1
|
da Fonseca STO, Alves CC, Dias CT, Mendes-da-Silva C. Probiotics and undernourishment impact on brain 5-Hydroxytryptamine system and neurotrophin BDNF in rats: Risk of depression and anxiety? Nutrition 2024; 132:112680. [PMID: 39904121 DOI: 10.1016/j.nut.2024.112680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/14/2024] [Accepted: 12/26/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Undernourishment can significantly affect the serotonergic system, potentially increasing the risk of neuropsychiatric disorders such as depression and anxiety. Probiotic therapy has emerged as a potential modulator of the serotonergic system and brain-derived neurotrophic factor (BDNF). Our hypothesis posits that probiotic treatment positively influences the serotonergic system and BDNF levels in the prefrontal cortex (PFC) and hippocampus (HIP), mitigating the effects of malnutrition. METHODS We conducted an experiment using 38 adult isogenic rats, divided into four groups: nourished control (n = 9), undernourished control (n = 9), nourished probiotic (n = 10), and undernourished probiotic (n = 10). The animals experienced undernourishment for 10 days, followed by probiotic supplementation while continuing food restriction for an additional 15 days. On the 25th day of the experiment, we euthanized the animals, microdissected their brains, and extracted samples from the HIP and PFC. We performed immunoblotting analysis to assess the expression levels of the following proteins: BDNF, tryptophan hydroxylase 2 enzyme, and 5-HT1A and 5-HT2C serotonergic receptors. RESULTS Our findings revealed the following effects of probiotic administration: tryptophan hydroxylase 2 expression increased in the PFC of nourished rats (P = 0.033) and in the HIP of undernourished rats (P = 0.013); improved 5-HT2C expression was observed in the PFC under both nutritional conditions (P < 0.01). The proBDNF levels were elevated in the HIP of undernourished rats (P = 0.001). CONCLUSION Probiotic administration effectively modulated the gut-microbiota-brain axis by enhancing serotonergic system proteins in the prefrontal cortex and hippocampus of both nourished and undernourished rats.
Collapse
Affiliation(s)
| | - Cláudia Cristina Alves
- Department of Biosciences, Clinical and Experimental Nutrition Research Laboratory, Federal University of São Paulo, Santos, São Paulo, Brazil
| | - Clarissa Tavares Dias
- Department of Biosciences, Laboratory of Neuroscience and Nutrition, Federal University of São Paulo, Santos, São Paulo, Brazil
| | - Cristiano Mendes-da-Silva
- Department of Biosciences, Laboratory of Neuroscience and Nutrition, Federal University of São Paulo, Santos, São Paulo, Brazil.
| |
Collapse
|
2
|
Kendricks DR, Morrow C, Haste DA, Newland MC. Adult and adolescent antipsychotic exposure increases delay discounting and diminishes behavioral flexibility in male C57BL/6 mice. Pharmacol Biochem Behav 2024; 245:173866. [PMID: 39241867 DOI: 10.1016/j.pbb.2024.173866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Second-generation antipsychotics are frequently prescribed to adolescents, but the long-term consequences of their use remain understudied. These medications work via monoamine neurotransmitter systems, especially dopamine and serotonin, which undergo considerable development and pruning during adolescence. Dopamine and serotonin are linked to a wide host of behaviors, including impulsive choice and behavioral plasticity. In a murine model of adolescent antipsychotic use, male C57BL/6 mice were exposed to either 2.5 mg/kg/day risperidone or 5 mg/kg/day olanzapine via drinking water from postnatal days 22-60. To determine whether the adolescent period was uniquely sensitive to antipsychotic exposure, long-term effects on behavior were compared to an equivalently exposed group of adults where mice were exposed to 2.5 mg/kg risperidone from postnatal days 101-138. Motor activity and body weight in adolescent animals were assessed. Thirty days after exposure terminated animal's behavioral flexibility and impulsive choice were assessed using spatial discrimination reversal and delay discounting. Antipsychotic exposure produced a modest change in behavior flexibility during the second reversal. There was a robust and reproducible difference in impulsive choice: exposed animals devalued the delayed alternative reward substantially more than controls. This effect was observed both following adolescent and adult exposure, indicating that an irreversible change in impulsive choice occurs regardless of the age of exposure.
Collapse
|
3
|
Cory-Slechta DA, Marvin E, Welle K, Goeke C, Chalupa D, Oberdörster G, Sobolewski M. Male-biased vulnerability of mouse brain tryptophan/kynurenine and glutamate systems to adolescent exposures to concentrated ambient ultrafine particle air pollution. Neurotoxicology 2024; 104:20-35. [PMID: 39002649 PMCID: PMC11377152 DOI: 10.1016/j.neuro.2024.07.004] [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: 04/29/2024] [Revised: 06/27/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Air pollution (AP) exposures have been associated with numerous neurodevelopmental and psychiatric disorders, including autism spectrum disorder, attention deficit hyperactivity disorder and schizophrenia, all male-biased disorders with onsets from early life to late adolescence/early adulthood. While prior experimental studies have focused on effects of AP exposures during early brain development, brain development actually extends well into early adulthood. The current study in mice sought to extend the understanding of developmental brain vulnerability during adolescence, a later but significant period of brain development and maturation to the ultrafine particulate (UFPs) component of AP, considered its most reactive component. Additionally, it examined adolescent response to UFPs when preceded by earlier developmental exposures, to ascertain the trajectory of effects and potential enhancement or mitigation of adverse consequences. Outcomes focused on shared features associated with multiple neurodevelopmental disorders. For this purpose, C57Bl/6 J mice of both sexes were exposed to ambient concentrated UFPs or filtered air from PND (postnatal day) 4-7 and PND10-13, and again at PND39-42 and 45-49, resulting in 3 exposure postnatal/adolescent treatment groups per sex: Air/Air, Air/UFP, and UFP/UFP. Features common to neurodevelopmental disorders were examined at PND50. Mass exposure concentration from postnatal exposure averaged 44.34 μg/m3 and the adolescent exposure averaged 49.18 μg/m3. Male brain showed particular vulnerability to UFP exposures in adolescence, with alterations in frontal cortical and striatal glutamatergic and tryptophan/serotonergic neurotransmitters and concurrent reductions in levels of astrocytes in corpus callosum and in serum cytokine levels, with combined exposures resulting in significant reductions in corpus callosum myelination and serum corticosterone. Reductions in serum corticosterone in males correlated with reductions in neurotransmitter levels, and reductions in striatal glutamatergic function specifically correlated with reductions in corpus callosum astrocytes. UFP-induced changes in neurotransmitter levels in males were mitigated by prior postnatal exposure, suggesting potential adaptation, whereas reductions in corticosterone and in corpus callosum neuropathological effects were further strengthened by combined postnatal and adolescent exposures. UFP-induced changes in females occurred primarily in striatal dopamine systems and as reductions in serum cytokines only in response to combined postnatal and adolescent exposures. Findings in males underscore the importance of more integrated physiological assessments of mechanisms of neurotoxicity. Further, these findings provide biological plausibility for an accumulating epidemiologic literature linking air pollution to neurodevelopmental and psychiatric disorders. As such, they support a need for consideration of the regulation of the UFP component of air pollution.
Collapse
Affiliation(s)
- D A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical School, Box EHSC, Rochester, NY 14642, United States.
| | - E Marvin
- Department of Environmental Medicine, University of Rochester Medical School, Box EHSC, Rochester, NY 14642, United States
| | - K Welle
- Mass Spectrometry Resource Laboratory, University of Rochester Medical School, Box EHSC, Rochester, NY 14642, United States
| | - C Goeke
- Department of Environmental Medicine, University of Rochester Medical School, Box EHSC, Rochester, NY 14642, United States
| | - D Chalupa
- Department of Environmental Medicine, University of Rochester Medical School, Box EHSC, Rochester, NY 14642, United States
| | - G Oberdörster
- Department of Environmental Medicine, University of Rochester Medical School, Box EHSC, Rochester, NY 14642, United States
| | - M Sobolewski
- Department of Environmental Medicine, University of Rochester Medical School, Box EHSC, Rochester, NY 14642, United States
| |
Collapse
|
4
|
Medeiros AC, Medeiros P, Pigatto GR, Maione S, Coimbra NC, de Freitas RL. Cannabidiol in the dorsal hippocampus attenuates emotional and cognitive impairments related to neuropathic pain: The role of prelimbic neocortex-hippocampal connections. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111039. [PMID: 38797491 DOI: 10.1016/j.pnpbp.2024.111039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND AND PURPOSE Chronic neuropathic pain (NP) is commonly associated with cognitive and emotional impairments. Cannabidiol (CBD) presents a broad spectrum of action with a potential analgesic effect. This work investigates the CBD effect on comorbidity between chronic NP, depression, and memory impairment. EXPERIMENTAL APPROACH The connection between the neocortex and the hippocampus was investigated with biotinylated dextran amine (BDA) deposits in the prelimbic cortex (PrL). Wistar rats were submitted to chronic constriction injury (CCI) of the sciatic nerve and CA1 treatment with CBD (15, 30, 60 nmol). KEY RESULTS BDA-labeled perikarya and terminal buttons were found in CA1 and dentate gyrus. CCI-induced mechanical and cold allodynia increased c-Fos protein expression in the PrL and CA1. The number of astrocytes in PrL and CA1 increased, and the number of neuroblasts decreased in CA1. Animals submitted to CCI procedure showed increasing depressive-like behaviors, such as memory impairment. CBD (60 nmol) treatment decreased mechanical and cold allodynia, attenuated depressive-associated behaviors, and improved memory performance. Cobalt chloride (CoCl2: 1 nM), WAY-100635 (0.37 nmol), and AM251 (100 nmol) intra-PrL reversed the effect of CA1 treatment with CBD (60 nmol) on nociceptive, cognitive, and depressive behaviors. CONCLUSION CBD represents a promising therapeutic perspective in the pharmacological treatment of chronic NP and associated comorbidities such as depression and memory impairments. The CBD effects possibly recruit the CA1-PrL pathway, inducing neuroplasticity. CBD acute treatment into the CA1 produces functional and molecular morphological improvements.
Collapse
Affiliation(s)
- Ana Carolina Medeiros
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Laboratory of Neurosciences of Pain & Emotions, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Ribeirão Preto, SP 14050-220, Brazil
| | - Priscila Medeiros
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Laboratory of Neurosciences of Pain & Emotions, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Department of General and Specialized Nursing, Ribeirão Preto Nursing School of the University of São Paulo (EERP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Glauce Regina Pigatto
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Norberto Cysne Coimbra
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Ribeirão Preto, SP 14050-220, Brazil
| | - Renato Leonardo de Freitas
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Laboratory of Neurosciences of Pain & Emotions, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Ribeirão Preto, SP 14050-220, Brazil; Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy.
| |
Collapse
|
5
|
Jiang J, Tan S, Feng X, Peng Y, Long C, Yang L. Distinct ACC Neural Mechanisms Underlie Authentic and Transmitted Anxiety Induced by Maternal Separation in Mice. J Neurosci 2023; 43:8201-8218. [PMID: 37845036 PMCID: PMC10697407 DOI: 10.1523/jneurosci.0558-23.2023] [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: 03/27/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023] Open
Abstract
It is known that humans and rodents are capable of transmitting stress to their naive partners via social interaction. However, a comprehensive understanding of transmitted stress, which may differ from authentic stress, thus revealing unique neural mechanisms of social interaction resulting from transmitted stress and the associated anxiety, is missing. We used, in the present study, maternal separation (MS) as a stress model to investigate whether MS causes abnormal behavior in adolescence. A key concern in the analysis of stress transmission is whether the littermates of MS mice who only witness MS stress ("Partners") exhibit behavioral abnormalities similar to those of MS mice themselves. Of special interest is the establishment of the neural mechanisms underlying transmitted stress and authentic stress. The results show that Partners, similar to MS mice, exhibit anxiety-like behavior and hyperalgesia after witnessing littermates being subjected to early-life repetitive MS. Electrophysiological analysis revealed that mice subjected to MS demonstrate a reduction in both the excitatory and inhibitory synaptic activities of parvalbumin interneurons (PVINs) in the anterior cingulate cortex (ACC). However, Partners differed from MS mice in showing an increase in the number and excitability of GABAergic PVINs in the ACC and in the ability of chemogenetic PVIN inactivation to eliminate abnormal behavior. Furthermore, the social transfer of anxiety-like behavior required intact olfactory, but not visual, perception. This study suggests a functional involvement of ACC PVINs in mediating the distinct neural basis of transmitted anxiety.SIGNIFICANCE STATEMENT The anterior cingulate cortex (ACC) is a critical brain area in physical and social pain and contributes to the exhibition of abnormal behavior. ACC glutamatergic neurons have been shown to encode transmitted stress, but it remains unclear whether inhibitory ACC neurons also play a role. We evaluate, in this study, ACC neuronal, synaptic and network activities and uncover a critical role of parvalbumin interneurons (PVINs) in the expression of transmitted stress in adolescent mice who had witnessed MS of littermates in infancy. Furthermore, inactivation of ACC PVINs blocks transmitted stress. The results suggest that emotional contagion has a severe effect on brain function, and identify a potential target for the treatment of transmitted anxiety.
