1
|
Zaniewska M, Brygider S, Majcher-Maślanka I, Gawliński D, Głowacka U, Glińska S, Balcerzak Ł. The impact of voluntary wheel-running exercise on hippocampal neurogenesis and behaviours in response to nicotine cessation in rats. Psychopharmacology (Berl) 2024; 241:2585-2607. [PMID: 39463206 PMCID: PMC11569017 DOI: 10.1007/s00213-024-06705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
RATIONALE The literature indicates that nicotine exposure or its discontinuation impair adult hippocampal neurogenesis in rats, though the impact of exercise on this process remains unclear. We have previously shown that disturbances in the number of doublecortin (DCX, a marker of immature neurons)-positive (DCX+) cells in the dentate gyrus (DG) of the hippocampus during nicotine deprivation may contribute to a depression-like state in rats. OBJECTIVES This study aimed to investigate the effect of running on hippocampal neurogenesis, depression-like symptoms, and drug-seeking behaviour during nicotine deprivation. METHODS The rats were subjected to nicotine (0.03 mg/kg/inf) self-administration via an increasing schedule of reinforcement. After 21 sessions, the animals entered a 14-day abstinence phase during which they were housed in either standard home cages without wheels, cages equipped with running wheels, or cages with locked wheels. RESULTS Wheel running increased the number of Ki-67+ and DCX+ cells in the DG of both nicotine-deprived and nicotine-naive rats. Wheel-running exercise evoked an antidepressant effect on abstinence Day 14 but had no effect on nicotine-seeking behaviour on abstinence Day 15 compared to rats with locked-wheel access. CONCLUSIONS In summary, long-term wheel running positively affected the number of immature neurons in the hippocampus, which corresponded with an antidepressant response in nicotine-weaned rats. One possible mechanism underlying the positive effect of running on the affective state during nicotine cessation may be the reduction in deficits in DCX+ cells in the hippocampus.
Collapse
Affiliation(s)
- Magdalena Zaniewska
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland.
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, Kraków, 31-343, Poland.
- Affective Cognitive Neuroscience Laboratory, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland.
| | - Sabina Brygider
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, Kraków, 31-343, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| | - Dawid Gawliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, Kraków, 31-343, Poland
| | - Urszula Głowacka
- Department of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, Kraków, 31- 531, Poland
| | - Sława Glińska
- Faculty of Biology and Environmental Protection, Laboratory of Microscopic Imaging and Specialized Biological Techniques, University of Lodz, Banacha 12/16, Lodz, 90-237, Poland
| | - Łucja Balcerzak
- Faculty of Biology and Environmental Protection, Laboratory of Microscopic Imaging and Specialized Biological Techniques, University of Lodz, Banacha 12/16, Lodz, 90-237, Poland
| |
Collapse
|
2
|
Zhang Y, Wang S, Hei M. Maternal separation as early-life stress: Mechanisms of neuropsychiatric disorders and inspiration for neonatal care. Brain Res Bull 2024; 217:111058. [PMID: 39197670 DOI: 10.1016/j.brainresbull.2024.111058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/01/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
The establishment of positive early parent-infant relationships provide essential nourishment and social stimulation for newborns. During the early stages of postnatal brain development, events such as synaptogenesis, neuronal maturation and glial differentiation occur in a highly coordinated manner. Maternal separation, as an early-life stress introducer, can disrupt the formation of parent-child bonds and exert long-term adverse effects throughout life. When offspring are exposed to maternal separation, the body regulates the stress of maternal separation through multiple mechanisms, including neuroinflammatory responses, neuroendocrinology, and neuronal electrical activity. In adulthood, early maternal separation has long-term effects, such as the induction of neuropsychiatric disorders such as anxiety, depression, and cognitive dysfunction. This review summarized the application of maternal separation models and the mechanisms of stress system response in neuropsychiatric disorders, serving as both a reminder and inspiration for approaches to improve neonatal care, "from bench to bedside".
Collapse
Affiliation(s)
- Yuan Zhang
- Neonatal Center, Beijing Children's Hospital Capital Medical University, National Center of Children's Health, Beijing 100045, China
| | - Shu Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Mingyan Hei
- Neonatal Center, Beijing Children's Hospital Capital Medical University, National Center of Children's Health, Beijing 100045, China.
| |
Collapse
|
3
|
Alves J, Dos Santos APB, Vieira ADS, Martini APR, de Lima RMS, Smaniotto TÂ, de Moraes RO, Gomes RF, Acerbi GCDA, de Assis EZB, Lampert C, Dalmaz C, Couto Pereira NDS. Coping with the experience of frustration throughout life: Sex- and age-specific effects of early life stress on the susceptibility to reward devaluation. Neuroscience 2024; 553:160-171. [PMID: 38960089 DOI: 10.1016/j.neuroscience.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024]
Abstract
Early life stress may lead to lifelong impairments in psychophysiological functions, including emotional and reward systems. Unpredicted decrease in reward magnitude generates a negative emotional state (frustration) that may be involved with susceptibility to psychiatric disorders. We evaluated, in adolescents and adult rats of both sexes, whether maternal separation (MS) alters the ability to cope with an unexpected reduction of reward later in life. Litters of Wistar rats were divided into controls (non handled - NH) or subjected to MS. Animals were trained to find sugary cereal pellets; later the amount was reduced. Increased latency to reach the reward-associated area indicates higher inability to regulate frustration. The dorsal hippocampus (dHC) and basolateral amygdala (BLA) were evaluated for protein levels of NMDA receptor subunits (GluN2A/GluN2B), synaptophysin, PSD95, SNAP-25 and CRF1. We found that adult MS males had greater vulnerability to reward reduction, together with decreased GluN2A and increased GluN2B immunocontent in the dHC. MS females and adolescents did not differ from controls. We concluded that MS enhances the response to frustration in adult males. The change in the ratio of GluN2A and GluN2B subunits in dHC could be related to a stronger, more difficult to update memory of the aversive experience.
Collapse
Affiliation(s)
- Joelma Alves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Paula Bosquetti Dos Santos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Aline Dos Santos Vieira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Paula Rodrigues Martini
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Randriely Merscher Sobreira de Lima
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Thiago Ângelo Smaniotto
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Rafael Oliveira de Moraes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Roger Ferreira Gomes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Giulia Conde de Albite Acerbi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Eduardo Z B de Assis
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carine Lampert
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carla Dalmaz
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Natividade de Sá Couto Pereira
- Psychological Neuroscience Laboratory, Psychology Research Centre (CIPsi), School of Psychology, University of Minho, Braga, Portugal.
| |
Collapse
|
4
|
Smith AM, Grayson BE. A strike to the head: Parallels between the pediatric and adult human and the rodent in traumatic brain injury. J Neurosci Res 2024; 102:e25364. [PMID: 38953607 DOI: 10.1002/jnr.25364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024]
Abstract
Traumatic brain injury (TBI) is a condition that occurs commonly in children from infancy through adolescence and is a global health concern. Pediatric TBI presents with a bimodal age distribution, with very young children (0-4 years) and adolescents (15-19 years) more commonly injured. Because children's brains are still developing, there is increased vulnerability to the effects of head trauma, which results in entirely different patterns of injury than in adults. Pediatric TBI has a profound and lasting impact on a child's development and quality of life, resulting in long-lasting consequences to physical, cognitive, and emotional development. Chronic issues like learning disabilities, behavioral problems, and emotional disturbances can develop. Early intervention and ongoing support are critical for minimizing these long-term deficits. Many animal models of TBI exist, and each varies significantly, displaying different characteristics of clinical TBI. The neurodevelopment differs in the rodent from the human in timing and effect, so TBI outcomes in the juvenile rodent can thus vary from the human child. The current review compares findings from preclinical TBI work in juvenile and adult rodents to clinical TBI research in pediatric and adult humans. We focus on the four brain regions most affected by TBI: the prefrontal cortex, corpus callosum, hippocampus, and hypothalamus. Each has its unique developmental projections and thus is impacted by TBI differently. This review aims to compare the healthy neurodevelopment of these four brain regions in humans to the developmental processes in rodents.
