cortisol, and L-DOPA. The current body of research reveals important new insights into the neurobiology and neurochemistry of fear extinction and holds significant promise for pharmacologically-augmented psychotherapy as an improved approach to treat trauma and anxiety-related disorders in a more efficient and persistent way promoting enhanced symptom remission and recovery.
Collapse
Affiliation(s)
- N Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria.
| | - C Schmuckermair
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - N Whittle
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - A Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - K J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
29
|
Colley KJ, Kitajima K, Sato C. Polysialic acid: biosynthesis, novel functions and applications. Crit Rev Biochem Mol Biol 2014; 49:498-532. [PMID: 25373518 DOI: 10.3109/10409238.2014.976606] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As an anti-adhesive, a reservoir for key biological molecules, and a modulator of signaling, polysialic acid (polySia) is critical for nervous system development and maintenance, promotes cancer metastasis, tissue regeneration and repair, and is implicated in psychiatric diseases. In this review, we focus on the biosynthesis and functions of mammalian polySia, and the use of polySia in therapeutic applications. PolySia modifies a small subset of mammalian glycoproteins, with the neural cell adhesion molecule, NCAM, serving as its major carrier. Studies show that mammalian polysialyltransferases employ a unique recognition mechanism to limit the addition of polySia to a select group of proteins. PolySia has long been considered an anti-adhesive molecule, and its impact on cell adhesion and signaling attributed directly to this property. However, recent studies have shown that polySia specifically binds neurotrophins, growth factors, and neurotransmitters and that this binding depends on chain length. This work highlights the importance of considering polySia quality and quantity, and not simply its presence or absence, as its various roles are explored. The capsular polySia of neuroinvasive bacteria allows these organisms to evade the host immune response. While this "stealth" characteristic has made meningitis vaccine development difficult, it has also made polySia a worthy replacement for polyetheylene glycol in the generation of therapeutic proteins with low immunogenicity and improved circulating half-lives. Bacterial polysialyltransferases are more promiscuous than the protein-specific mammalian enzymes, and new studies suggest that these enzymes have tremendous therapeutic potential, especially for strategies aimed at neural regeneration and tissue repair.
Collapse
Affiliation(s)
- Karen J Colley
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, IL , USA and
| | | | | |
Collapse
|
30
|
Ballaz SJ, Perez J, Waselus M, Akil H, Watson SJ. Interaction between cholecystokinin and the fibroblast growth factor system in the ventral tegmental area of selectively bred high- and low-responder rats. Neuroscience 2013; 255:68-75. [PMID: 24121132 DOI: 10.1016/j.neuroscience.2013.09.063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 11/26/2022]
Abstract
Individual differences in the locomotor response to novelty have been linked to basal differences in dopaminergic neurotransmission. Mesolimbic dopaminergic outputs are regulated by cholecystokinin (CCK), a neuropeptide implicated in anxiety. In turn, CCK expression is regulated by fibroblast growth factor-2 (FGF2), which has recently been identified as an endogenous regulator of anxiety. FGF2 binds to the high-affinity fibroblast growth factor receptor-1 (FGF-R1) to regulate the development and maintenance of dopamine neurons in the ventral tegmental area (VTA). However, the relationship between the FGF and CCK systems in the VTA is not well understood. Therefore, we utilized the selectively-bred low-responder (bLR; high-anxiety) and high-responder (bHR; low-anxiety) rats to examine the effects of repeated (21-day) FGF2 treatment on CCK and FGF-R1 mRNA in the rostral VTA (VTAr). In vehicle-treated controls, both CCK and FGF-R1 mRNA levels were increased in the VTAr of bLR rats relative to bHR rats. Following FGF2 treatment, however, bHR-bLR differences in CCK and FGF-R1 mRNA expression were eliminated, due to decreased CCK mRNA levels in the VTAr of bLR rats and increased FGF-R1 expression in bHR rats. Differences after FGF2 treatment may denote distinct interactions between the CCK and FGF systems in the VTAr of bHR vs. bLR rats. Indeed, significant correlations between CCK and FGF-R1 mRNA expression were found in bHR, but not bLR rats. Colocalization studies suggest that CCK and FGF-R1 are coexpressed in some VTAr neurons. Taken together, our findings suggest that the FGF system is poised to modulate both CCK and FGF-R1 expression in the VTAr, which may be associated with individual differences in mesolimbic pathways associated with anxiety-like behavior.
Collapse
Affiliation(s)
- S J Ballaz
- Molecular and Behavioral Neuroscience Institute, University of Michigan, 205 Zina Pitcher Place, Ann Arbor, MI 48109-0720, USA.
| | | | | | | | | |
Collapse
|
31
|
Kopec AM, Carew TJ. Growth factor signaling and memory formation: temporal and spatial integration of a molecular network. Learn Mem 2013; 20:531-9. [PMID: 24042849 PMCID: PMC3768197 DOI: 10.1101/lm.031377.113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Growth factor (GF) signaling is critically important for developmental plasticity. It also plays a crucial role in adult plasticity, such as that required for memory formation. Although different GFs interact with receptors containing distinct types of kinase domains, they typically signal through converging intracellular cascades (e.g., Ras–MEK–MAPK) to mediate overlapping functional endpoints. Several GFs have been implicated in memory formation, but due to a high level of convergent signaling, the unique contributions of individual GFs as well as the interactions between GF signaling cascades during the induction of memory is not well known. In this review, we highlight the unique roles of specific GFs in dendritic plasticity, and discuss the spatial and temporal profiles of different GFs during memory formation. Collectively, the data suggest that the roles of GF signaling in long-lasting behavioral and structural plasticity may be best viewed as interactive components in a complex molecular network.