Collapse
Affiliation(s)
- Jinxiang Jiang
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Shuyi Tan
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiaoyi Feng
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yigang Peng
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|
6
|
Herselman MF, Bobrovskaya L. The Effects of Chronic Unpredictable Mild Stress and Semi-Pure Diets on the Brain, Gut and Adrenal Medulla in C57BL6 Mice. Int J Mol Sci 2023; 24:14618. [PMID: 37834073 PMCID: PMC10572190 DOI: 10.3390/ijms241914618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Chronic stress is known to perturb serotonergic regulation in the brain, leading to mood, learning and memory impairments and increasing the risk of developing mood disorders. The influence of the gut microbiota on serotonergic regulation in the brain has received increased attention recently, justifying the investigation of the role of diet on the gut and the brain in mood disorders. Here, using a 4-week chronic unpredictable mild stress (CUMS) model in mice, we aimed to investigate the effects of a high-fat high-glycaemic index (HFD) and high-fibre fruit & vegetable "superfood" (SUP) modifications of a semi-pure AIN93M diet on behaviour, serotonin synthesis and metabolism pathway regulation in the brain and the gut, as well as the gut microbiota and the peripheral adrenal medullary system. CUMS induced anxiety-like behaviour, dysregulated the tryptophan and serotonin metabolic pathways in the hippocampus, prefrontal cortex, and colon, and altered the composition of the gut microbiota. CUMS reduced the catecholamine synthetic capacity of the adrenal glands. Differential effects were found in these parameters in the HFD and SUP diet. Thus, dietary modifications may profoundly affect the multiple dynamic systems involved in mood disorders.
Collapse
Affiliation(s)
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| |
Collapse
|
7
|
LeDuke DO, Borio M, Miranda R, Tye KM. Anxiety and depression: A top-down, bottom-up model of circuit function. Ann N Y Acad Sci 2023; 1525:70-87. [PMID: 37129246 PMCID: PMC10695657 DOI: 10.1111/nyas.14997] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
A functional interplay of bottom-up and top-down processing allows an individual to appropriately respond to the dynamic environment around them. These processing modalities can be represented as attractor states using a dynamical systems model of the brain. The transition probability to move from one attractor state to another is dependent on the stability, depth, neuromodulatory tone, and tonic changes in plasticity. However, how does the relationship between these states change in disease states, such as anxiety or depression? We describe bottom-up and top-down processing from Marr's computational-algorithmic-implementation perspective to understand depressive and anxious disease states. We illustrate examples of bottom-up processing as basolateral amygdala signaling and projections and top-down processing as medial prefrontal cortex internal signaling and projections. Understanding these internal processing dynamics can help us better model the multifaceted elements of anxiety and depression.
Collapse
Affiliation(s)
- Deryn O. LeDuke
- Salk Institute for Biological Studies, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, USA
| | - Matilde Borio
- Salk Institute for Biological Studies, La Jolla, California, USA
| | - Raymundo Miranda
- Salk Institute for Biological Studies, La Jolla, California, USA
- Neurosciences Graduate Program, University of California San Diego, La Jolla, California, USA
| | - Kay M. Tye
- Salk Institute for Biological Studies, La Jolla, California, USA
- Howard Hughes Medical Institute, La Jolla, California, USA
- Kavli Institute for the Brain and Mind, La Jolla, California, USA
| |
Collapse
|
8
|
Inhibition of norepinephrine signaling during a sensitive period disrupts locus coeruleus circuitry and emotional behaviors in adulthood. Sci Rep 2023; 13:3077. [PMID: 36813805 PMCID: PMC9946949 DOI: 10.1038/s41598-023-29175-x] [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: 07/06/2022] [Accepted: 01/31/2023] [Indexed: 02/24/2023] Open
Abstract
Deficits in arousal and stress responsiveness are a feature of numerous psychiatric disorders including depression and anxiety. Arousal is supported by norepinephrine (NE) released from specialized brainstem nuclei, including the locus coeruleus (LC) neurons into cortical and limbic areas. During development, the NE system matures in concert with increased exploration of the animal's environment. While several psychiatric medications target the NE system, the possibility that its modulation during discreet developmental periods can have long-lasting consequences has not yet been explored. We used a chemogenetic strategy in mice to reversibly inhibit NE signaling during brief developmental periods and then evaluated any long-lasting impact of our intervention on adult NE circuit function and on emotional behavior. We also tested whether developmental exposure to the α2 receptor agonist guanfacine, which is commonly used in the pediatric population and is not contraindicated during pregnancy and nursing, recapitulates the effect seen with the chemogenetic strategy. Our results reveal that postnatal days 10-21 constitute a sensitive period during which alterations in NE signaling lead to changes in baseline anxiety, increased anhedonia, and passive coping behaviors in adulthood. Disruption of NE signaling during this sensitive period also caused altered LC autoreceptor function, along with circuit specific changes in LC-NE target regions at baseline, and in response to stress. Our findings indicate an early critical role for NE in sculpting brain circuits that support adult emotional function. Interfering with this role by guanfacine and similar clinically used drugs can have lasting implications for mental health.
Collapse
|
9
|
Argañaraz CV, Adjimann TS, Perissinotti PP, Soiza-Reilly M. Selective refinement of glutamate and GABA synapses on dorsal raphe 5-HT neurons during postnatal life. Development 2022; 149:285818. [PMID: 36458556 DOI: 10.1242/dev.201121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) neurons are implicated in the etiology and therapeutics of anxiety and depression. Critical periods of vulnerability during brain development enable maladaptive mechanisms to produce detrimental consequences on adult mood and emotional responses. 5-HT plays a crucial role in these mechanisms; however, little is known about how synaptic inputs and modulatory systems that shape the activity of early 5-HT networks mature during postnatal development. We investigated in mice the postnatal trajectory of glutamate and GABA synaptic inputs to dorsal raphe nucleus (DRN) 5-HT neurons, the main source of forebrain 5-HT. High-resolution quantitative analyses with array tomography and ex vivo electrophysiology indicate that cortical glutamate and subcortical GABA synapses undergo a profound refinement process after the third postnatal week, whereas subcortical glutamate inputs do not. This refinement of DRN inputs is not accompanied by changes in 5-HT1A receptor-mediated inhibition over 5-HT neurons. Our study reveals a precise developmental pattern of synaptic refinement of DRN excitatory and inhibitory afferents, when 5-HT-related inhibitory mechanisms are in place. These findings contribute to the understanding of neurodevelopmental vulnerability to psychiatric disorders. This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Carla V Argañaraz
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos AiresC1428EGA, Argentina
| | - Tamara S Adjimann
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos AiresC1428EGA, Argentina
| | - Paula P Perissinotti
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos AiresC1428EGA, Argentina
| | - Mariano Soiza-Reilly
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos AiresC1428EGA, Argentina
| |
Collapse
|
10
|
Fabio MC, Servin-Bernal IJC, Degano AL, Pautassi RM. Serotonin disruption at gestation alters expression of genes associated with serotonin synthesis and reuptake at weaning. Psychopharmacology (Berl) 2022; 239:3355-3366. [PMID: 36063206 DOI: 10.1007/s00213-022-06228-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/27/2022] [Indexed: 12/28/2022]
Abstract
RATIONALE Serotonin (5-HT) is a monoamine neuromodulator that plays a key role in the organization of the central nervous system. 5-HT alterations may be associated to the emergence of social deficits and psychiatric disorders, including anxiety, depression, and substance abuse disorders. Notably, disruption of the 5-HT system during sensitive periods of development seems to exert long-term consequences, including altered anxiety responses and problematic use of alcohol. OBJECTIVE We analyzed, in mice, the effects of transient 5-HT depletion at gestation (a developmental stage when medial prefrontal cortex (mPFC) 5-HT levels depend exclusively on placental 5-HT availability) on 5-HT central synthesis and reuptake at weaning. We also explored if 5-HT disruption at the embryonic stage influences behavioral outcomes that may serve as a proxy for autistic- or anxiety-like phenotypes. METHODS C57/BL6 male and female mice, born from dams treated with a 5-HT synthesis inhibitor (PCPA; 4-Chloro-DL-phenylalanine methyl ester hydrochloride) at gestational days (G)13.5-16.5, were subjected to a behavioral battery that assesses social preference and novelty, compulsive behavior, stereotypies, and ethanol's anti-anxiety effects, at postnatal days (P) 21-28. Afterwards, expression of the genes that encode for 5-HT synthesis (Tph2) and SERT (5-HT transporter) were analyzed in mPFC via real-time RT-PCR. Dopamine 2 receptor (D2R) expression was also analyzed via RT-PCR to further explore possible effects of PCPA on dopaminergic transmission. RESULTS Transient 5-HT disruption at G13.5-16.5 reduced Tph2 expression of both male and female mice in mPFC at P23. Notably, female mice also exhibited higher SERT expression and reduced D2R expression in mPFC. Mice derived from 5-HT depleted dams displayed heightened compulsive behavior at P21, when compared to control mice. Alcohol anti-anxiety effects at early adolescence (P28) were exhibited by mice derived from 5-HT depleted dams, but not by control counterparts. No social deficits or stereotyped behaviors were observed. CONCLUSION Transient 5-HT inhibition at gestation resulted in altered expression of genes involved in 5-HT synthesis and reuptake in mPFC at weaning, a period in which the 5-HT system is still developing. These alterations may exert lingering effects, which translate to significant compulsivity and heightened sensitivity to the anxiolytic effects of alcohol at early adolescence.
Collapse
Affiliation(s)
- M C Fabio
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Cordoba, Argentina. .,Facultad de Psicología, Universidad Nacional de Córdoba, Cordoba, Argentina.
| | - I J C Servin-Bernal
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Cordoba, Argentina
| | - A L Degano
- Departamento de Química Biológica Ranwel CaputtoFacultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba, Argentina.,Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Universidad Nacional de Córdoba, Cordoba, Argentina
| | - R M Pautassi
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Cordoba, Argentina.,Facultad de Psicología, Universidad Nacional de Córdoba, Cordoba, Argentina
| |
Collapse
|
11
|
Cortes-Altamirano JL, Yáñes-Pizaña A, Reyes-Long S, Angélica GM, Bandala C, Bonilla-Jaime H, Alfaro-Rodríguez A. Potential Neuroprotective Effect of Cannabinoids in Covid-19 Patients. Curr Top Med Chem 2022; 22:1326-1345. [PMID: 35382723 DOI: 10.2174/1568026622666220405143003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/29/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022]
Abstract
The global pandemic caused by the SARS-CoV-2 virus began in early 2020 and is still present. The respiratory symptoms caused by COVID-19 are well established, however, neurological manifestations that may result from direct or indirect neurological damage after SARS-CoV-2 infection have been reported frequently. The main proposed pathophysiological processes leading to neurological damage in COVID-19 are cerebrovascular disease, and indirect mechanisms of inflammatory / autoimmune origin. A growing number of studies confirm that neuroprotective measures should be maintained in COVID-19 patients. On the other hand, cannabinoids have been the subject of various studies that propose them as potential promising drugs in chronic neurodegenerative diseases due to their powerful neuroprotective potential. In this review we address the possible mechanism of action of cannabinoids as a neuroprotective treatment in patients infected by SARS-CoV-2. The endocannabinoid system is found in multiple systems within the body, including the immune system. Its activation can lead to beneficial results, such as a decrease in viral entry, a decrease in viral replication, and a decrease in pro-inflammatory cytokines such as IL-2, IL-4, IL-6, IL-12, TNF-α or IFN-c through CB2R expression induced during inflammation by SARS-CoV-2 infection in the central nervous system.