Collapse
Affiliation(s)
- Allie M Smith
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Bernadette E Grayson
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Population Health Science, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
5
|
Francis-Oliveira J, Higa GSV, Viana FJC, Cruvinel E, Carlos-Lima E, da Silva Borges F, Zampieri TT, Rebello FP, Ulrich H, De Pasquale R. TREK-1 inhibition promotes synaptic plasticity in the prelimbic cortex. Exp Neurol 2024; 373:114652. [PMID: 38103709 DOI: 10.1016/j.expneurol.2023.114652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/28/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Synaptic plasticity is one of the putative mechanisms involved in the maturation of the prefrontal cortex (PFC) during postnatal development. Early life stress (ELS) affects the shaping of cortical circuitries through impairment of synaptic plasticity supporting the onset of mood disorders. Growing evidence suggests that dysfunctional postnatal maturation of the prelimbic division (PL) of the PFC might be related to the emergence of depression. The potassium channel TREK-1 has attracted particular interest among many factors that modulate plasticity, concerning synaptic modifications that could underlie mood disorders. Studies have found that ablation of TREK-1 increases the resilience to depression, while rats exposed to ELS exhibit higher TREK-1 levels in the PL. TREK-1 is regulated by multiple intracellular transduction pathways including the ones activated by metabotropic receptors. In the hippocampal neurons, TREK-1 interacts with the serotonergic system, one of the main factors involved in the action of antidepressants. To investigate possible mechanisms related to the antidepressant role of TREK-1, we used brain slice electrophysiology to evaluate the effects of TREK-1 pharmacological blockade on synaptic plasticity at PL circuitry. We extended this investigation to animals subjected to ELS. Our findings suggest that in non-stressed animals, TREK-1 activity is required for the reduction of synaptic responses mediated by the 5HT1A receptor activation. Furthermore, we demonstrate that TREK-1 blockade promotes activity-dependent long-term depression (LTD) when acting in synergy with 5HT1A receptor stimulation. On the other hand, in ELS animals, TREK-1 blockade reduces synaptic transmission and facilitates LTD expression. These results indicate that TREK-1 inhibition stimulates synaptic plasticity in the PL and this effect is more pronounced in animals subjected to ELS during postnatal development.
Collapse
Affiliation(s)
- José Francis-Oliveira
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil; Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, SP 05508-900, Brazil; Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, SP 09210-580, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Estevão Carlos-Lima
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Fernando da Silva Borges
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Thais Tessari Zampieri
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Fernanda Pereira Rebello
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, SP 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil.
| |
Collapse
|
6
|
Gildawie KR, Wang K, Budge KE, Byrnes EM. Effects of Maternal Separation on Effort-based Responding for Sucrose Are Associated with c-Fos Expression in the Nucleus Accumbens Core. Neuroscience 2024; 537:174-188. [PMID: 38036058 PMCID: PMC10872495 DOI: 10.1016/j.neuroscience.2023.11.030] [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: 08/04/2023] [Revised: 11/06/2023] [Accepted: 11/26/2023] [Indexed: 12/02/2023]
Abstract
In both people and animals, exposure to adverse experiences early in life can alter neurodevelopment and lead to long-term behavioral effects, including effects on reward processing. In the current study, we use a well-validated rodent model of maternal neglect, maternal separation (MS), to investigate the impact of early life adversity on reward learning and motivation and identify associated modifications in cellular activation in reward-relevant areas. Litters of Long-Evans rats were separated from the dam for either 15 min (brief) or 180 min (prolonged)/day from postnatal day (PND)2 to PND14. As adults, offspring were trained to lever press for a sucrose pellet using fixed ratio (FR) schedules and motivation was tested using a progressive ratio (PR) schedule over 10 daily sessions to assess sustained effects on effort-based responding. Immunohistochemical staining for c-Fos was conducted in a subset of animals that underwent an additional PR session. While there were no effects on reward learning, both MS180 males and females demonstrated increased effort-based responding on the first day of PR testing, while only MS180 males demonstrated a sustained increase in effort across all 10 days. MS180-induced changes in c-Fos expression in the dorsal and ventral striatum were observed, with subregion-specific effects along the rostrocaudal axis. Moreover, regression analyses suggest that motivated responding for a sucrose food reward in MS180-exposed, but not MS15-exposed animals, was associated with increased c-Fos expression in the rostral nucleus accumbens core. These findings implicate specific striatal regions in sex-specific modulation of sustained effort-based reward behavior following early life adversity.
Collapse
Affiliation(s)
- Kelsea R Gildawie
- Department of Comparative Pathobiology, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA 01536, USA
| | - Katherine Wang
- Department of Comparative Pathobiology, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA 01536, USA
| | - Kerri E Budge
- Department of Comparative Pathobiology, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA 01536, USA
| | - Elizabeth M Byrnes
- Department of Comparative Pathobiology, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA 01536, USA.
| |
Collapse
|
7
|
Ayala-Rodríguez JD, García-Colunga J. Maternal separation modifies spontaneous synaptic activity in the infralimbic cortex of stress-resilient male rats. PLoS One 2023; 18:e0294151. [PMID: 37943747 PMCID: PMC10635473 DOI: 10.1371/journal.pone.0294151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Glutamate and GABA signaling systems are necessary to maintain proper function of the central nervous system through excitation/inhibition (E/I) balance. Alteration of this balance in the medial prefrontal cortex (mPFC), as an effect of early-life stress, may lead to the development of anxiety and depressive disorders. Few studies exist in the infralimbic division of the mPFC to understand the effect of early-life stress at different ages, which is the purpose of the present work. Newborn Sprague Dawley male rats were subjected to maternal separation (MS) for two weeks. First, tests measuring anxiety- and depression-like behaviors were performed on adolescent and adult rats subjected to MS (MS-rats). Then, to establish a relationship with behavioral results, electrophysiological recordings were performed in neurons of the infralimbic cortex in acute brain slices of infant, adolescent, and adult rats. In the behavioral tests, there were no significant differences in MS-rats compared to control rats at any age. Moreover, MS had no effect on the passive membrane properties nor neuronal excitability in the infralimbic cortex, whereas spontaneous synaptic activity in infralimbic neurons was altered. The frequency of spontaneous glutamatergic synaptic events increased in infant MS-rats, whereas in adolescent MS-rats both the frequency and the amplitude of spontaneous GABAergic events increased without any effect on glutamatergic synaptic responses. In adult MS-rats, these two parameters decreased in spontaneous GABAergic synaptic events, whereas only the frequency of glutamatergic events decreased. These data suggest that rats subjected to MS did not exhibit behavioral changes and presented an age-dependent E/I imbalance in the infralimbic cortex, possibly due to differential changes in neurotransmitter release and/or receptor expression.