Collapse
Affiliation(s)
- Ashley M Kopec
- Center for Neural Science, New York University, New York, New York 10003, USA
| | | |
Collapse
|
32
|
FGF2 blocks PTSD symptoms via an astrocyte-based mechanism. Behav Brain Res 2013; 256:472-80. [PMID: 24013012 DOI: 10.1016/j.bbr.2013.08.048] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 12/20/2022]
Abstract
Although posttraumatic stress disorder (PTSD) is characterized by traumatic memories or experiences and increased arousal, which can be partly alleviated by antidepressants, the underlying cellular mechanisms are not fully understood. As emerging studies have focused on the critical role of astrocytes in pathological mood disorders, we hypothesized that several 'astrocyte-related' mechanisms underlying PTSD exist. In the present study, using the single prolonged stress (SPS) model, we investigated the effects of intraperitoneal FGF2 on SPS-induced PTSD behavior response as well as the astrocytic activation after FGF2 administration in SPS rats. Behavioral data showed that intraperitoneal FGF2 inhibited SPS-induced hyperarousal and anxiety behavior; however, immunohistochemistry showed that SPS-induced astrocytic inhibition was activated by intraperitoneal FGF2. Quantitative western blotting showed that intraperitoneal FGF2 up-regulated glial fibrillary acidic protein (GFAP), but not NeuN, expression in the hippocampus. We suggest that intraperitoneal FGF2 could block the SPS-induced fear response and anxiety behavior in PTSD via astrocyte-based but not neuron-based mechanisms.
Collapse
|
33
|
Callaghan BL, Graham BM, Li S, Richardson R. From resilience to vulnerability: mechanistic insights into the effects of stress on transitions in critical period plasticity. Front Psychiatry 2013; 4:90. [PMID: 23964249 PMCID: PMC3741646 DOI: 10.3389/fpsyt.2013.00090] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/31/2013] [Indexed: 11/24/2022] Open
Abstract
While early experiences are proposed to be important for the emergence of anxiety and other mental health problems, there is little empirical research examining the impact of such experiences on the development of emotional learning. Of the research that has been performed in this area, however, a complex picture has emerged in which the maturation of emotion circuits is influenced by the early experiences of the animal. For example, under typical laboratory rearing conditions infant rats rapidly forget learned fear associations (infantile amnesia) and express a form of extinction learning which is relapse-resistant (i.e., extinction in infant rats may be due to fear erasure). In contrast, adult rats exhibit very long-lasting memories of past learned fear associations, and express a form of extinction learning that is relapse-prone (i.e., the fear returns in a number of situations). However, when rats are reared under stressful conditions then they exhibit adult-like fear retention and extinction behaviors at an earlier stage of development (i.e., good retention of learned fear and relapse-prone extinction learning). In other words, under typical rearing conditions infant rats appear to be protected from exhibiting anxiety whereas after adverse rearing fear learning appears to make those infants more vulnerable to the later development of anxiety. While the effects of different experiences on infant rats' fear retention and extinction are becoming better documented, the mechanisms which mediate the early transition seen following stress remain unclear. Here we suggest that rearing stress may lead to an early maturation of the molecular and cellular signals shown to be involved in the closure of critical period plasticity in sensory modalities (e.g., maturation of GABAergic neurons, development of perineuronal nets), and speculate that these signals could be manipulated in adulthood to reopen infant forms of emotional learning (i.e., those that favor resilience).
Collapse
Affiliation(s)
- Bridget L Callaghan
- School of Psychology, The University of New South Wales , Sydney, NSW , Australia
| | | | | | | |
Collapse
|
34
|
Turner CA, Watson SJ, Akil H. The fibroblast growth factor family: neuromodulation of affective behavior. Neuron 2012; 76:160-74. [PMID: 23040813 DOI: 10.1016/j.neuron.2012.08.037] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2012] [Indexed: 12/20/2022]
Abstract
In this review, we propose a broader view of the role of the fibroblast growth factor (FGF) family in modulating brain function. We suggest that some of the FGF ligands together with the FGF receptors are altered in individuals with affective disorder and modulate emotionality in animal models. Thus, we propose that members of the FGF family may be genetic predisposing factors for anxiety, depression, or substance abuse; that they play a key organizing role during early development but continue to play a central role in neuroplasticity in adulthood; and that they work not only over extended time frames, but also via rapid signaling mechanisms, allowing them to exert an "on-line" influence on behavior. Therefore, the FGF family appears to be a prototype of "switch genes" that are endowed with organizational and modulatory properties across the lifespan, and that may represent molecular candidates as biomarkers and treatment targets for affective and addictive disorders.
Collapse
Affiliation(s)
- Cortney A Turner
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
35
|
Milad MR, Quirk GJ. Fear extinction as a model for translational neuroscience: ten years of progress. Annu Rev Psychol 2012; 63:129-51. [PMID: 22129456 DOI: 10.1146/annurev.psych.121208.131631] [Citation(s) in RCA: 1007] [Impact Index Per Article: 83.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The psychology of extinction has been studied for decades. Approximately 10 years ago, however, there began a concerted effort to understand the neural circuits of extinction of fear conditioning, in both animals and humans. Progress during this period has been facilitated by a high degree of coordination between rodent and human researchers examining fear extinction. Here we review the major advances and highlight new approaches to understanding and exploiting fear extinction. Research in fear extinction could serve as a model for translational research in other areas of behavioral neuroscience.
Collapse
Affiliation(s)
- Mohammed R Milad
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, 02129, USA
| | | |
Collapse
|