Collapse
Affiliation(s)
- José Luis Cortes-Altamirano
- Division de Neurociencias, Instituto Nacional de Rehabilitación, Secretaría de Salud, Mexico City, 14389, Mexico.,Departamento de Quiropráctica, Universidad Estatal del Valle de Ecatepec, Estado de Mexico, 55210, Mexico
| | - Ariadna Yáñes-Pizaña
- Escuela de Ciencias de la Salud, Medicina Veterinaria y Zootecnia, Universidad del Valle de Mexico, Mexico City, 04910, México.,Escuela de Medicina Veterinaria y Zootecnia en Pequeñas Especies, Federación Canofila Mexicana, Mexico City, 14430, México
| | - Samuel Reyes-Long
- Division de Neurociencias, Instituto Nacional de Rehabilitación, Secretaría de Salud, Mexico City, 14389, Mexico.,Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, 07738, México
| | - González-Maciel Angélica
- Laboratory of Cell and Tissue Morphology, Instituto Nacional de Pediatría, Secretaría de Salud, Insurgentes Sur No. 3700-C, Mexico City, C. P. 04530, Mexico
| | - Cindy Bandala
- Division de Neurociencias, Instituto Nacional de Rehabilitación, Secretaría de Salud, Mexico City, 14389, Mexico.,Escuela de Medicina Veterinaria y Zootecnia en Pequeñas Especies, Federación Canofila Mexicana, Mexico City, 14430, México
| | - Herlinda Bonilla-Jaime
- Departamento de Biología de la reproducción, Universidad Autónoma Metropolitana, Mexico City, 09340, Mexico
| | - Alfonso Alfaro-Rodríguez
- Division de Neurociencias, Instituto Nacional de Rehabilitación, Secretaría de Salud, Mexico City, 14389, Mexico
| |
Collapse
|
12
|
Sex-based changes in rat brain serotonin and behavior in a model of altitude-related vulnerability to treatment-resistant depression. Psychopharmacology (Berl) 2021; 238:2867-2881. [PMID: 34159421 DOI: 10.1007/s00213-021-05902-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/08/2021] [Indexed: 01/03/2023]
Abstract
RATIONALE Rates of depression and suicide increase with altitude. In our animal model, rats housed at moderate altitude vs. at sea level exhibit increased depressive symptoms in the forced swim test (FST) and lack of response to selective serotonin reuptake inhibitors (SSRIs). Depression and SSRI resistance are linked to disrupted serotonergic function, and hypobaric hypoxia may reduce the oxygen-dependent synthesis of serotonin. We therefore tested brain serotonin in rats housed at altitude. METHODS Sprague-Dawley rats were housed at altitude (4,500 ft, 10,000 ft) vs. sea level for 7-36 days. Brain serotonin was measured by ELISA, or behavior evaluated in the FST, sucrose preference (SPT), or open-field tests (OFT). RESULTS After 2 weeks at 4,500 ft or 10,000ft vs. sea level, serotonin levels decreased significantly at altitude in the female prefrontal cortex, striatum, hippocampus, and brainstem, but increased with altitude in the male hippocampus and brainstem. Female brain serotonin decreased from 7 to 36 days at 4,500 ft, but males did not vary. At 2 weeks and 24 days, females at altitude exhibit lower brain serotonin and increased depressive symptoms in the FST and SPT, with motor behavior unaltered. In males, serotonin, passive coping in the FST and OFT immobility increased with altitude at 2 weeks, but not at 24 days. Male SPT behavior did not change with altitude. CONCLUSIONS Females may be more vulnerable to depressive symptoms at altitude, while males may be resilient. Chronic hypoxic stress at altitudes as low as 4,500 ft may cause a brain serotonin imbalance to worsen vulnerability to depression and SSRI resistance, and potentially worsen suicide risk.
Collapse
|
13
|
Salman T, Afroz R, Nawaz S, Mahmood K, Haleem DJ, Zarina S. Differential effects of memory enhancing and impairing doses of methylphenidate on serotonin metabolism and 5-HT1A, GABA, glutamate receptor expression in the rat prefrontal cortex. Biochimie 2021; 191:51-61. [PMID: 34454977 DOI: 10.1016/j.biochi.2021.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 07/28/2021] [Accepted: 08/24/2021] [Indexed: 02/05/2023]
Abstract
Methylphenidate (MPD), a psychostimulant, is a prescription medicine for treating attention deficit hyperactivity disorder (ADHD). Previously we have shown that moderate doses of MPD enhanced learning and memory while higher doses impaired it. To understand neurochemical mechanisms and receptors involved in memory enhancing and impairing effects of MPD, the present study concerns the effects of these doses of MPD on serotonin, 5-HT1A, GABA, and NMDA receptor mRNA expression in the prefrontal cortex (PFC). We found that low doses (2.5 mg/kg) of MPD improved performance in the water-maze test but higher doses (5 mg/kg) impaired memory retention. Animals showing improved performance had high 5-HT metabolism in the PFC while these levels were not affected in the group treated with higher MPD doses and exhibiting impaired memory. There was downregulation of 5-HT1A receptors in the PFC of rats treated with higher dose MPD, which didn't occur in low dose of MPD treated animals. Further, a decrease in GABAAreceptor mRNA expression occurred in low doses of MPD treated animals and GluN2A expression was reduced in higher doses of MPD treated animals. The findings suggest that memory enhancing doses of MPD increase 5-HT and reduce GABAA receptor mRNA expression in the PFC to release excitatory glutamate neurons from the inhibitory influence of GABA. Conversely, higher dose of MPD downregulates 5-HT1A receptor mRNA expression to enhance inhibitory GABA influence on glutamate neurons and impair cognitive performance. The findings show an important role of 5-HT1A heteroreceptors in the PFC for improving therapeutic use of MPD and developing novel cognitive enhancers.
Collapse
Affiliation(s)
- Tabinda Salman
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan.
| | - Rushda Afroz
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Shazia Nawaz
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Khalid Mahmood
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Darakhshan J Haleem
- Neuroscience Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Shamshad Zarina
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
14
|
Palamarchuk IS, Vaillancourt T. Mental Resilience and Coping With Stress: A Comprehensive, Multi-level Model of Cognitive Processing, Decision Making, and Behavior. Front Behav Neurosci 2021; 15:719674. [PMID: 34421556 PMCID: PMC8377204 DOI: 10.3389/fnbeh.2021.719674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
Aversive events can evoke strong emotions that trigger cerebral neuroactivity to facilitate behavioral and cognitive shifts to secure physiological stability. However, upon intense and/or chronic exposure to such events, the neural coping processes can be maladaptive and disrupt mental well-being. This maladaptation denotes a pivotal point when psychological stress occurs, which can trigger subconscious, "automatic" neuroreactivity as a defence mechanism to protect the individual from potential danger including overwhelming unpleasant feelings and disturbing or threatening thoughts.The outcomes of maladaptive neural activity are cognitive dysfunctions such as altered memory, decision making, and behavior that impose a risk for mental disorders. Although the neurocognitive phenomena associated with psychological stress are well documented, the complex neural activity and pathways related to stressor detection and stress coping have not been outlined in detail. Accordingly, we define acute and chronic stress-induced pathways, phases, and stages in relation to novel/unpredicted, uncontrollable, and ambiguous stressors. We offer a comprehensive model of the stress-induced alterations associated with multifaceted pathophysiology related to cognitive appraisal and executive functioning in stress.
Collapse
Affiliation(s)
- Iryna S Palamarchuk
- Counselling Psychology, Faculty of Education, University of Ottawa, Ottawa, ON, Canada
| | - Tracy Vaillancourt
- Counselling Psychology, Faculty of Education, University of Ottawa, Ottawa, ON, Canada.,School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
15
|
Hayley S, Hakim AM, Albert PR. Depression, dementia and immune dysregulation. Brain 2021; 144:746-760. [PMID: 33279966 DOI: 10.1093/brain/awaa405] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/26/2020] [Accepted: 09/20/2020] [Indexed: 12/17/2022] Open
Abstract
Major depression is a prevalent illness that increases the risk of several neurological conditions. These include stroke, cardiovascular disease, and dementia including Alzheimer's disease. In this review we ask whether certain types of depression and associated loneliness may be a harbinger of cognitive decline and possibly even dementia. We propose that chronic stress and inflammation combine to compromise vascular and brain function. The resulting increases in proinflammatory cytokines and microglial activation drive brain pathology leading to depression and mild cognitive impairment, which may progress to dementia. We present evidence that by treating the inflammatory changes, depression can be reversed in many cases. Importantly, there is evidence that anti-inflammatory and antidepressant treatments may reduce or prevent dementia in people with depression. Thus, we propose a model in which chronic stress and inflammation combine to increase brain permeability and cytokine production. This leads to microglial activation, white matter damage, neuronal and glial cell loss. This is first manifest as depression and mild cognitive impairment, but can eventually evolve into dementia. Further research may identify clinical subgroups with inflammatory depression at risk for dementia. It would then be possible to address in clinical trials whether effective treatment of the depression can delay the onset of dementia.
Collapse
Affiliation(s)
- Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Antoine M Hakim
- Ottawa Hospital Research Institute (Neuroscience), uOttawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), uOttawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
16
|
Kanekar S, Ettaro R, Hoffman MD, Ombach HJ, Brown J, Lynch C, Sheth CS, Renshaw PF. Sex-Based Impact of Creatine Supplementation on Depressive Symptoms, Brain Serotonin and SSRI Efficacy in an Animal Model of Treatment-Resistant Depression. Int J Mol Sci 2021; 22:ijms22158195. [PMID: 34360959 PMCID: PMC8348220 DOI: 10.3390/ijms22158195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 02/05/2023] Open
Abstract
Background: Rates of major depressive disorder (MDD) increase with living at altitude. In our model, rats housed at moderate altitude (in hypobaric hypoxia) exhibit increased depression-like behavior, altered brain serotonin and a lack of antidepressant response to most selective serotonin reuptake inhibitors (SSRIs). A forebrain deficit in the bioenergetic marker creatine is noted in people living at altitude or with MDD. Methods: Rats housed at 4500 ft were given dietary creatine monohydrate (CRMH, 4% w/w, 5 weeks) vs. un-supplemented diet, and impact on depression-like behavior, brain bioenergetics, serotonin and SSRI efficacy assessed. Results: CRMH significantly improved brain creatine in a sex-based manner. At altitude, CRMH increased serotonin levels in the female prefrontal cortex and striatum but reduced male striatal and hippocampal serotonin. Dietary CRMH was antidepressant in the forced swim test and anti-anhedonic in the sucrose preference test in only females at altitude, with motor behavior unchanged. CRMH improved fluoxetine efficacy (20 mg/kg) in only males at altitude: CRMH + SSRI significantly improved male striatal creatine and serotonin vs. CRMH alone. Conclusions: Dietary CRMH exhibits sex-based efficacy in resolving altitude-related deficits in brain biomarkers, depression-like behavior and SSRI efficacy, and may be effective clinically for SSRI-resistant depression at altitude. This is the first study to link CRMH treatment to improving brain serotonin.
Collapse
Affiliation(s)
- Shami Kanekar
- Diagnostic Neuroimaging, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA; (R.E.); (M.D.H.); (H.J.O.); (J.B.); (C.L.); (C.S.S.); (P.F.R.)