Collapse
Affiliation(s)
- Jesús David Ayala-Rodríguez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Jesús García-Colunga
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| |
Collapse
|
8
|
Edwards PT, Soni KG, Conner ME, Fowler SW, Foong JPP, Stavely R, Cheng LS, Preidis GA. Site-specific pathophysiology in a neonatal mouse model of gastroparesis. Neurogastroenterol Motil 2023; 35:e14676. [PMID: 37772676 PMCID: PMC11023621 DOI: 10.1111/nmo.14676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 07/30/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023]
Abstract
BACKGROUND Early-life events impact maturation of the gut microbiome, enteric nervous system, and gastrointestinal motility. We examined three regions of gastric tissue to determine how maternal separation and gut microbes influence the structure and motor function of specific regions of the neonatal mouse stomach. METHODS Germ-free and conventionally housed C57BL/6J mouse pups underwent timed maternal separation (TmSep) or nursed uninterrupted (controls) until 14 days of life. We assessed gastric emptying by quantifying the progression of gavaged fluorescein isothiocyanate (FITC)-dextran. With isolated rings of forestomach, corpus, and antrum, we measured tone and contractility by force transduction, gastric wall thickness by light microscopy, and myenteric plexus neurochemistry by whole-mount immunostaining. KEY RESULTS Regional gastric sampling revealed site-specific differences in contractile patterns and myenteric plexus structure. In neonatal mice, TmSep prolonged gastric emptying. In the forestomach, TmSep increased contractile responses to carbachol, decreased muscularis externa and mucosa thickness, and increased the relative proportion of myenteric plexus nNOS+ neurons. Germ-free conditions did not appreciably alter the structure or function of the neonatal mouse stomach and did not impact the changes caused by TmSep. CONCLUSIONS AND INFERENCES A regional sampling approach facilitates site-specific investigations of murine gastric motor physiology and histology to identify site-specific alterations that may impact gastrointestinal function. Delayed gastric emptying in TmSep is associated with a thinner muscle wall, exaggerated cholinergic contractile responses, and increased proportions of inhibitory myenteric plexus nNOS+ neurons in the forestomach. Gut microbes do not profoundly affect the development of the neonatal mouse stomach or the gastric pathophysiology that results from TmSep.
Collapse
Affiliation(s)
- Price T. Edwards
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Krishnakant G. Soni
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Margaret E. Conner
- Molecular Virology and Microbiology, Department of Education, Innovation and Technology, Baylor College of Medicine, Houston, TX, USA
| | - Stephanie W. Fowler
- Molecular Virology and Microbiology, Department of Education, Innovation and Technology, Baylor College of Medicine, Houston, TX, USA
| | - Jaime P. P. Foong
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Vic., Australia
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lily S. Cheng
- Michael E. DeBakey Department of Surgery, Division of Pediatric Surgery, Texas Children’s Surgical Oncology Program, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
9
|
Walker SL, Sud N, Beyene R, Palin N, Glasper ER. Paternal deprivation induces vigilance-avoidant behavior and accompanies sex-specific alterations in stress reactivity and central proinflammatory cytokine response in California mice (Peromyscus californicus). Psychopharmacology (Berl) 2023; 240:2317-2334. [PMID: 36988696 PMCID: PMC10599166 DOI: 10.1007/s00213-023-06354-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023]
Abstract
RATIONALE Early-life stress (ELS) can increase anxiety, reduce prosocial behaviors, and impair brain regions that facilitate emotional and social development. This knowledge greatly stems from assessing disrupted mother-child relationships, while studies investigating the long-term effects of father-child relationships on behavioral development in children are scarce. However, available evidence suggests that fathers may uniquely influence a child's behavioral development in a sex-specific manner. Rodent models examining mother-offspring interaction demonstrate relationships among ELS, neuroinflammatory mediators, and behavioral development; yet, the role paternal care may play in neuroimmune functioning remains unreported. OBJECTIVES Using the biparental California mouse (Peromyscus californicus), we examined to what extent paternal deprivation impairs social and anxiety-like behaviors, augments peripheral corticosterone (CORT) response, and alters central proinflammatory cytokine production following an acute stressor in adulthood. METHODS Biparentally reared and paternally deprived (permanent removal of the sire 24 h post-birth) adult mice were assessed for sociability, preference for social novelty, social vigilance, and social avoidance behaviors, followed by novelty-suppressed feeding (NSF) testing for general anxiety-like behavior. Following an acute stressor, circulating CORT concentrations and region-specific proinflammatory cytokine concentrations were determined via radioimmunoassay and Luminex multianalyte analysis, respectively. RESULTS In response to a novel same-sex conspecific, social vigilance behavior was associated with reduced sociability and increased avoidance in paternally deprived mice-an effect not observed in biparentally reared counterparts. Yet, in response to a familiar same-sex conspecific, social vigilance persisted but only in paternally deprived females. The latency to consume during NSF testing was not significantly altered by paternal deprivation. In response to an acute physical stressor, lower circulating CORT concentrations were observed in paternally deprived females. Compared to control-reared males, paternal deprivation increased hypothalamic interleukin-1β, but decreased hippocampal IL-6 protein concentration. CONCLUSION Greater social vigilance behavior was demonstrated in paternally deprived mice while they avoided social interaction with a novel same-sex conspecific; however, in response to a familiar same-sex conspecific, paternal deprivation increased social vigilance behavior but only in females. It is possible that different neurobiological mechanisms underlie these observed behavioral outcomes as sex-specific central proinflammatory cytokine and stress responsivity were observed in paternally deprived offspring.
Collapse
Affiliation(s)
- Shakeera L Walker
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
- Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD, 20742, USA
| | - Neilesh Sud
- Department of Psychology, University of Maryland, College Park, MD, 20742, USA
| | - Rita Beyene
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Nicole Palin
- Department of Psychology, University of Maryland, College Park, MD, 20742, USA
| | - Erica R Glasper
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA.
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, 43210, USA.
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, 43210, USA.
- Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD, 20742, USA.
- Department of Psychology, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
10
|
Li Y, Shi DD, Wang Z. Adolescent nonpharmacological interventions for early-life stress and their mechanisms. Behav Brain Res 2023; 452:114580. [PMID: 37453516 DOI: 10.1016/j.bbr.2023.114580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Those with a negative experience of psychosocial stress during the early stage of life not only have a high susceptibility of the psychiatric disorder in all phases of their life span, but they also demonstrate more severe symptoms and poorer response to treatment compared to those without a history of early-life stress. The interventions targeted to early-life stress may improve the effectiveness of treating and preventing psychiatric disorders. Brain regions associated with mood and cognition develop rapidly and own heightened plasticity during adolescence. So, manipulating nonpharmacological interventions in fewer side effects and higher acceptance during adolescence, which is a probable window of opportunity, may ameliorate or even reverse the constantly deteriorating impact of early-life stress. The present article reviews animal and people studies about adolescent nonpharmacological interventions for early-life stress. We aim to discuss whether those adolescent nonpharmacological interventions can promote individuals' psychological health who expose to early-life stress.
Collapse
Affiliation(s)
- Yi Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong-Dong Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Psychological and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
11
|
Meng X, Bao B, Yue G. Global research trends on maternal separation paradigms as an early life stress model: A bibliometric analysis. Heliyon 2023; 9:e18469. [PMID: 37533990 PMCID: PMC10392086 DOI: 10.1016/j.heliyon.2023.e18469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 08/04/2023] Open
Abstract
Background Maternal separation (MS) is an early life stress model that is often studied to determine how early life stress affects brain development and psychopathological adaptation. As society has developed, public health problems have become increasingly prominent, and this research area has attracted significant attention. However, to date, there has been no systematic bibliometric study on MS. The aim of this study was to analyze the trends and frontiers in MS using bibliometrics and provide a scientific reference to researchers in the field. Methods Utilizing VOSviewer, CiteSpace, and Microsoft Excel, examined data obtained from the WoSCC, which encompasses the years 2002-2021. Results In this bibliometric study, we analyzed 6209 articles related to MS authored by 24,174 researchers across 121 countries and regions and published in 2219 journals. The United States had the most publications (2,232, 35.95%) and both the United States and the United Kingdom had the highest h-index. Institutions in the United States and France had the most published articles and citations. Keyword clustering analysis revealed associations between MS and adverse early life experiences, the hypothalamic-pituitary-adrenal (HPA) axis, stress, gene expression, and depression. Conclusions This bibliometric analysis highlights the current research focus on the long-term effects of MS on emotional cognition, the HPA axis, epigenetic changes, and their links to gut microbiome imbalances. Future research may expand on these findings to investigate the underlying mechanisms and broader health and societal implications of MS. These results provide a comprehensive overview of the current research landscape in MS and offer valuable insights for researchers to guide future investigations in this field.
Collapse
Affiliation(s)
- Xiaoying Meng
- Institute of Basic Theory for Chinese Medicine,China Academy of Chinese Medical Sciences, Beijing, China
| | - Binghao Bao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Guangxin Yue
- Institute of Basic Theory for Chinese Medicine,China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Modulation of the endoplasmic reticulum stress and unfolded protein response mitigates the behavioral effects of early-life stress. Pharmacol Rep 2023; 75:293-319. [PMID: 36843201 PMCID: PMC10060333 DOI: 10.1007/s43440-023-00456-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/28/2023]
Abstract
BACKGROUND Early-life stress (ELS) affects brain development and increases the risk of mental disorders associated with the dysfunction of the medial prefrontal cortex (mPFC). The mechanisms of ELS action are not well understood. Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) are cellular processes involved in brain maturation through the regulation of pro-survival or proapoptotic processes. We hypothesized that ER stress and the UPR in the mPFC are involved in the neurobiology of ELS. METHODS We performed a maternal separation (MS) procedure from postnatal days 1 to 14 in rats. Before each MS, pups were injected with an inhibitor of ER stress, salubrinal or a vehicle. The mRNA and protein expression of UPR and apoptotic markers were evaluated in the mPFC using RT-qPCR and Western blot methods, respectively. We also estimated the numbers of neurons and glial cells using stereological methods. Additionally, we assessed behavioral phenotypes related to fear, anhedonia and response to psychostimulants. RESULTS MS slightly enhanced the activation of the UPR in juveniles and modulated the expression of apoptotic markers in juveniles and preadolescents but not in adults. Additionally, MS did not affect the numbers of neurons and glial cells at any age. Both salubrinal and vehicle blunted the expression of UPR markers in juvenile and preadolescent MS rats, often in a treatment-specific manner. Moreover, salubrinal and vehicle generally alleviated the behavioral effects of MS in preadolescent and adult rats. CONCLUSIONS Modulation of ER stress and UPR processes may potentially underlie susceptibility or resilience to ELS.
Collapse
|
13
|
Solarz A, Majcher-Maślanka I, Kryst J, Chocyk A. Early-life stress affects peripheral, blood-brain barrier, and brain responses to immune challenge in juvenile and adult rats. Brain Behav Immun 2023; 108:1-15. [PMID: 36400335 DOI: 10.1016/j.bbi.2022.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/21/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022] Open
Abstract
Early-life stress (ELS) may affect brain maturation and neuroimmune interactions and, consequently, the inflammatory response to subsequent environmental factors later in life. Recently, the coexistence of blood-brain barrier (BBB) dysfunction and inflammation has been implicated in the etiology and progression of mental and/or neurodegenerative diseases. There are sex differences in the prevalence and outcomes of these disorders. The number of studies reporting the effects of ELS and sex on BBB functioning and neuroinflammatory processes in response to immune challenge is very limited, and the data are inconsistent. In the present study, we examined whether ELS, based on the maternal separation (MS) paradigm in rats, can condition male and female subjects to subsequent lipopolysaccharide (LPS)-induced immune challenge in juvenility or adulthood. Twenty-four hours after acute LPS injection, serum proinflammatory cytokines were measured, and BBB permeability in the medial prefrontal cortex (mPFC) and hippocampus (HP) was evaluated. Additionally, the mRNA expression of neuroinflammatory markers and BBB-related genes was also studied. We found that a single LPS challenge induced a proinflammatory response both in the periphery and in the mPFC and HP and increased BBB permeability in a sex-dependent fashion. Moreover, MS enhanced the neuroinflammatory response to LPS challenge in males (especially juveniles), whereas MS females showed no difference or a blunted central response to LPS compared with control females, mainly during adulthood. These results suggest that ELS may precondition individuals to subsequent environmental factors later in life in a sex-specific manner and potentially determine their susceptibility or resilience to mental and/or neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna Solarz
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland
| | - Joanna Kryst
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland; Department of Chemistry and Biochemistry, Institute for Basics Sciences, Faculty of Physiotherapy, University of Physical Education, Jana Pawła II Av. 78, 31-571 Kraków, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Smętna Street 12, Poland.
| |
Collapse
|
14
|
Drzewiecki CM, Brinks AS, Sellinger EP, Doshi AD, Koh JY, Juraska JM. Brief postnatal exposure to bisphenol A affects apoptosis and gene expression in the medial prefrontal cortex and social behavior in rats with sex specificity. Neurotoxicology 2023; 94:126-134. [PMID: 36442689 PMCID: PMC9839503 DOI: 10.1016/j.neuro.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Bisphenol A (BPA) is an endocrine disruptor found in polycarbonate plastics and exposure in humans is nearly ubiquitous and it has widespread effects on cognitive, emotional, and reproductive behaviors in both humans and animal models. In our laboratory we previously found that perinatal BPA exposure results in a higher number of neurons in the adult male rat prefrontal cortex (PFC) and less play in adolescents of both sexes. Here we examine changes in the rate of postnatal apoptosis in the rat prefrontal cortex and its timing with brief BPA exposure. Because an increased number of neurons in the PFC is a characteristic of a subtype of autism spectrum disorder, we tested social preference following brief BPA exposure and also expression of a small group of genes. Males and females were exposed to BPA from postnatal days (P) 6 through 8 or from P10 through 12. Both exposures significantly decreased indicators of cell death in the developing medial prefrontal cortex in male subjects only. Additionally, males exposed to BPA from P6 - 8 showed decreased social preference and decreased cortical expression of Shank3 and Homer1, two synaptic scaffolding genes that have been implicated in social deficits. There were no significant effects of BPA in the female subjects. These results draw attention to the negative consequences following brief exposure to BPA during early development.