- VISN19 MIRECC, 500 Foothill Drive, Salt Lake City, UT 84148, USA
- Veterans Affairs Salt Lake City Health Care System, 500 Foothill Drive, Salt Lake City, UT 84148, USA
- Correspondence: ; Tel.: +1-801-587-1477 or +1-801-585-5375
| | - Robert Ettaro
- Diagnostic Neuroimaging, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA; (R.E.); (M.D.H.); (H.J.O.); (J.B.); (C.L.); (C.S.S.); (P.F.R.)
| | - Michael D. Hoffman
- Diagnostic Neuroimaging, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA; (R.E.); (M.D.H.); (H.J.O.); (J.B.); (C.L.); (C.S.S.); (P.F.R.)
| | - Hendrik J. Ombach
- Diagnostic Neuroimaging, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA; (R.E.); (M.D.H.); (H.J.O.); (J.B.); (C.L.); (C.S.S.); (P.F.R.)
| | - Jadeda Brown
- Diagnostic Neuroimaging, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA; (R.E.); (M.D.H.); (H.J.O.); (J.B.); (C.L.); (C.S.S.); (P.F.R.)
| | - Cayla Lynch
- Diagnostic Neuroimaging, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA; (R.E.); (M.D.H.); (H.J.O.); (J.B.); (C.L.); (C.S.S.); (P.F.R.)
| | - Chandni S. Sheth
- Diagnostic Neuroimaging, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA; (R.E.); (M.D.H.); (H.J.O.); (J.B.); (C.L.); (C.S.S.); (P.F.R.)
| | - Perry F. Renshaw
- Diagnostic Neuroimaging, Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA; (R.E.); (M.D.H.); (H.J.O.); (J.B.); (C.L.); (C.S.S.); (P.F.R.)
- VISN19 MIRECC, 500 Foothill Drive, Salt Lake City, UT 84148, USA
- Veterans Affairs Salt Lake City Health Care System, 500 Foothill Drive, Salt Lake City, UT 84148, USA
| |
Collapse
|
17
|
Stupin KN, Zenko MY, Rybnikova EA. Comparative Analysis of Pathobiochemical Changes in Major Depression and Post-Traumatic Stress Disorder. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:729-736. [PMID: 34225595 DOI: 10.1134/s0006297921060109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 06/13/2023]
Abstract
Comparative analysis of available literature data on the pathogenetic neuroendocrine mechanisms of depression and post-traumatic stress disorder (PTSD) is provided in this review to identify their common features and differences. We discuss the multidirectional modifications of the activity of cortical and subcortical structures of the brain, levels of neurotransmitters and their receptors, and functions of the hypothalamic-pituitary-adrenocortical axis in depression and PTSD. The analysis shows that these disorders are examples of opposite failures in the system of adaptive stress response of the body to stressful psychotraumatic events. On this basis, it is concluded that the currently widespread use of similar approaches to treat these disorders is not justified, despite the significant similarity of their anxiety-depressive symptoms; development of differential therapeutic strategies is required.
Collapse
Affiliation(s)
- Konstantin N Stupin
- Pavlov Institute of Physiology, Russian Academy of Sciences, St.-Petersburg, 199034, Russia
| | - Mikhail Y Zenko
- Pavlov Institute of Physiology, Russian Academy of Sciences, St.-Petersburg, 199034, Russia
| | - Elena A Rybnikova
- Pavlov Institute of Physiology, Russian Academy of Sciences, St.-Petersburg, 199034, Russia.
| |
Collapse
|
18
|
Wiring of higher-order cortical areas: Spatiotemporal development of cortical hierarchy. Semin Cell Dev Biol 2021; 118:35-49. [PMID: 34034988 DOI: 10.1016/j.semcdb.2021.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 01/04/2023]
Abstract
A hierarchical development of cortical areas was suggested over a century ago, but the diversity and complexity of cortical hierarchy properties have so far prevented a formal demonstration. The aim of this review is to clarify the similarities and differences in the developmental processes underlying cortical development of primary and higher-order areas. We start by recapitulating the historical and recent advances underlying the biological principle of cortical hierarchy in adults. We then revisit the arguments for a hierarchical maturation of cortical areas, and further integrate the principles of cortical areas specification during embryonic and postnatal development. We highlight how the dramatic expansion in cortical size might have contributed to the increased number of association areas sustaining cognitive complexification in evolution. Finally, we summarize the recent observations of an alteration of cortical hierarchy in neuropsychiatric disorders and discuss their potential developmental origins.
Collapse
|
19
|
Sałaciak K, Pytka K. Biased agonism in drug discovery: Is there a future for biased 5-HT 1A receptor agonists in the treatment of neuropsychiatric diseases? Pharmacol Ther 2021; 227:107872. [PMID: 33905796 DOI: 10.1016/j.pharmthera.2021.107872] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022]
Abstract
Serotonin (5-HT) is one of the fundamental neurotransmitters that contribute to the information essential for an organism's normal, physiological function. Serotonin acts centrally and systemically. The 5-HT1A receptor is the most widespread serotonin receptor, and participates in many brain-related disorders, including anxiety, depression, and cognitive impairments. The 5-HT1A receptor can activate several different biochemical pathways and signals through both G protein-dependent and G protein-independent pathways. Preclinical experiments indicate that distinct signaling pathways in specific brain regions may be crucial for antidepressant-like, anxiolytic-like, and procognitive responses. Therefore, the development of new ligands that selectively target a particular signaling pathway(s) could open new possibilities for more effective and safer pharmacotherapy. This review discusses the current state of preclinical studies focusing on the concept of functional selectivity (biased agonism) regarding the 5-HT1A receptor and its role in antidepressant-like, anxiolytic-like, and procognitive regulation. Such work highlights not only the differential effects of targeted autoreceptors, vs. heteroreceptors, but also the importance of targeting specific downstream intracellular signaling processes, thereby enhancing favorable over unfavorable signaling activation.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| |
Collapse
|
20
|
Abstract
This paper introduces a new construct, the 'pivotal mental state', which is defined as a hyper-plastic state aiding rapid and deep learning that can mediate psychological transformation. We believe this new construct bears relevance to a broad range of psychological and psychiatric phenomena. We argue that pivotal mental states serve an important evolutionary function, that is, to aid psychological transformation when actual or perceived environmental pressures demand this. We cite evidence that chronic stress and neurotic traits are primers for a pivotal mental state, whereas acute stress can be a trigger. Inspired by research with serotonin 2A receptor agonist psychedelics, we highlight how activity at this particular receptor can robustly and reliably induce pivotal mental states, but we argue that the capacity for pivotal mental states is an inherent property of the human brain itself. Moreover, we hypothesize that serotonergic psychedelics hijack a system that has evolved to mediate rapid and deep learning when its need is sensed. We cite a breadth of evidences linking stress via a variety of inducers, with an upregulated serotonin 2A receptor system (e.g. upregulated availability of and/or binding to the receptor) and acute stress with 5-HT release, which we argue can activate this primed system to induce a pivotal mental state. The pivotal mental state model is multi-level, linking a specific molecular gateway (increased serotonin 2A receptor signaling) with the inception of a hyper-plastic brain and mind state, enhanced rate of associative learning and the potential mediation of a psychological transformation.
Collapse
Affiliation(s)
- Ari Brouwer
- Centre for Psychedelic Research, Imperial College London, London, United Kingdom
| | | |
Collapse
|
21
|
Lages YVM, Rossi AD, Krahe TE, Landeira-Fernandez J. Effect of chronic unpredictable mild stress on the expression profile of serotonin receptors in rats and mice: a meta-analysis. Neurosci Biobehav Rev 2021; 124:78-88. [PMID: 33524415 DOI: 10.1016/j.neubiorev.2021.01.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/17/2022]
Abstract
Chronic-stress-induced depression is recognized as a widespread public health concern. Selective serotonin reuptake inhibitors (SSRIs) have been the most common treatment for this illness. However, the role of 5-hydroxytryptamine (5-HT) receptor subtypes in stress-induced depression remains unclear. Evidence from Animal studies has reported a variety of results regarding the effects of chronic unpredictable mild stress (CUMS) on serotonin signaling pathways and 5-HT receptor subtypes. This divergence may rely on differences in protocols, methods, and studied pathways. Thus, the aim of this systematic review was to weigh the currently available findings regarding serotonin receptor changes in animal models of CUMS. Overall, our meta-analysis results showed the association of altered expression of 5-HT1A receptors in the frontal cortex and 5-HT2A receptors both in the whole cortex and the hypothalamus of rats following CUMS. Moreover, by using a qualitative-structured analysis and the application of risk-of-bias tools, we identified possible sources of data variation between the studied literature, which should be taken into account in future animal studies of chronic-stress induced depression.
Collapse
Affiliation(s)
- Y V M Lages
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - A D Rossi
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - T E Krahe
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J Landeira-Fernandez
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
22
|
Transient serotonin depletion at adolescence, but not at early infancy, reduced subsequent anxiety-like behavior and alcohol intake in female mice. Psychopharmacology (Berl) 2021; 238:215-225. [PMID: 33011817 DOI: 10.1007/s00213-020-05670-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/21/2020] [Indexed: 12/27/2022]
Abstract
RATIONALE Serotonin (5-HT) plays an important role in the organization of the central nervous system and in the development of social interaction deficits and psychiatric disorders, including anxiety, depression, and addiction disorders. Notably, disruption of the 5-HT system during sensitive periods of development exerts long-term consequences, including altered anxiety response and problematic use of alcohol. OBJECTIVE we analyzed, in mice, the effects of transient 5-HT depletion at infancy or adolescence on subsequent anxiety-like behavior and alcohol intake during adolescence. METHODS C57/BL6 male and female mice were administered a 5-HT synthesis inhibitor (PCPA; 4-chloro-DL-phenylalanine methyl ester hydrochloride) at infancy (postnatal days 14-16 [PD14-16]) or adolescence (PD40-42). Eleven (± 1) days after treatment, mice were assessed for ethanol intake in daily two-bottle choice tests and for anxiety response via the elevated plus maze. RESULTS Female, but not male, mice transiently depleted of 5-HT at adolescence (but not those depleted at the perinatal stage) exhibited a significant reduction in anxiety response, which was accompanied by a significant reduction on alcohol intake. CONCLUSION Transient 5-HT inhibition at adolescence may act, in females, as a protective factor for the emergence of anxiety disorders and problematic use of alcohol during adolescence.