Collapse
Affiliation(s)
- Carly M Drzewiecki
- Program in Neuroscience, University of Illinois at Urbana-Champaign, Champaign, IL 61820, USA; Currently at California National Primate Research Center, University of California-Davis, Davis, CA, 95616, USA
| | - Amara S Brinks
- Program in Neuroscience, University of Illinois at Urbana-Champaign, Champaign, IL 61820, USA
| | - Elli P Sellinger
- Program in Neuroscience, University of Illinois at Urbana-Champaign, Champaign, IL 61820, USA
| | - Aditi D Doshi
- Department of Psychology, University of Illinois at Urbana-Champaign, 603 E Daniel St, Champaign, IL, 61820, USA; Currently at Department of Psychology, University of Illinois at Chicago, 1007W Harrison St, Chicago, IL 60607, USA
| | - Jessie Y Koh
- Department of Psychology, University of Illinois at Urbana-Champaign, 603 E Daniel St, Champaign, IL, 61820, USA
| | - Janice M Juraska
- Program in Neuroscience, University of Illinois at Urbana-Champaign, Champaign, IL 61820, USA; Department of Psychology, University of Illinois at Urbana-Champaign, 603 E Daniel St, Champaign, IL, 61820, USA.
| |
Collapse
|
15
|
5-HT-dependent synaptic plasticity of the prefrontal cortex in postnatal development. Sci Rep 2022; 12:21015. [PMID: 36470912 PMCID: PMC9723183 DOI: 10.1038/s41598-022-23767-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022] Open
Abstract
Important functions of the prefrontal cortex (PFC) are established during early life, when neurons exhibit enhanced synaptic plasticity and synaptogenesis. This developmental stage drives the organization of cortical connectivity, responsible for establishing behavioral patterns. Serotonin (5-HT) emerges among the most significant factors that modulate brain activity during postnatal development. In the PFC, activated 5-HT receptors modify neuronal excitability and interact with intracellular signaling involved in synaptic modifications, thus suggesting that 5-HT might participate in early postnatal plasticity. To test this hypothesis, we employed intracellular electrophysiological recordings of PFC layer 5 neurons to study the modulatory effects of 5-HT on plasticity induced by theta-burst stimulation (TBS) in two postnatal periods of rats. Our results indicate that 5-HT is essential for TBS to result in synaptic changes during the third postnatal week, but not later. TBS coupled with 5-HT2A or 5-HT1A and 5-HT7 receptors stimulation leads to long-term depression (LTD). On the other hand, TBS and synergic activation of 5-HT1A, 5-HT2A, and 5-HT7 receptors lead to long-term potentiation (LTP). Finally, we also show that 5-HT dependent synaptic plasticity of the PFC is impaired in animals that are exposed to early-life chronic stress.
Collapse
|
16
|
Sex-specific effects of neonatal paternal deprivation on microglial cell density in adult California mouse (Peromyscus californicus) dentate gyrus. Brain Behav Immun 2022; 106:1-10. [PMID: 35908654 DOI: 10.1016/j.bbi.2022.07.161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
Adverse early-life experiences are risk factors for psychiatric disease development, resulting in stress-related neuronal modeling and neurobehavioral changes. Stressful experiences modulate the immune system, contributing to neuronal damage in higher cortical regions, like the hippocampus. Moreover, early-life stressors dysregulate the function of microglia, the resident immune cells of the brain, in the developing hippocampus. Paternal deprivation, an early-life stressor in many biparental species, facilitates sex-dependent inhibitions in hippocampal plasticity, but parental contributors to these sex-specific outcomes are unknown. Also, neurobiological mechanisms contributing to impairments in hippocampal neuroplasticity are less known. Thus, our goals were to 1) determine whether parental behavior is altered in maternal females following removal of the paternal male, 2) assess the effects of paternal deprivation on dentate gyrus (DG) volume and microglia proliferation, and 3) determine if early-life experimental handling mitigates sex-specific reductions in DG cell survival. California mice were born to multiparous breeders and reared by both parents (biparental care) or by their mother alone (i.e., father removed on postnatal day 1; paternal deprivation). One cohort of offspring underwent offspring retrieval tests for eight days beginning on postnatal day 2. On PND 68, these offspring (and a second cohort of mice without behavioral testing) were euthanized and brains visualized for bromodeoxyuridine (BrdU) and neuron-specific class III beta-tubulin (TuJ-1) or ionized calcium binding adaptor molecule 1 (Iba1). While mate absence did not impair maternal retrieval, paternal deprivation reduced DG volume, but Iba1+ cell density was only higher in paternally-deprived females. Neither sex or paternal deprivation significantly altered the number of BrdU+ or Tuj1+ cells in the DG - an absence of a reduction in cell survival may be related to daily handing during early offspring retrieval tests. Together, these data suggest that paternal deprivation impairs hippocampal plasticity; however, sex and early environment may influence the magnitude of these outcomes.
Collapse
|
17
|
Effects of early life stress on brain cytokines: A systematic review and meta-analysis of rodent studies. Neurosci Biobehav Rev 2022; 139:104746. [PMID: 35716876 DOI: 10.1016/j.neubiorev.2022.104746] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/13/2022] [Accepted: 06/11/2022] [Indexed: 12/21/2022]
Abstract
Exposure to early life stress (ELS) may lead to long-lasting neurobiological and behavioral impairments. Alterations in the immune system and neuroinflammatory state induced by ELS exposure are considered risk factors for developing psychiatric disorders. Here, we performed a systematic review and meta-analysis of rodent studies investigating the short and long-term effects of ELS exposure on anti and pro-inflammatory cytokines in brain tissues. Our analysis shows that animals exposed to ELS present an increase in pro-inflammatory cytokines IL-1β, IL-6, and TNF-α. On the other hand, no alteration was observed in the anti-inflammatory cytokine IL-10. Meta-regression revealed that alterations were more prominent in the hippocampus of adult animals that were exposed to more extended periods of ELS. These inflammatory effects were not permanent since few alterations were identified in aged animals. Our findings suggest that ELS exposure alters pro-inflammatory cytokines expression and may act as a primer for a secondary challenge that may induce lifelong immune alterations. Moreover, the actual evidence is insufficient to comprehend the relationship between anti-inflammatory cytokines and ELS fully.
Collapse
|
18
|
Poleksic J, Aksic M, Kapor S, Aleksic D, Stojkovic T, Radovic M, Djulejic V, Markovic B, Stamatakis A. Effects of Maternal Deprivation on the Prefrontal Cortex of Male Rats: Cellular, Neurochemical, and Behavioral Outcomes. Front Behav Neurosci 2021; 15:666547. [PMID: 34819843 PMCID: PMC8606589 DOI: 10.3389/fnbeh.2021.666547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/08/2021] [Indexed: 01/13/2023] Open
Abstract
Stressful events experienced during early life are associated with increased vulnerability of developing psychopathology in adulthood. In the present study, we exposed 9-day-old Wistar rats to 24 h maternal deprivation (MD) with the aim to investigate the impact of early life stress (ELS) on morphological, biochemical, and functional aspects of the prefrontal cortex (PFC), a brain region particularly sensitive to stress. We found that in the superficial medial orbital cortex (MO), young adult male rats had reduced density of GAD67 and CCK immunopositive cells, while the rostral part of the ventral lateral orbital cortex (roVLO) showed a decrease in the density of GAD67 immunopositive cells in both superficial and deep layers. In addition, the superficial rostral part of area 1 of the cingulate cortex (roCg1) and deep prelimbic cortex (PrL) was also affected by MD indicated by the reduction in PV immunopositive cellular density. Furthermore, MD induced upregulation of brain-derived neurotrophic factor (BDNF), while it did not affect the overall expression of Iba1 in neonatal or young adult PFC as measured by Western blot, however, microglial activation in young adult MD rats was detected immunohistochemically in deep layers of MO and infralimbic cortex (IL). Interestingly, when young adult male rats were subjected to a behavioral flexibility test in a T-maze, MD rats showed a subtle impairment in T-maze reversal learning indicating a mildly affected PFC function. Taken together, our findings demonstrated that MD reduced the density of interneurons and induced microglial activation, in particular, PFC areas at young adulthood, and could alter synaptic plasticity accompanied by PFC dysfunction.