Collapse
|
23
|
Sałaciak K, Głuch-Lutwin M, Siwek A, Szafarz M, Kazek G, Bednarski M, Nowiński L, Mitchell E, Jastrzębska-Więsek M, Partyka A, Wesołowska A, Kołaczkowski M, Szkaradek N, Marona H, Sapa J, Pytka K. The antidepressant-like activity of chiral xanthone derivatives may be mediated by 5-HT1A receptor and β-arrestin signalling. J Psychopharmacol 2020; 34:1431-1442. [PMID: 33103555 DOI: 10.1177/0269881120959605] [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] [Indexed: 01/14/2023]
Abstract
BACKGROUND Our previous studies showed that xanthone derivatives with N-(2-methoxyphenyl)piperazine fragment have an affinity to the 5-HT1A receptor and show antidepressant-like properties in rodents. In this study, we tested three xanthone derivatives, HBK-1 (R, S) and its enantiomers, in which we increased the distance between the piperazine and xanthone fragments by using a hydroxypropoxy linker. We hypothesized that this would increase the binding to the 5-HT1A receptor and consequently, pharmacological activity. AIMS We aimed to assess the in vitro and in vivo pharmacological activity of the xanthone derivatives. METHODS We evaluated the in vitro affinity for serotonin 5-HT1A and 5-HT2A receptors and serotonin transporter. We also determined the intrinsic activity at the 5-HT1A receptor. We investigated the antidepressant-like properties and safety after acute administration (dose range: 1.25-20 mg/kg) using the forced swim, tail suspension, locomotor activity, rotarod and chimney tests in mice. We also evaluated the basic pharmacokinetic parameters. RESULTS Our results indicated that the compounds showed a high affinity for the 5-HT1A receptor but very weak antagonistic properties in the Ca2+ mobilization assay; however, they showed significant agonistic properties in the β-arrestin recruitment assay. In both behavioural tests the studied xanthone derivatives showed antidepressant-like activity. Pre-treatment with p-chlorophenylalanine or WAY-100635 abolished their antidepressant-like activity. None of the compounds caused motor impairments at antidepressant-like doses. The racemate penetrated the blood-brain barrier and had a relatively high bioavailability after intraperitoneal administration. CONCLUSIONS Xanthone derivatives with N-(2-methoxyphenyl)piperazine fragment and hydroxypropoxy linker show increased binding to the 5-HT1A receptor and may represent an attractive putative treatment candidate for depression.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland
| | - Agata Siwek
- Department of Pharmacobiology, Jagiellonian University Medical College, Kraków, Poland
| | - Małgorzata Szafarz
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Kazek
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Marek Bednarski
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Leszek Nowiński
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Emma Mitchell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | | | - Anna Partyka
- Department of Clinical Pharmacy, Jagiellonian University Medical College Kraków, Kraków, Poland
| | - Anna Wesołowska
- Department of Clinical Pharmacy, Jagiellonian University Medical College Kraków, Kraków, Poland
| | - Marcin Kołaczkowski
- Department of Medicinal Chemistry, Jagiellonian University Medical College Kraków, Kraków, Poland
| | - Natalia Szkaradek
- Department of Bioorganic Chemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Henryk Marona
- Department of Bioorganic Chemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Sapa
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
24
|
Murack M, Chandrasegaram R, Smith KB, Ah-Yen EG, Rheaume É, Malette-Guyon É, Nanji Z, Semchishen SN, Latus O, Messier C, Ismail N. Chronic sleep disruption induces depression-like behavior in adolescent male and female mice and sensitization of the hypothalamic-pituitary-adrenal axis in adolescent female mice. Behav Brain Res 2020; 399:113001. [PMID: 33197456 DOI: 10.1016/j.bbr.2020.113001] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/01/2020] [Accepted: 11/01/2020] [Indexed: 01/05/2023]
Abstract
Depression is a prevalent mood disorder responsible for reduced quality of life for over 264 million people. Depression commonly develops during adolescence and becomes twice as prevalent in females than in males. However, the mechanisms underlying adolescent depression onset and sex differences in the prevalence rate remain unclear. Adolescent exposure to stress and subsequent sensitization of the hypothalamic-pituitary-adrenal (HPA) axis contributes to mood disorder development, and females are particularly vulnerable to HPA sensitization. Repeated exposure to stressors common to adolescent development, like sleep disruption, could partially be responsible for adolescent female susceptibility to depression. To address this possibility, 80 adolescent and adult CD-1 mice (Male, n = 40; Female, n = 40) were manually sleep disrupted for the first four hours of each rest cycle or allowed normal rest for eight consecutive days. Depression-like behavior was assessed with the forced swim test. 5-HT1A and glucocorticoid receptor expression and concurrent cellular activation via glucocorticoid receptor/c-Fos colocalization were examined in various brain regions to assess cellular correlates of depression and HPA-axis activation. Both adolescent male and female mice displayed significantly greater depression-like behavior and prelimbic c-Fos expression after chronic sleep disruption than non-sleep disrupted adolescent and sleep disrupted adult counterparts. However, sleep disrupted adolescent females demonstrated greater dorsal raphe 5-HT1A expression than sleep disrupted adolescent males. Adolescent females and males had decreased medial prefrontal 5-HT1A expression after chronic sleep disruption, but only adolescent females expressed decreased hippocampal 5-HT1A expression compared to controls. Chronic sleep disruption significantly increased corticosterone release, glucocorticoid expression in the CA1, and activation of glucocorticoid immunoreactive cells in the prelimbic cortex of adolescent females but not in adolescent males. These findings suggest that chronic sleep disruption during adolescence could give rise to depressive symptoms in male and female adolescents through differing signaling mechanisms.
Collapse
Affiliation(s)
- Michael Murack
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Kevin B Smith
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Emily G Ah-Yen
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Étienne Rheaume
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Zahra Nanji
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Olivia Latus
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Claude Messier
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Canada
| | - Nafissa Ismail
- Department of Psychology, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Canada.
| |
Collapse
|
25
|
Tripathi SJ, Chakraborty S, Rao BSS. Remediation of chronic immobilization stress-induced negative affective behaviors and altered metabolism of monoamines in the prefrontal cortex by inactivation of basolateral amygdala. Neurochem Int 2020; 141:104858. [PMID: 33010391 DOI: 10.1016/j.neuint.2020.104858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/06/2020] [Accepted: 09/26/2020] [Indexed: 01/28/2023]
Abstract
Exposure to chronic stress precipitates depression and anxiety. Stress-induced responses are differentially regulated by the prefrontal cortex (PFC) and basolateral amygdala (BLA). For instance, repeated stress leads to hypertrophy of BLA, resulting in the emergence of affective symptoms. Chronic stress-induced changes in the metabolism of monoamines are central in the manifestation of affective symptoms. Interestingly, BLA via its reciprocal connections modulates prefrontal cortical monoaminergic responses to acute stress. However, the effects of BLA inactivation on chronic stress-induced affective behaviors and monoaminergic changes in the PFC are relatively unknown. Thus, we hypothesized that inactivation of BLA might prevent chronic immobilization stress (CIS)-induced depressive-, anxiety-like behaviors, and associated monoaminergic alterations in the prelimbic (PrL) and anterior cingulate cortex (ACC) subregions of PFC. We used two different BLA silencing strategies, namely ibotenic acid lesion and reversible temporary inactivation using lidocaine. We found that CIS precipitates depressive- and anxiety-like behaviors. Further, CIS-induced negative affective behaviors were associated with decreased levels of 5-HT, DA, and NE, and increased 5-HIAA/5-HT, DOPAC + HVA/DA, and MHPG/NE ratio in the PrL and ACC, suggesting enhanced metabolism. Interestingly, BLA lesion prior to CIS blocked the emergence of depressive- and anxiety-like behaviors. Moreover, the lesion of BLA prior to CIS was sufficient to prevent alterations in levels of monoamines and their metabolites in the PrL and ACC. Thereafter, we evaluated whether the effects of BLA lesion could be mirrored by temporary inactivation of BLA, specifically during stress. Remarkably, temporary inactivation of BLA during stress recapitulated the effects of lesion. Our results have implications for understanding the role of BLA in chronic stress-induced metabolic alterations in prefrontal cortical monoaminergic systems, and associated mood and anxiety disorders. The current study supports the hypothesis that combating amygdalar hyperactivity might be a viable strategy for the management of stress and associated affective disorders.
Collapse
Affiliation(s)
- Sunil Jamuna Tripathi
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bengaluru, 560 029, India
| | - Suwarna Chakraborty
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bengaluru, 560 029, India
| | - B S Shankaranarayana Rao
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bengaluru, 560 029, India.
| |
Collapse
|
26
|
Psychological mechanisms and functions of 5-HT and SSRIs in potential therapeutic change: Lessons from the serotonergic modulation of action selection, learning, affect, and social cognition. Neurosci Biobehav Rev 2020; 119:138-167. [PMID: 32931805 DOI: 10.1016/j.neubiorev.2020.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022]
Abstract
Uncertainty regarding which psychological mechanisms are fundamental in mediating SSRI treatment outcomes and wide-ranging variability in their efficacy has raised more questions than it has solved. Since subjective mood states are an abstract scientific construct, only available through self-report in humans, and likely involving input from multiple top-down and bottom-up signals, it has been difficult to model at what level SSRIs interact with this process. Converging translational evidence indicates a role for serotonin in modulating context-dependent parameters of action selection, affect, and social cognition; and concurrently supporting learning mechanisms, which promote adaptability and behavioural flexibility. We examine the theoretical basis, ecological validity, and interaction of these constructs and how they may or may not exert a clinical benefit. Specifically, we bridge crucial gaps between disparate lines of research, particularly findings from animal models and human clinical trials, which often seem to present irreconcilable differences. In determining how SSRIs exert their effects, our approach examines the endogenous functions of 5-HT neurons, how 5-HT manipulations affect behaviour in different contexts, and how their therapeutic effects may be exerted in humans - which may illuminate issues of translational models, hierarchical mechanisms, idiographic variables, and social cognition.
Collapse
|
27
|
Fukumoto K, Fogaça MV, Liu RJ, Duman CH, Li XY, Chaki S, Duman RS. Medial PFC AMPA receptor and BDNF signaling are required for the rapid and sustained antidepressant-like effects of 5-HT 1A receptor stimulation. Neuropsychopharmacology 2020; 45:1725-1734. [PMID: 32396921 PMCID: PMC7419563 DOI: 10.1038/s41386-020-0705-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 04/26/2020] [Accepted: 05/05/2020] [Indexed: 12/28/2022]
Abstract
We previously reported that the serotonergic system is important for the antidepressant-like effects of ketamine, a non-competitive N-methyl-D-aspartate receptor antagonist, which produces rapid and long-lasting antidepressant effects in patients with major depressive disorder (MDD). In particular, selective stimulation of the 5-HT1A receptor in the medial prefrontal cortex (mPFC), as opposed to the somatic 5-HT1A autoreceptor, has been shown to play a critical role in the antidepressant-like actions of ketamine. However, the detailed mechanisms underlying mPFC 5-HT1A receptor-mediated antidepressant-like effects are not fully understood. Here we examined the involvement of the glutamate AMPA receptor and brain-derived neurotrophic factor (BDNF) in the antidepressant-like effects of 5-HT1A receptor activation in the mPFC. The results show that intra-mPFC infusion of the 5-HT1A receptor agonist 8-OH-DPAT induces rapid and long-lasting antidepressant-like effects in the forced swim, novelty-suppressed feeding, female urine sniffing, and chronic unpredictable stress tests. In addition, the results demonstrate that the antidepressant-like effects of intra-mPFC infusion of 8-OH-DPAT are blocked by co-infusion of an AMPA receptor antagonist or an anti-BDNF neutralizing antibody. In addition, mPFC infusion of 8-OH-DPAT increased the phosphorylation of signaling proteins downstream of BDNF, including mTOR, ERK, 4EBP1, and p70S6K. Finally, selective stimulation of the 5-HT1A receptor increased levels of synaptic proteins and synaptic function in the mPFC. Collectively, these results indicate that selective stimulation of 5-HT1A receptor in the mPFC exerts rapid and sustained antidepressant-like effects via activation of AMPA receptor/BDNF/mTOR signaling in mice, which subsequently increase synaptic function in the mPFC, and provide evidence for the 5-HT1A receptor as a target for the treatment of MDD.
Collapse
Affiliation(s)
- Kenichi Fukumoto
- Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06520, USA. .,Research Headquarters, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama, 331-9530, Japan.
| | - Manoela V. Fogaça
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| | - Rong-Jian Liu
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| | - Catharine H. Duman
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| | - Xiao-Yuan Li
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| | - Shigeyuki Chaki
- grid.419836.10000 0001 2162 3360Research Headquarters, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama 331-9530 Japan
| | - Ronald S. Duman
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| |
Collapse
|
28
|
Ma L, Tian MX, Sun QY, Liu NN, Dong JF, Feng K, Wu YK, Wang YX, Wang GY, Chen W, Xi JJ, Kang JH. Fetal growth restriction mice are more likely to exhibit depression-like behaviors due to stress-induced loss of dopaminergic neurons in the VTA. FASEB J 2020; 34:13257-13271. [PMID: 32860269 DOI: 10.1096/fj.202000534r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/05/2020] [Accepted: 07/16/2020] [Indexed: 11/11/2022]
Abstract
Fetal growth restriction (FGR) is a severe perinatal complication that can increase risk for mental illness. To investigate the mechanism by which FGR mice develop mental illness in adulthood, we established the FGR mouse model and the FGR mice did not display obvious depression-like behaviors, but after environmental stress exposure, FGR mice were more likely to exhibit depression-like behaviors than control mice. Moreover, FGR mice had significantly fewer dopaminergic neurons in the ventral tegmental area but no difference in serotoninergic neurons in the dorsal raphe. RNA-seq analysis showed that the downregulated genes in the midbrain of FGR mice were associated with many mental diseases and were especially involved in the regulation of NMDA-selective glutamate receptor (NMDAR) activity. Furthermore, the NMDAR antagonist memantine can relieve the stress-induced depression-like behaviors of FGR mice. In summary, our findings provide a theoretical basis for future research and treatment of FGR-related depression.
Collapse
Affiliation(s)
- Li Ma
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meng-Xue Tian
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Institute of Translational Research, Tongji Hospital, School of Life Sciences and Technology, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| | - Qiao-Yi Sun
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Na-Na Liu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jian-Feng Dong
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ke Feng
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yu-Kang Wu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yu-Xi Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Gui-Ying Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wen Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jia-Jie Xi
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jiu-Hong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
29
|
Esquivel-Franco DC, de Boer SF, Waldinger M, Olivier B, Olivier JDA. Pharmacological Studies on the Role of 5-HT 1 A Receptors in Male Sexual Behavior of Wildtype and Serotonin Transporter Knockout Rats. Front Behav Neurosci 2020; 14:40. [PMID: 32296313 PMCID: PMC7136541 DOI: 10.3389/fnbeh.2020.00040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
Brain serotonin (5-HT) neurotransmission plays an important role in male sexual behavior and it is well established that activating 5-HT1 A receptors in rats facilitate ejaculatory behavior. However, the relative contribution of 5-HT1 A somatodendritic autoreceptors and heteroreceptors in this pro-sexual behavior is unclear. Moreover, it is unclear whether the contribution of somatodendritic 5-HT1 A autoreceptors and postsynaptic 5-HT1 A heteroreceptors alter when extracellular 5-HT levels are chronically increased. Serotonin transporter knockout (SERT-/-) rats exhibit enhanced extracellular 5-HT levels and desensitized 5-HT1 A receptors. These rats model neurochemical changes underlying chronic SSRI-induced sexual dysfunction. We want to determine the role of presynaptic versus postsynaptic 5-HT1 A receptors in the pro-sexual effects of 5-HT1 A receptor agonists in SERT+/+ and in SERT-/- rats. Therefore, acute effects of the biased 5-HT1 A receptor agonists F-13714, a preferential 5-HT1 A autoreceptor agonist, or F-15599, a preferential 5-HT1 A heteroreceptor agonist, and S15535 a mixed 5-HT1 A autoreceptor agonist/heteroreceptor antagonist, on male sexual behavior were assessed. A clear and stable genotype effect was found after training where SERT+/+ performed sexual behavior at a higher level than SERT-/- rats. Both F-15599 and F-13714 induced pro-sexual activity in SERT+/+ and SERT-/- animals. Compared to SERT+/+, the F13714-dose-response curve in SERT-/- rats was shifted to the right. SERT+/+ and SERT-/- rats responded similar to F15599. Within both SERT+/+ and SERT-/- rats the potency of F-13714 was much stronger compared to F-15599. S15535 had no effect on sexual behavior in either genotype. In SERT+/+ and SERT-/- rats that were selected on comparable low sexual activity (SERT+/+ 3 or less ejaculations and SERT-/- 5 or less ejaculations in 10 weeks) S15535 also did not influence sexual behavior. The two biased compounds with differential effects on 5-HT1 A auto- and hetero-receptors, exerted pro-sexual activity in both SERT+/+ and SERT-/- rats. Applying these specific pharmacological tools has not solved whether pre- or post-synaptic 5-HT1 A receptors are involved in pro-sexual activity. Moreover, the inactivity of S15535 in male sexual behavior in either genotype was unexpected. The question is whether the in vivo pharmacological profile of the different 5-HT1 A receptor ligands used, is sufficient to differentiate pre- and/or post-synaptic 5-HT1 A receptor contributions in male rat sexual behavior.
Collapse
Affiliation(s)
- Diana Carolina Esquivel-Franco
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands.,Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Instituto de Investigaciones Biomédicas (IIB), Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sietse F de Boer
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Marcel Waldinger
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Berend Olivier
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands.,Department of Psychopharmacology, Utrecht Institute for Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, Netherlands.,Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
| | - Jocelien D A Olivier
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| |
Collapse
|
30
|
Abstract
ABSTRACT:Cannabidiol (CBD) has been generating increasing interest in medicine due to its therapeutic properties and an apparent lack of negative side effects. Research has suggested that high dosages of CBD can be taken acutely and chronically with little to no risk. This review focuses on the neuroprotective effects of a CBD, with an emphasis on its implications for recovering from a mild traumatic brain injury (TBI) or concussion. CBD has been shown to influence the endocannabinoid system, both by affecting cannabinoid receptors and other receptors involved in the endocannabinoid system such as vanilloid receptor 1, adenosine receptors, and 5-hydroxytryptamine via cannabinoid receptor-independent mechanisms. Concussions can result in many physiological consequences, potentially resulting in post-concussion syndrome. While impairments in cerebrovascular and cardiovascular physiology following concussion have been shown, there is unfortunately still no single treatment available to enhance recovery. CBD has been shown to influence the blood brain barrier, brain-derived neurotrophic factors, cognitive capacity, the cerebrovasculature, cardiovascular physiology, and neurogenesis, all of which have been shown to be altered by concussion. CBD can therefore potentially provide treatment to enhance neuroprotection by reducing inflammation, regulating cerebral blood flow, enhancing neurogenesis, and protecting the brain against reactive oxygen species. Double-blind randomized controlled trials are still required to validate the use of CBD as medication following mild TBIs, such as concussion.
Collapse
|
31
|
Houwing DJ, de Waard J, Ramsteijn AS, Woelders T, de Boer SF, Wams EJ, Olivier JDA. Perinatal fluoxetine exposure disrupts the circadian response to a phase-shifting challenge in female rats. Psychopharmacology (Berl) 2020; 237:2555-2568. [PMID: 32533210 PMCID: PMC7351858 DOI: 10.1007/s00213-020-05556-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/11/2020] [Indexed: 12/11/2022]
Abstract
RATIONALE Selective serotonin reuptake inhibitor (SSRI) antidepressants are increasingly prescribed during pregnancy. Changes in serotonergic signaling during human fetal development have been associated with changes in brain development and with changes in affective behavior in adulthood. The suprachiasmatic nucleus (SCN) is known to be modulated by serotonin and it is therefore assumed that SSRIs may affect circadian rhythms. However, effects of perinatal SSRI treatment on circadian system functioning in the offspring are largely unknown. OBJECTIVE Our aim was to investigate the effects of perinatal exposure to the SSRI fluoxetine (FLX) on circadian behavior, affective behavior, and 5-HT1A receptor sensitivity in female rats. In addition, we studied the expression of clock genes and the 5-HT1A receptor in the SCN, as they are potentially involved in underlying mechanisms contributing to changes in circadian rhythms. RESULTS Perinatal FLX exposure shortened the free-running tau in response to the 5-HT1A/7 agonist 8-OH-DPAT. However, FLX exposure did not alter anxiety, stress coping, and 5-HT1A receptor sensitivity. No differences were found in 5-HT1A receptor and clock genes Per1, Per2, Cry1, and Cry2 SCN gene expression. CONCLUSIONS Perinatal FLX exposure altered the response to a phase-shifting challenge in female rats, whether this may pose health risks remains to be investigated.
Collapse
Affiliation(s)
- Danielle J Houwing
- Department of Neurobiology, unit Behavioral Neuroscience, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Jolien de Waard
- Department of Neurobiology, unit Behavioral Neuroscience, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Anouschka S Ramsteijn
- Department of Neurobiology, unit Behavioral Neuroscience, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Tom Woelders
- Department of Neurobiology, unit Chronobiology, GELIFES, Univ. Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Sietse F de Boer
- Department of Neurobiology, unit Behavioral Neuroscience, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Emma J Wams
- Department of Neurobiology, unit Behavioral Neuroscience, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Jocelien D A Olivier
- Department of Neurobiology, unit Behavioral Neuroscience, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands.
| |
Collapse
|
32
|
Rantala MJ, Luoto S, Krama T, Krams I. Eating Disorders: An Evolutionary Psychoneuroimmunological Approach. Front Psychol 2019; 10:2200. [PMID: 31749720 PMCID: PMC6842941 DOI: 10.3389/fpsyg.2019.02200] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/12/2019] [Indexed: 12/18/2022] Open
Abstract
Eating disorders are evolutionarily novel conditions. They lead to some of the highest mortality rates of all psychiatric disorders. Several evolutionary hypotheses have been proposed for eating disorders, but only the intrasexual competition hypothesis is extensively supported by evidence. We present the mismatch hypothesis as a necessary extension to the current theoretical framework of eating disorders. This hypothesis explains the evolutionarily novel adaptive metaproblem that has arisen when mating motives conflict with the large-scale and easy availability of hyper-rewarding but obesogenic foods. This situation is exacerbated particularly in those contemporary environments that are characterized by sedentary lifestyles, ever-present junk foods, caloric surplus and the ubiquity of social comparisons that take place via social media. Our psychoneuroimmunological model connects ultimate-level causation with proximate mechanisms by showing how the adaptive metaproblem between mating motives and food rewards leads to chronic stress and, further, to disordered eating. Chronic stress causes neuroinflammation, which increases susceptibility to OCD-like behaviors that typically co-occur with eating disorders. Chronic stress upregulates the serotonergic system and causes dysphoric mood in anorexia nervosa patients. Dieting, however, reduces serotonin levels and dysphoric mood, leading to a vicious serotonergic-homeostatic stress/starvation cycle whereby cortisol and neuroinflammation increase through stringent dieting. Our psychoneuroimmunological model indicates that between-individual and within-individual variation in eating disorders partially arises from (co)variation in gut microbiota and stress responsivity, which influence neuroinflammation and the serotonergic system. We review the advances that have been made in recent years in understanding how to best treat eating disorders, outlining directions for future clinical research. Current evidence indicates that eating disorder treatments should aim to reduce the chronic stress, neuroinflammation, stress responsivity and gut dysbiosis that fuel the disorders. Connecting ultimate causes with proximate mechanisms and treating biopsychosocial causes rather than manifest symptoms is expected to bring more effective and sophisticated long-term interventions for the millions of people who suffer from eating disorders.
Collapse
Affiliation(s)
| | - Severi Luoto
- English, Drama and Writing Studies, University of Auckland, Auckland, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
| | - Tatjana Krama
- Department of Biotechnology, Daugavpils University, Daugavpils, Latvia
| | - Indrikis Krams
- Department of Biotechnology, Daugavpils University, Daugavpils, Latvia
- Institute of Ecology and Earth Sciences, University of Tartu, Tartu, Estonia
| |
Collapse
|
33
|
Dopamine D 2L Receptor Deficiency Causes Stress Vulnerability through 5-HT 1A Receptor Dysfunction in Serotonergic Neurons. J Neurosci 2019; 39:7551-7563. [PMID: 31371425 DOI: 10.1523/jneurosci.0079-19.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/16/2019] [Accepted: 05/28/2019] [Indexed: 12/27/2022] Open
Abstract
Mental disorders are caused by genetic and environmental factors. We here show that deficiency of an isoform of dopamine D2 receptor (D2R), D2LR, causes stress vulnerability in mouse. This occurs through dysfunction of serotonin [5-hydroxytryptamine (5-HT)] 1A receptor (5-HT1AR) on serotonergic neurons in the mouse brain. Exposure to forced swim stress significantly increased anxiety- and depressive-like behaviors in D2LR knock-out (KO) male mice compared with wild-type mice. Treatment with 8-OH-DPAT, a 5-HT1AR agonist, failed to alleviate the stress-induced behaviors in D2LR-KO mice. In forced swim-stressed D2LR-KO mice, 5-HT efflux in the medial prefrontal cortex was elevated and the expression of genes related to 5-HT levels was upregulated by the transcription factor PET1 in the dorsal raphe nucleus. Notably, D2LR formed a heteromer with 5-HT1AR in serotonergic neurons, thereby suppressing 5-HT1AR-activated G-protein-activated inwardly rectifying potassium conductance in D2LR-KO serotonergic neurons. Finally, D2LR overexpression in serotonergic neurons in the dorsal raphe nucleus alleviated stress vulnerability observed in D2LR-KO mice. Together, we conclude that disruption of the negative feedback regulation by the D2LR/5-HT1A heteromer causes stress vulnerability.SIGNIFICANCE STATEMENT Etiologies of mental disorders are multifactorial, e.g., interactions between genetic and environmental factors. In this study, using a mouse model, we showed that genetic depletion of an isoform of dopamine D2 receptor, D2LR, causes stress vulnerability associated with dysfunction of serotonin 1A receptor, 5-HT1AR in serotonergic neurons. The D2LR/5-HT1AR inhibitory G-protein-coupled heteromer may function as a negative feedback regulator to suppress psychosocial stress.