Collapse
Affiliation(s)
- Joko Poleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Aksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Slobodan Kapor
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dubravka Aleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tihomir Stojkovic
- Institute of Clinical and Medical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marina Radovic
- Institute of Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vuk Djulejic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branka Markovic
- Faculty of Sport and Physical Education, University of Belgrade, Belgrade, Serbia
| | - Antonios Stamatakis
- Biology-Biochemistry Lab, School of Health Sciences, Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
19
|
Solarz A, Majcher-Maślanka I, Chocyk A. Effects of early-life stress and sex on blood-brain barrier permeability and integrity in juvenile and adult rats. Dev Neurobiol 2021; 81:861-876. [PMID: 34320279 DOI: 10.1002/dneu.22846] [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: 05/12/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/22/2022]
Abstract
Early-life stress (ELS) is considered a relevant etiological factor for neurodegenerative and mental disorders. In the present study, we hypothesized that ELS may persistently and sex dependently influence blood-brain barrier (BBB) integrity and function during critical periods of brain development and consequently determine susceptibility to and sex-related prevalence of chronic diseases in adult life. We used the maternal separation (MS) procedure in rats to model ELS and evaluated BBB permeability and gene expression of selected tight junction (TJ) proteins, glucose transporter type 1 (Slc2a1) and aquaporin 4 (Aqp4) in the medial prefrontal cortex (mPFC), dorsal striatum (dSTR) and hippocampus of juvenile and adult rats. Serum concentrations of a peripheral marker of BBB function (S100β) and proinflammatory cytokines were also assessed. We observed developmental sealing of the BBB and sex differences in the permeability of the BBB and the mRNA expression of TJ proteins and Slc2a1. Adult females showed lower BBB permeability and higher levels of Cldn3, Cldn5, Ocln, and Slc2a1 in the mPFC and dSTR than males. MS temporarily increased BBB permeability in the dSTR of juvenile males and affected mRNA expression of the majority of studied proteins related to BBB function in age-, region- and sex-dependent manners. Additionally, MS sex dependently decreased serum S100β levels and did not affect proinflammatory cytokine concentrations. In general, our study did not reveal a clear or strong negative effect of MS on BBB integrity. However, the results suggest that ELS may induce adaptive/maladaptive changes or compensatory mechanisms within the BBB of unknown yet consequences.
Collapse
Affiliation(s)
- Anna Solarz
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Iwona Majcher-Maślanka
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Agnieszka Chocyk
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
20
|
Solarz A, Majcher-Maślanka I, Kryst J, Chocyk A. A Search for Biomarkers of Early-life Stress-related Psychopathology: Focus on 70-kDa Heat Shock Proteins. Neuroscience 2021; 463:238-253. [PMID: 33662529 DOI: 10.1016/j.neuroscience.2021.02.026] [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: 07/09/2020] [Revised: 02/12/2021] [Accepted: 02/21/2021] [Indexed: 12/31/2022]
Abstract
Clinical studies clearly indicate that early-life stress (ELS) may cause physical and mental health problems later in life. Therefore, the identification of universal biomarkers of ELS-related diseases is very important. The 70-kDa heat shock proteins (HSP70s), specifically HSPA5 and HSPA1B, have been recently shown to be potentially associated with occurrence of anxiety, mood disorders, and schizophrenia; thus, we hypothesized that HSP70s are potential candidate biomarkers of ELS-induced psychopathologies. A maternal separation (MS) procedure in rats was used to model ELS, and the expression of HSPA5 and HSPA1B was investigated in the blood, medial prefrontal cortex (mPFC), and hippocampus of juvenile, preadolescent, and adult animals. We also studied the effects of MS on the long-term potentiation (LTP) and behavioral phenotypes of adult rats. We found that MS enhanced the expression of HSPA1B mRNA in the blood and mPFC of juvenile and preadolescent rats. This increase was accompanied by an increase in the HSPA1A/1B protein levels in the mPFC and hippocampus of juvenile rats that persisted in the mPFC until adulthood. MS juvenile and adult rats showed enhanced HSPA5 mRNA expression in the blood and increased HSPA5 protein expression in the mPFC (juveniles) and hippocampus (adults). Concurrently, MS adult rats exhibited aberrations in LTP in the mPFC and hippocampus and a less anxious behavioral phenotype. These results indicate that MS may produce enduring overexpression of HSPA1B and HSPA5 in the brain and blood. Therefore, both HSP70 family members may be potential candidate peripheral and brain biomarkers of ELS-induced changes in brain functioning.
Collapse
Affiliation(s)
- Anna Solarz
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Iwona Majcher-Maślanka
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Joanna Kryst
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland
| | - Agnieszka Chocyk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Street 12, 31-343 Kraków, Poland.
| |
Collapse
|
21
|
Cui Y, Che Y, Wang H. Nono-titanium dioxide exposure during the adolescent period induces neurotoxicities in rats: Ameliorative potential of bergamot essential oil. Brain Behav 2021; 11:e02099. [PMID: 33694318 PMCID: PMC8119869 DOI: 10.1002/brb3.2099] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/18/2021] [Accepted: 02/18/2021] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION In adolescence, the brain is still maturing, and disorders in maturation may affect the normal development of the brain. Exposure to titanium dioxide nanoparticles (TiO2 NPs) has various potential negative effects on the central nervous system. Bergamot essential oil (BEO) has been found to be effective for neuroprotection. METHODS The rats were injected intraperitoneally with TiO2 NPs (20 mg/kg) and/or BEO (200 mg/kg). The endogenous antioxidant state and inflammatory parameters were estimated using ELISA kits, and then the memory ability and anxiety-like behavior in rats were assessed. RESULTS TiO2 NPs exposure during the adolescent period induced anxiety-like behavior, cognitive impairment, neuroinflammation and oxidative damage in hippocampus, and BEO treatment could significantly ameliorate the neurotoxicities induced by TiO2 NPs exposure. CONCLUSION Our results suggest that the negative effects of TiO2 NPs exposure during the adolescent period on anxiety-like behavior and cognitive function may be related to oxidative stress and neuroinflammation induced by TiO2 NPs exposure. In addition, BEO may ameliorate the neurotoxicities induced by TiO2 NPs exposure in adolescent rats through the antioxidant and anti-inflammatory activity of BEO.
Collapse
Affiliation(s)
- Yonghua Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,Medical College, Soochow University, Suzhou, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yi Che
- Medical College, Soochow University, Suzhou, China
| | - Hongxin Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Wuxi, China
| |
Collapse
|
22
|
Sellinger EP, Drzewiecki CM, Willing J, Juraska JM. Cell death in the male and female rat medial prefrontal cortex during early postnatal development. IBRO Neurosci Rep 2021; 10:186-190. [PMID: 33870262 PMCID: PMC8044638 DOI: 10.1016/j.ibneur.2021.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Abstract
Apoptosis, programmed cell death, is a critical component of neurodevelopment occurring in temporal, spatial, and at times, sex-specific, patterns across the cortex during the early postnatal period. During this time, the brain is particularly susceptible to environmental influences that are often used in animal models of neurodevelopmental disorders. In the present study, the timing of peak cell death was assessed by the presence of pyknotic cells in the male and female rat medial prefrontal cortex (mPFC), a cortical region that in humans, is often involved in developmental disorders. One male and one female rat per litter were sacrificed at the following ages: postnatal day (P)2, 4, 6, 8, 10, 12, 14, 16, 18, and 25. The mPFC was Nissl-stained, the densities of pyknotic cells and live neurons were stereologically collected, and the number of pyknotic cells per 100 live neurons, pyknotic cell density, and neuron density were analyzed. Males and females showed a significant peak in the ratio of pyknotic to live neurons on P8, and in females, this elevation persisted through P12. Likewise, the density of pyknotic cells peaked on P8 in both sexes and persisted through P12 in females. The timing of cell death within the rat mPFC will inform study design in experiments that employ early environmental manipulations that might disrupt this process. The number of pyknotic cells per live neuron was quantified. Postnatal cell death peaked on P8 in the male rat medial prefrontal cortex. In females, postnatal cell death peaked from P8 to P12.