Collapse
|
34
|
Sargin D, Jeoung HS, Goodfellow NM, Lambe EK. Serotonin Regulation of the Prefrontal Cortex: Cognitive Relevance and the Impact of Developmental Perturbation. ACS Chem Neurosci 2019; 10:3078-3093. [PMID: 31259523 DOI: 10.1021/acschemneuro.9b00073] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The prefrontal cortex is essential for both executive function and emotional regulation. The interrelationships among these behavioral domains are increasingly recognized, as well as their sensitivity to serotonin (5-hydroxytryptamine, 5-HT). Prefrontal cortex receives serotonergic inputs from the dorsal and median raphe nuclei and is modulated by multiple subtypes of 5-HT receptor across its layers and cell types. Extremes of serotonergic modulation alter mood regulation in vulnerable individuals, yet the impact of serotonin under more typical physiological parameters remains unclear. In this regard, new tools are permitting a closer examination of the behavioral impact of the serotonin system. Optogenetic and chemogenetic manipulations of dorsal raphe 5-HT neurons reveal that serotonin has a greater impact on executive function than previously appreciated. Domains that appear sensitive to fluctuations in 5-HT neuronal excitability include patience and cognitive flexibility. This work is broadly consistent with ex vivo research investigating how 5-HT regulates prefrontal cortex and its output projections. A growing literature suggests 5-HT modulation of these prefrontal circuits is unexpectedly flexible to alteration during development by genetic, behavioral, environmental or pharmacological manipulations, with lasting repercussions for cognition and emotional regulation. Here, we review the cellular and circuit mechanisms of prefrontal serotonergic modulation, investigate recent research into the cognitive consequences of the serotonergic system, and probe the lasting consequences of developmental perturbations. Understanding both the complexity of the prefrontal serotonin system and its sensitivity during development are essential to learn more about the vulnerabilities of this system in mood and anxiety disorders and the underappreciated cognitive consequences of these disorders and their treatment.
Collapse
Affiliation(s)
- Derya Sargin
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Ha-Seul Jeoung
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Evelyn K. Lambe
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of OBGYN, University of Toronto, Toronto, ON M5G 1E2, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
| |
Collapse
|
35
|
Albert PR, Le François B, Vahid-Ansari F. Genetic, epigenetic and posttranscriptional mechanisms for treatment of major depression: the 5-HT1A receptor gene as a paradigm. J Psychiatry Neurosci 2019; 44:164-176. [PMID: 30807072 PMCID: PMC6488484 DOI: 10.1503/jpn.180209] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/10/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023] Open
Abstract
Major depression and anxiety are highly prevalent and involve chronic dysregulation of serotonin, but they remain poorly understood. Here, we review novel transcriptional (genetic, epigenetic) and posttranscriptional (microRNA, alternative splicing) mechanisms implicated in mental illness, focusing on a key serotonin-related regulator, the serotonin 1A (5-HT1A) receptor. Functional single-nucleotide polymorphisms and stress-induced DNA methylation of the 5-HT1A promoter converge to differentially alter pre- and postsynaptic 5-HT1A receptor expression associated with major depression and reduced therapeutic response to serotonergic antidepressants. Major depression is also associated with altered levels of splice factors and microRNA, posttranscriptional mechanisms that regulate RNA stability. The human 5-HT1A 3′-untranslated region is alternatively spliced, removing microRNA sites and increasing 5-HT1A expression, which is reduced in major depression and may be genotype-dependent. Thus, the 5-HT1A receptor gene illustrates the convergence of genetic, epigenetic and posttranscriptional mechanisms in gene expression, neurodevelopment and neuroplasticity, and major depression. Understanding gene regulatory mechanisms could enhance the detection, categorization and personalized treatment of major depression.
Collapse
Affiliation(s)
- Paul R. Albert
- From the Department of Neuroscience, Ottawa Hospital Research Institute, UOttawa Brain and Mind Research Institute, Ottawa, Ont., Canada
| | - Brice Le François
- From the Department of Neuroscience, Ottawa Hospital Research Institute, UOttawa Brain and Mind Research Institute, Ottawa, Ont., Canada
| | - Faranak Vahid-Ansari
- From the Department of Neuroscience, Ottawa Hospital Research Institute, UOttawa Brain and Mind Research Institute, Ottawa, Ont., Canada
| |
Collapse
|
36
|
Vahid-Ansari F, Zhang M, Zahrai A, Albert PR. Overcoming Resistance to Selective Serotonin Reuptake Inhibitors: Targeting Serotonin, Serotonin-1A Receptors and Adult Neuroplasticity. Front Neurosci 2019; 13:404. [PMID: 31114473 PMCID: PMC6502905 DOI: 10.3389/fnins.2019.00404] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/09/2019] [Indexed: 12/14/2022] Open
Abstract
Major depressive disorder (MDD) is the most prevalent mental illness contributing to global disease burden. Selective serotonin (5-HT) reuptake inhibitors (SSRIs) are the first-line treatment for MDD, but are only fully effective in 30% of patients and require weeks before improvement may be seen. About 30% of SSRI-resistant patients may respond to augmentation or switching to another antidepressant, often selected by trial and error. Hence a better understanding of the causes of SSRI resistance is needed to provide models for optimizing treatment. Since SSRIs enhance 5-HT, in this review we discuss new findings on the circuitry, development and function of the 5-HT system in modulating behavior, and on how 5-HT neuronal activity is regulated. We focus on the 5-HT1A autoreceptor, which controls 5-HT activity, and the 5-HT1A heteroreceptor that mediates 5-HT actions. A series of mice models now implicate increased levels of 5-HT1A autoreceptors in SSRI resistance, and the requirement of hippocampal 5-HT1A heteroreceptor for neurogenic and behavioral response to SSRIs. We also present clinical data that show promise for identifying biomarkers of 5-HT activity, 5-HT1A regulation and regional changes in brain activity in MDD patients that may provide biomarkers for tailored interventions to overcome or bypass resistance to SSRI treatment. We identify a series of potential strategies including inhibiting 5-HT auto-inhibition, stimulating 5-HT1A heteroreceptors, other monoamine systems, or cortical stimulation to overcome SSRI resistance.
Collapse
Affiliation(s)
| | | | | | - Paul R. Albert
- Brain and Mind Research Institute, Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
37
|
Zhang J, He ZX, Wang LM, Yuan W, Li LF, Hou WJ, Yang Y, Guo QQ, Zhang XN, Cai WQ, An SC, Tai FD. Voluntary Wheel Running Reverses Deficits in Social Behavior Induced by Chronic Social Defeat Stress in Mice: Involvement of the Dopamine System. Front Neurosci 2019; 13:256. [PMID: 31019446 PMCID: PMC6458241 DOI: 10.3389/fnins.2019.00256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/05/2019] [Indexed: 01/04/2023] Open
Abstract
Voluntary exercise has been reported to have a therapeutic effect on many psychiatric disorders and social stress is known to impair social interaction. However, whether voluntary exercise could reverse deficits in social behaviors induced by chronic social defeat stress (CSDS) and the underlying mechanism remain unclear. The present study shows CSDS impaired social preference and induced social interaction deficiency in susceptible mice. Voluntary wheel running (VWR) reversed these effects. In addition, CSDS decreased the levels of tyrosine hydroxylase in the ventral tegmental area and the D2 receptor (D2R) in the nucleus accumbens (NAc) shell. These changes can be recovered by VWR. Furthermore, the recovery effect of VWR on deficits in social behaviors in CSDS mice was blocked by the microinjection of D2R antagonist raclopride into the NAc shell. Thus, these results suggest that the mechanism underlying CSDS-induced social interaction disorder might be caused by an alteration of the dopamine system. VWR may be a novel means to treat CSDS-induced deficits in social behaviors via modifying the dopamine system.
Collapse
Affiliation(s)
- Jing Zhang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhi-Xiong He
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Li-Min Wang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wei Yuan
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Lai-Fu Li
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wen-Juan Hou
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yang Yang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Qian-Qian Guo
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xue-Ni Zhang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wen-Qi Cai
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Shu-Cheng An
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Fa-Dao Tai
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
38
|
Wang QQ, Wang CM, Cheng BH, Yang CQ, Bai B, Chen J. Signaling transduction regulated by 5-hydroxytryptamine 1A receptor and orexin receptor 2 heterodimers. Cell Signal 2018; 54:46-58. [PMID: 30481562 DOI: 10.1016/j.cellsig.2018.11.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/08/2018] [Accepted: 11/15/2018] [Indexed: 12/09/2022]
Abstract
As G-protein-coupled receptors (GPCRs), 5-hydroxytryptamine 1A receptor (5-HT1AR) and orexin receptor 2 (OX2R) regulate the levels of the cellular downstream molecules. The heterodimers of different GPCRs play important roles in various of neurological diseases. Moreover, 5-HT1AR and OX2R are involved in the pathogenesis of neurological diseases such as depression with deficiency of hippocampus plasticity. However, the direct interaction of the two receptors remains elusive. In the present study, we firstly demonstrated the heterodimer formation of 5-HT1AR and OX2R. Exchange protein directly activated by cAMP (Epac) cAMP bioluminescence resonance energy transfer (BRET) biosensor analysis revealed that the expression levels of cellular cAMP significantly increased in HEK293T cells transfected with the two receptors compared with the 5-HT1AR group. Additionally, the cellular level of calcium was upregulated robustly in HEK293T cells co-transfected with 5-HT1AR and OX2R group after agonist treatment. Furthermore, western blotting data showed that 5-HT1AR and OX2R heterodimer decreased the levels of phosphorylation of extracellular signal-regulated kinase (ERK) and cAMP-response element-binding protein (CREB). These results not only unraveled the formation of 5-HT1AR and OX2R heterodimer but also suggested that the heterodimer affected the downstream signaling pathway, which will provide new insights into the function of the two receptors in the brain.
Collapse
Affiliation(s)
- Qin-Qin Wang
- Neurobiology Key Laboratory, Jining Medical University, Colleges of Shandong, Jining 272067, PR China
| | - Chun-Mei Wang
- Neurobiology Key Laboratory, Jining Medical University, Colleges of Shandong, Jining 272067, PR China
| | - Bao-Hua Cheng
- Neurobiology Key Laboratory, Jining Medical University, Colleges of Shandong, Jining 272067, PR China
| | - Chun-Qing Yang
- Neurobiology Key Laboratory, Jining Medical University, Colleges of Shandong, Jining 272067, PR China
| | - Bo Bai
- Neurobiology Key Laboratory, Jining Medical University, Colleges of Shandong, Jining 272067, PR China.
| | - Jing Chen
- Neurobiology Key Laboratory, Jining Medical University, Colleges of Shandong, Jining 272067, PR China; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
39
|
Wang L, Zhu Z, Hou W, Zhang X, He Z, Yuan W, Yang Y, Zhang S, Jia R, Tai F. Serotonin Signaling Trough Prelimbic 5-HT1A Receptors Modulates CSDS-Induced Behavioral Changes in Adult Female Voles. Int J Neuropsychopharmacol 2018; 22:208-220. [PMID: 30445535 PMCID: PMC6403097 DOI: 10.1093/ijnp/pyy093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/02/2018] [Accepted: 11/14/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Most previous studies have focused on the effects of social defeat in male juvenile individuals. Whether chronic social defeat stress in adulthood affects female emotion and the underlying mechanisms remains unclear. METHODS Using highly aggressive adult female mandarin voles (Microtus mandarinus), the present study aimed to determine the effects of chronic social defeat stress on anxiety- and depression-like behaviors in adult female rodents and investigate the neurobiological mechanisms underlying these effects. RESULTS Exposure of adult female voles to social defeat stress for 14 days reduced the time spent in the central area of the open field test and in the open arms of the elevated plus maze and lengthened the immobility time in the tail suspension and forced swimming tests, indicating increased anxiety- and depression-like behaviors. Meanwhile, defeated voles exhibited increased neural activity in the prelimbic cortex of the medial prefrontal cortex. Furthermore, chronic social defeat stress reduced serotonin projections and levels of serotonin 1A receptors in the medial prefrontal cortex-prelimbic cortex. Intra-prelimbic cortex microinjections of the serotonin 1A receptor agonist 8-OH-DPAT reversed the alterations in emotional behaviors, whereas injections of the serotonin 1A receptor antagonist WAY-100635 into the prelimbic cortex of control voles increased the levels of anxiety- and depression-like behaviors. CONCLUSIONS Taken together, our results demonstrated that chronic social defeat stress increased anxiety- and depression-like behaviors in adult female voles, and these effects were mediated by the action of serotonin on the serotonin 1A receptors in the prelimbic cortex. The serotonin system may be a promising target to treat emotional disorders induced by chronic social defeat stress.
Collapse
Affiliation(s)
- Limin Wang
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Zhenxiang Zhu
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Wenjuan Hou
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Xueni Zhang
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Zhixiong He
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Wei Yuan
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Yang Yang
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Siyi Zhang
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Rui Jia
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Fadao Tai
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China,Correspondence: Fadao Tai, PhD, Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi 710062, China ()
| |
Collapse
|
40
|
Żmudzka E, Sałaciak K, Sapa J, Pytka K. Serotonin receptors in depression and anxiety: Insights from animal studies. Life Sci 2018; 210:106-124. [PMID: 30144453 DOI: 10.1016/j.lfs.2018.08.050] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/13/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022]
Abstract
Serotonin regulates many physiological processes including sleep, appetite, and mood. Thus, serotonergic system is an important target in the treatment of psychiatric disorders, such as major depression and anxiety. This natural neurotransmitter interacts with 7 families of its receptors (5-HT1-7), which cause a variety of pharmacological effects. Using genetically modified animals and selective or preferential agonists and antagonist, numerous studies demonstrated the involvement of almost all serotonin receptor subtypes in antidepressant- or anxiolytic-like effects. In this review, based on animal studies, we discuss the possible involvement of serotonin receptor subtypes in depression and anxiety.
Collapse
Affiliation(s)
- Elżbieta Żmudzka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Sapa
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
41
|
Rawat A, Guo J, Renoir T, Pang TY, Hannan AJ. Hypersensitivity to sertraline in the absence of hippocampal 5-HT 1AR and 5-HTT gene expression changes following paternal corticosterone treatment. ENVIRONMENTAL EPIGENETICS 2018; 4:dvy015. [PMID: 30046455 PMCID: PMC6054191 DOI: 10.1093/eep/dvy015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/02/2018] [Accepted: 05/15/2018] [Indexed: 06/08/2023]
Abstract
The male germ line is capable of transmitting a legacy of stress exposure to the next generation of offspring. This transgenerational process manifests by altering offspring affective behaviours, cognition and metabolism. Paternal early life trauma causes hippocampal serotonergic dysregulation in male offspring. We previously showed a transgenerational modification to male offspring anxiety-like behaviours by treatment of adult male breeders with corticosterone (CORT) prior to mating. In the present study, we used offspring from our paternal CORT model and characterised offspring serotonergic function by examining their responses to the 5HT1AR agonist, 8-OH-DPAT, and the selective serotonin reuptake inhibitor, sertraline. We also examined whether post-weaning environmental enrichment, a paradigm well-known to modulate serotonergic signalling in the brain, had the capacity to normalise the anxiety phenotype of male offspring. Finally, we assessed gene expression levels of 5HT1AR and serotonin transporter in the offspring hippocampus to determine whether deficits in gene transcription contributed to the male-only anxiety phenotype. We report that male and female offspring of CORT-treated fathers are hypersensitive to sertraline but have normal hypothermic responses to 8-OH-DPAT. No deficits in htr1a and sert were found in association with paternal CORT treatment, and environmental enrichment did not rescue the anxiety phenotype of male offspring on the elevated-plus maze. These findings indicate that varying forms of paternal stress exert different effects on offspring brain serotonergic function.
Collapse
Affiliation(s)
- Arina Rawat
- Behavioural Neuroscience Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jackey Guo
- Behavioural Neuroscience Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Thibault Renoir
- Behavioural Neuroscience Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Terence Y Pang
- Behavioural Neuroscience Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Anthony J Hannan
- Behavioural Neuroscience Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
42
|
Fukumoto K, Iijima M, Funakoshi T, Chaki S. Role of 5-HT1A Receptor Stimulation in the Medial Prefrontal Cortex in the Sustained Antidepressant Effects of Ketamine. Int J Neuropsychopharmacol 2017; 21:371-381. [PMID: 29309585 PMCID: PMC5888010 DOI: 10.1093/ijnp/pyx116] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/21/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND We previously reported that serotonergic transmission plays an important role in antidepressant effects of ketamine. However, detailed mechanisms have not been elucidated. Among the serotonin receptor subtypes, the serotonin1A receptor in the medial prefrontal cortex has an important role in depression. Here, we investigated the role of the medial prefrontal cortex serotonin1A receptor and its signaling mechanism in the antidepressant effects of ketamine. METHODS The role of serotonin1A receptor-mediated signaling mechanism (phosphoinositide-3 kinase/Akt) in the medial prefrontal cortex was examined in the mouse forced swimming test and western blotting. RESULTS Ketamine exerted antidepressant effects that lasted for 24 hours, and the sustained antidepressant effects were attenuated by intra-medial prefrontal cortex injection of a serotonin1A receptor antagonist, WAY100635. The sustained antidepressant effects were mimicked by intra- medial prefrontal cortex, but not systemic, administration of a serotonin1A receptor agonist, (±)-8-hydroxy-2-dipropylaminotetralin hydrobromide (8-OH-DPAT). The sustained antidepressant effects of ketamine and 8-OH-DPAT were abrogated by intra- medial prefrontal cortex injection of a phosphoinositide-3 kinase inhibitor. Ketamine increased the phosphorylation of Akt in the medial prefrontal cortex at 60 minutes after administration, which was blocked by a serotonin1A receptor antagonist and a phosphoinositide-3 kinase inhibitor. Furthermore, the sustained antidepressant effects of ketamine and 8-OH-DPAT were attenuated by pretreatment of intra-medial prefrontal cortex injection of a mechanistic target of rapamycin complex-1 inhibitor. CONCLUSIONS These results indicate that selective stimulation of the medial prefrontal cortex serotonin1A receptor and subsequent activation of the phosphoinositide-3 kinase/Akt/mechanistic target of rapamycin complex-1 pathway may be necessary for ketamine to exert the sustained antidepressant effects, and that this mechanism could be targeted to develop a novel and effective approach for treating depression.
Collapse
Affiliation(s)
| | | | - Takeo Funakoshi
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Japan
| | - Shigeyuki Chaki
- Research Headquarters, Taisho Pharmaceutical Co., Ltd., Japan,Correspondence: Shigeyuki Chaki, PhD, Research Headquarters, Taisho Pharmaceutical Co., Ltd., 1–403 Yoshino-cho, Kita-ku, Saitama, Saitama 331–9530, Japan ()
| |
Collapse
|
43
|
Pilar-Cuéllar F, Vidal R, Díaz Á, Garro-Martínez E, Linge R, Castro E, Haberzettl R, Fink H, Bert B, Brosda J, Romero B, Crespo-Facorro B, Pazos Á. Enhanced Stress Response in 5-HT 1AR Overexpressing Mice: Altered HPA Function and Hippocampal Long-Term Potentiation. ACS Chem Neurosci 2017; 8:2393-2401. [PMID: 28777913 DOI: 10.1021/acschemneuro.7b00156] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Postsynaptic 5-HT1A receptors (5-HT1AR) play an important role in anxiety and stress, although their contribution is still controversial. Previous studies report that mice overexpressing postsynaptic 5-HT1ARs show no changes in basal anxiety, though the influence of stress conditions has not been addressed yet. In this study, we used this animal model to evaluate the role of 5-HT1ARs in anxiety response after pre-exposure to an acute stressor. Under basal conditions, 5-HT1AR overexpressing animals presented high corticosterone levels and a lower mineralocorticoid/glucocorticoid receptor ratio. After pre-exposure to a single stressor, they showed a high anxiety-like response, associated with a blunted increase in corticosterone levels and higher c-Fos activation in the prefrontal cortex. Moreover, these mice also presented a lack of downregulation of hippocampal long-term potentiation after stress exposure. Therefore, higher postsynaptic 5-HT1AR activation might predispose to a high anxious phenotype and an impaired stress coping behavior.
Collapse
Affiliation(s)
- Fuencisla Pilar-Cuéllar
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Rebeca Vidal
- Departamento
de Farmacología, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Red de Trastornos Adictivos del Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Álvaro Díaz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Emilio Garro-Martínez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Raquel Linge
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Elena Castro
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Robert Haberzettl
- Institut
für Pharmakologie und Toxikologie, Fachbereich Veterinärmedizin, Freie Universität Berlin, 14195 Berlin, Germany
| | - Heidrun Fink
- Institut
für Pharmakologie und Toxikologie, Fachbereich Veterinärmedizin, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bettina Bert
- Institut
für Pharmakologie und Toxikologie, Fachbereich Veterinärmedizin, Freie Universität Berlin, 14195 Berlin, Germany
| | - Jan Brosda
- Institut
für Pharmakologie und Toxikologie, Fachbereich Veterinärmedizin, Freie Universität Berlin, 14195 Berlin, Germany
| | - Beatriz Romero
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Benedicto Crespo-Facorro
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Hospital Universitario Marqués de Valdecilla, University of Cantabria-IDIVAL, School of Medicine, Department of Psychiatry, 39008 Santander, Spain
| | - Ángel Pazos
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| |
Collapse
|
44
|
Evidence of an inverse correlation between serotonergic activity and spreading depression propagation in the rat cortex. Brain Res 2017; 1672:29-34. [DOI: 10.1016/j.brainres.2017.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/12/2017] [Accepted: 07/14/2017] [Indexed: 11/19/2022]
|
45
|
Teissier A, Soiza-Reilly M, Gaspar P. Refining the Role of 5-HT in Postnatal Development of Brain Circuits. Front Cell Neurosci 2017; 11:139. [PMID: 28588453 PMCID: PMC5440475 DOI: 10.3389/fncel.2017.00139] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/26/2017] [Indexed: 11/30/2022] Open
Abstract
Changing serotonin (5-hydroxytryptamine, 5-HT) brain levels during critical periods in development has long-lasting effects on brain function, particularly on later anxiety/depression-related behaviors in adulthood. A large part of the known developmental effects of 5-HT occur during critical periods of postnatal life, when activity-dependent mechanisms remodel neural circuits. This was first demonstrated for the maturation of sensory brain maps in the barrel cortex and the visual system. More recently this has been extended to the 5-HT raphe circuits themselves and to limbic circuits. Recent studies overviewed here used new genetic models in mice and rats and combined physiological and structural approaches to provide new insights on the cellular and molecular mechanisms controlled by 5-HT during late stages of neural circuit maturation in the raphe projections, the somatosensory cortex and the visual system. Similar mechanisms appear to be also involved in the maturation of limbic circuits such as prefrontal circuits. The latter are of particular relevance to understand the impact of transient 5-HT dysfunction during postnatal life on psychiatric illnesses and emotional disorders in adult life.
Collapse
Affiliation(s)
- Anne Teissier
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| | - Mariano Soiza-Reilly
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| | - Patricia Gaspar
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839Paris, France.,Université Pierre et Marie CurieParis, France.,Institut du Fer à MoulinParis, France
| |
Collapse
|