Collapse
Affiliation(s)
- Elli P Sellinger
- Program in Neuroscience, University of Illinois at Urbana-Champaign, Champaign, IL 61801, United States
| | - Carly M Drzewiecki
- Program in Neuroscience, University of Illinois at Urbana-Champaign, Champaign, IL 61801, United States
| | - Jari Willing
- Department of Psychology, University of Illinois at Urbana-Champaign, 603 E Daniel St, Champaign, IL 61820, United States
| | - Janice M Juraska
- Department of Psychology, University of Illinois at Urbana-Champaign, 603 E Daniel St, Champaign, IL 61820, United States
| |
Collapse
|
23
|
Vilela FC, Vieira JS, Vitor-Vieira F, Kalil-Cutti B, da Silva JRT, Giusti-Paiva A, da Silva ML. Maternal separation increases pain sensitivity by reducing the activity of serotonergic neurons in the dorsal raphe nucleus and noradrenergic neurons in locus coeruleus. Neurosci Lett 2021; 748:135734. [PMID: 33596470 DOI: 10.1016/j.neulet.2021.135734] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 01/04/2023]
Abstract
Animals subjected to early life maternal separation exhibit increased sensitivity to chemical, thermal, and mechanical stimuli during adulthood. However, the mechanism by which maternal separation can alter pain sensitivity in adulthood has not yet been investigated. Thus, we aimed to evaluate the activity of serotonergic and noradrenergic neurons and the effect of serotonin (5-HT) and noradrenaline (NA) reuptake inhibitors in male and female Wistar rats subjected to maternal separation. This study consisted of two experiments: 1) to confirm whether maternal separation increased pain sensitivity (n = 8 per group) and to evaluate the activity of serotonergic neurons in the dorsal raphe nucleus and noradrenergic neurons in locus coeruleus in animals subjected to maternal separation in comparison to controls (n = 6 per group); and 2) to evaluate the effect of fluoxetine (a selective 5-HT reuptake inhibitor) and desipramine (a NA reuptake inhibitor) on sensitivity to chemical stimulation using formalin in animals subjected to maternal separation (n = 8 per group). Our findings indicated that maternal separation increases an animal's sensitivity to painful chemical stimulation and reduces the activity of 5-HT and NA neurons. In addition, acute pretreatment with a 5-HT or NA reuptake inhibitor prevented the increased response to painful stimulation induced by maternal separation. In conclusion, maternal separation increases pain sensitivity by reducing the activity of serotonergic neurons in the dorsal raphe nucleus and noradrenergic neurons in locus coeruleus. This study contributes to possible treatments for pain in individuals exposed to early life stress.
Collapse
Affiliation(s)
- Fabiana C Vilela
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil.
| | - Jádina S Vieira
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Fernando Vitor-Vieira
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Bruna Kalil-Cutti
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Josie R T da Silva
- Instituto de Ciências da Motricidade, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Alexandre Giusti-Paiva
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| | - Marcelo L da Silva
- Instituto de Ciências da Motricidade, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Brazil
| |
Collapse
|
24
|
Oldham Green N, Maniam J, Riese J, Morris MJ, Voineagu I. Transcriptomic signature of early life stress in male rat prefrontal cortex. Neurobiol Stress 2021; 14:100316. [PMID: 33796639 PMCID: PMC7995657 DOI: 10.1016/j.ynstr.2021.100316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 01/02/2023] Open
Abstract
Early life stress (ELS) is associated with adverse mental health outcomes including anxiety, depression and addiction-like behaviours. While ELS is known to affect the developing brain, leading to increased stress responsiveness and increased glucocorticoid levels, the molecular mechanisms underlying the detrimental effects of ELS remain incompletely characterised. Rodent models have been instrumental in beginning to uncover the molecular and cellular underpinnings of ELS. Limited nesting (LN), an ELS behavioural paradigm with significant improvements over maternal separation, mimics human maternal neglect. We have previously shown that LN leads to an increase in one of the behavioural measures of anxiety like-behaviours in rats (percent of entries in the EPM open arm). Here we assessed gene expression changes induced by ELS in rat prefrontal cortex by RNA-sequencing. We show that LN leads primarily to transcriptional repression and identify a molecular signature of LN in rat PFC that is observed across ELS protocols and replicable across rodent species (mouse and rat).
Collapse
Affiliation(s)
- Nicole Oldham Green
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jayanthi Maniam
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jessica Riese
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Margaret J Morris
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Irina Voineagu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Tremblay A, Lingrand L, Maillard M, Feuz B, Tompkins TA. The effects of psychobiotics on the microbiota-gut-brain axis in early-life stress and neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110142. [PMID: 33069817 DOI: 10.1016/j.pnpbp.2020.110142] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/28/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Psychobiotics are considered among potential avenues for modulating the bidirectional communication between the gastrointestinal tract and central nervous system, defined as the microbiota-gut-brain axis (MGBA). Even though causality has not yet been established, intestinal dysbiosis has emerged as a hallmark of several diseases, including neuropsychiatric disorders (NPDs). The fact that the microbiota and central nervous system are co-developing during the first years of life has provided a paradigm suggesting a potential role of psychobiotics for earlier interventions. Studies in animal models of early-life stress (ELS) have shown that they can counteract the pervasive effects of stress during this crucial developmental period, and rescue behavioral symptoms related to anxiety and depression later in life. In humans, evidence from clinical studies on the efficacy of psychobiotics at improving mental outcomes in most NPDs remain limited, except for major depressive disorder for which more studies are available. Consequently, the beneficial effect of psychobiotics on depression-related outcomes in adults are becoming clearer. While the specific mechanisms at play remain elusive, the effect of psychobiotics are generally considered to involve the hypothalamic-pituitary-adrenal axis, intestinal permeability, and inflammation. It is anticipated that future clinical studies will explore the potential role of psychobiotics at mitigating the risk developing NPDs in vulnerable individuals or in the context of childhood adversity. However, such studies remain challenging at present in terms of design and target populations; the profound impact of stress on the proper development of the MGBA during the first year of life is becoming increasingly recognized, but the trajectories post-ELS in humans and the mechanisms by which stress affects the susceptibility to various NPDs are still ill-defined. As psychobiotics are likely to exert both shared and specific mechanisms, a better definition of target subpopulations would allow to tailor psychobiotics selection by aligning mechanistic properties with known pathophysiological mechanisms or risk factors. Here we review the available evidence from clinical and preclinical studies supporting a role for psychobiotics at ameliorating depression-related outcomes, highlighting the knowledge gaps and challenges associated with conducting longitudinal studies to address outstanding key questions in the field.
Collapse
Affiliation(s)
- Annie Tremblay
- Rosell® Institute for Microbiome and Probiotics, 6100 Royalmount Avenue, Montreal, Quebec H4P 2R2, Canada
| | - Lucie Lingrand
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Morgane Maillard
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Berengere Feuz
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Thomas A Tompkins
- Rosell® Institute for Microbiome and Probiotics, 6100 Royalmount Avenue, Montreal, Quebec H4P 2R2, Canada.
| |
Collapse
|
26
|
Martynyuk AE, Ju LS, Morey TE, Zhang JQ. Neuroendocrine, epigenetic, and intergenerational effects of general anesthetics. World J Psychiatry 2020; 10:81-94. [PMID: 32477904 PMCID: PMC7243620 DOI: 10.5498/wjp.v10.i5.81] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/18/2020] [Accepted: 03/26/2020] [Indexed: 02/05/2023] Open
Abstract
The progress of modern medicine would be impossible without the use of general anesthetics (GAs). Despite advancements in refining anesthesia approaches, the effects of GAs are not fully reversible upon GA withdrawal. Neurocognitive deficiencies attributed to GA exposure may persist in neonates or endure for weeks to years in the elderly. Human studies on the mechanisms of the long-term adverse effects of GAs are needed to improve the safety of general anesthesia but they are hampered not only by ethical limitations specific to human research, but also by a lack of specific biological markers that can be used in human studies to safely and objectively study such effects. The latter can primarily be attributed to an insufficient understanding of the full range of the biological effects induced by GAs and the molecular mechanisms mediating such effects even in rodents, which are far more extensively studied than any other species. Our most recent experimental findings in rodents suggest that GAs may adversely affect many more people than is currently anticipated. Specifically, we have shown that anesthesia with the commonly used GA sevoflurane induces in exposed animals not only neuroendocrine abnormalities (somatic effects), but also epigenetic reprogramming of germ cells (germ cell effects). The latter may pass the neurobehavioral effects of parental sevoflurane exposure to the offspring, who may be affected even at levels of anesthesia that are not harmful to the exposed parents. The large number of patients who require general anesthesia, the even larger number of their future unexposed offspring whose health may be affected, and a growing number of neurodevelopmental disorders of unknown etiology underscore the translational importance of investigating the intergenerational effects of GAs. In this mini review, we discuss emerging experimental findings on neuroendocrine, epigenetic, and intergenerational effects of GAs.
Collapse
Affiliation(s)
- Anatoly E Martynyuk
- Department of Anesthesiology and the McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Ling-Sha Ju
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Timothy E Morey
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Jia-Qiang Zhang
- Department of Anesthesiology and Perioperative Medicine, Henan Provincial People’s Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| |
Collapse
|
27
|
Murthy S, Gould E. How Early Life Adversity Influences Defensive Circuitry. Trends Neurosci 2020; 43:200-212. [PMID: 32209452 DOI: 10.1016/j.tins.2020.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022]
Abstract
Childhood maltreatment increases the likelihood of developing anxiety disorders in humans. Early life adversity (ELA) paradigms in rodents produce lasting increases in avoidant and inhibitory responses to both immediate and nonspecific threats, collectively referred to as defensive behaviors. This approach provides an opportunity to thoroughly investigate the underlying mechanisms, an effort that is currently under way. In this review, we consider the growing literature indicating that ELA alters the rhythmic firing of neurons in brain regions associated with defensive behavior, as well as potential neuronal, glial, and extracellular matrix contributions to functional changes in this circuitry. We also consider how ELA studies in rodents may inform us about both susceptible and resilient outcomes in humans.
Collapse
Affiliation(s)
- Sahana Murthy
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
28
|
Dutcher EG, Pama EC, Lynall ME, Khan S, Clatworthy MR, Robbins TW, Bullmore ET, Dalley JW. Early-life stress and inflammation: A systematic review of a key experimental approach in rodents. Brain Neurosci Adv 2020; 4:2398212820978049. [PMID: 33447663 PMCID: PMC7780197 DOI: 10.1177/2398212820978049] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
Repeated maternal separation is the most widely used pre-clinical approach to investigate the relationship between early-life chronic stress and its neuropsychiatric and physical consequences. In this systematic review, we identified 46 studies that conducted repeated maternal separation or single-episode maternal separation and reported measurements of interleukin-1b, interleukin-6, interleukin-10, tumour necrosis factor-alpha, or microglia activation and density. We report that in the short-term and in the context of later-life stress, repeated maternal separation has pro-inflammatory immune consequences in diverse tissues. Repeated maternal separation animals exhibit greater microglial activation and elevated pro-inflammatory cytokine signalling in key brain regions implicated in human psychiatric disorders. Notably, repeated maternal separation generally has no long-term effect on cytokine expression in any tissue in the absence of later-life stress. These observations suggest that the elevated inflammatory signalling that has been reported in humans with a history of early-life stress may be the joint consequence of ongoing stressor exposure together with potentiated neural and/or immune responsiveness to stressors. Finally, our findings provide detailed guidance for future studies interrogating the causal roles of early-life stress and inflammation in disorders such as major depression.
Collapse
Affiliation(s)
- Ethan G. Dutcher
- Department of Psychology, University of Cambridge, Cambridge, UK
| | | | - Mary-Ellen Lynall
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Shahid Khan
- GlaxoSmithKline Research & Development, Stevenage, UK
| | | | | | | | - Jeffrey W. Dalley
- Department of Psychology, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
29
|
Zhang X, Li H, Sun H, Jiang Y, Wang A, Kong Y, Sun X, Zhu G, Li Q, Du Z, Sun H, Sun L. Effects of BDNF Signaling on Anxiety-Related Behavior and Spatial Memory of Adolescent Rats in Different Length of Maternal Separation. Front Psychiatry 2020; 11:709. [PMID: 32793001 PMCID: PMC7391957 DOI: 10.3389/fpsyt.2020.00709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
As an adverse form of early-life stress (ELS), maternal separation (MS) can interfere with the development of cognition and behaviors of adolescent rodents. Brain-derived neurotrophic factor (BDNF) is involved in the regulation of brain development and function, but the molecular mechanisms by which BDNF regulates brain function and behavior in MS with different stressor strengths remain unclear. This descriptive study characterized the levels of BDNF in the prefrontal cortex (PFC) and plasma corticosterone (CORT) from the offspring of rats exposed to early handling (EH, 15-min separation per day) and prolonged MS (PMS, 180-min separation per day), during postnatal days (PND) 1‑21. The behavioral and biochemical analyses were performed during adolescence (PND 42‑56). PMS resulted in reduced weight and decreased locomotor activity in the open field test and Y-maze task compared to control (CON) group, with EH showing an intermediate phenotype. BDNF protein levels in the PFC were lower in PMS compared to EH and further reduced in CON male rats. Plasma CORT levels were higher in PMS compared to CON with EH again showing intermediate levels. Neither PMS or EH affected spatial learning in the Y-maze task. These findings indicate that longer periods of maternal separation are necessary to increase anxiety-like behavior, elevate CORT levels, and further suppress BDNF levels in the PFC, providing a possible mechanism to explain why more severe forms of ELS lead to more significant psychiatric and medical consequences later in life.
Collapse
Affiliation(s)
- Xianqiang Zhang
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Haonan Li
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Haoran Sun
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yinghong Jiang
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Aihong Wang
- Department of Hematology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yujia Kong
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Xiue Sun
- Department of Nursing, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Guohui Zhu
- Depression Treatment Center, Weifang Mental Health Center, Weifang, China
| | - Qi Li
- Department of Psychiatry and Centre for Reproduction Growth and Development, University of Hong Kong, Hong Kong, Hong Kong
| | - Zhongde Du
- Department of Neurology, Sunshine Union Hospital, Weifang, China
| | - Hongwei Sun
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Lin Sun
- Department of Psychology, Weifang Medical University, Weifang, China
| |
Collapse
|