1
|
Dong MC, Chen YX, Sun XR, Jiang N, Chang Q, Liu XM, Pan RL. One-Week Maternal Separation Caused Sex-Specific Changes in Behavior and Hippocampal Metabolomics of Offspring Rats. Brain Sci 2024; 14:1275. [PMID: 39766474 PMCID: PMC11674698 DOI: 10.3390/brainsci14121275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
To investigate the effects of one-week maternal separation (MS) on anxiety- and depression-like behaviors in adolescent and adulthood as well as adult hippocampal metabolomics simultaneously in offspring female and male rats. In the MS group, newborn SD rats were separated from their mothers for 3 h per day from postnatal days (PND) 2 to 8. The open field test (OFT), elevated plus mazes (EPM), novelty suppressed feeding test (NSFT), and forced swimming test (FST) were conducted during adolescence and adulthood. Serum corticosterone, mRNA expression of hippocampal inflammatory cytokines, and hippocampal untargeted metabolomics of offspring adult rats were examined using an assay kit, qRT-PCR, and UPLC-Q-TOF/MS. Both MS female and male rats showed similar behaviors in OFT, EPM, NSFT, and SPT, except for the latency to feeding during adolescence and the open arm entries during adulthood, showed statistical significance only in MS female rats. Serum corticosterone and hippocampal pro-inflammatory cytokines IFN-γ were significantly elevated in both female and male rats, and IL-1β and TNF-α were significantly increased only in female rats. In hippocampal metabolism, the identification of differential metabolites displayed 53 and 37 in female rats and male rats, respectively (with 35 common metabolites), which were involved in 33 and 30 metabolic pathways with 28 common pathways. One-week MS induced sex-specific anxiety- and depression-like behaviors in female and male offspring rats during adolescence and adulthood, as well as sex-differentiated characteristics in the hippocampus inflammatory cytokines and metabolomics of adult MS rats. From the experimental data, the effects of MS on the female offspring rats were more severe than those of the male offspring rats.
Collapse
Affiliation(s)
- Meng-Chen Dong
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (M.-C.D.); (Y.-X.C.); (X.-R.S.); (N.J.); (Q.C.)
| | - Yu-Xin Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (M.-C.D.); (Y.-X.C.); (X.-R.S.); (N.J.); (Q.C.)
| | - Xin-Ran Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (M.-C.D.); (Y.-X.C.); (X.-R.S.); (N.J.); (Q.C.)
| | - Ning Jiang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (M.-C.D.); (Y.-X.C.); (X.-R.S.); (N.J.); (Q.C.)
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (M.-C.D.); (Y.-X.C.); (X.-R.S.); (N.J.); (Q.C.)
| | - Xin-Min Liu
- Institute of Drug Discovery Technology, Ningbo University, No. 818, Fenghua Road, Jiangbei District, Ningbo 315000, China
| | - Rui-Le Pan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China; (M.-C.D.); (Y.-X.C.); (X.-R.S.); (N.J.); (Q.C.)
| |
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
|
Krejcová LV, Bento-Torres J, Diniz DG, Pereira A, Batista-de-Oliveira M, de Morais AACL, Mendes-da-Silva RF, Abadie-Guedes R, dos Santos ÂA, Lima DS, Guedes RCA, Picanço-Diniz CW. Unraveling the Influence of Litter Size, Maternal Care, Exercise, and Aging on Neurobehavioral Plasticity and Dentate Gyrus Microglia Dynamics in Male Rats. Brain Sci 2024; 14:497. [PMID: 38790475 PMCID: PMC11119659 DOI: 10.3390/brainsci14050497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
This study explores the multifaceted influence of litter size, maternal care, exercise, and aging on rats' neurobehavioral plasticity and dentate gyrus microglia dynamics. Body weight evolution revealed a progressive increase until maturity, followed by a decline during aging, with larger litters exhibiting lower weights initially. Notably, exercised rats from smaller litters displayed higher body weights during the mature and aged stages. The dentate gyrus volumes showed no significant differences among groups, except for aged sedentary rats from smaller litters, which exhibited a reduction. Maternal care varied significantly based on litter size, with large litter dams showing lower frequencies of caregiving behaviors. Behavioral assays highlighted the detrimental impact of a sedentary lifestyle and reduced maternal care/large litters on spatial memory, mitigated by exercise in aged rats from smaller litters. The microglial dynamics in the layers of dentate gyrus revealed age-related changes modulated by litter size and exercise. Exercise interventions mitigated microgliosis associated with aging, particularly in aged rats. These findings underscore the complex interplay between early-life experiences, exercise, microglial dynamics, and neurobehavioral outcomes during aging.
Collapse
Affiliation(s)
- Lane Viana Krejcová
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - João Bento-Torres
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - Daniel Guerreiro Diniz
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
- Postgraduate Program in Oncology and Medical Sciences, João de Barros Barreto Universitary Hospital, Federal University of Pará, Belém 66075-110, Pará, Brazil
- Electron Microscopy Laboratory, Evandro Chagas Institute, Belém 66093-020, Pará, Brazil
| | - Antonio Pereira
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - Manuella Batista-de-Oliveira
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | | | | | - Ricardo Abadie-Guedes
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Ângela Amâncio dos Santos
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Denise Sandrelly Lima
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Rubem Carlos Araujo Guedes
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Cristovam Wanderley Picanço-Diniz
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| |
Collapse
|
4
|
He H, He H, Mo L, You Z, Zhang J. Priming of microglia with dysfunctional gut microbiota impairs hippocampal neurogenesis and fosters stress vulnerability of mice. Brain Behav Immun 2024; 115:280-294. [PMID: 37914097 DOI: 10.1016/j.bbi.2023.10.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Mental disorders may be involved in neuroinflammatory processes that are triggered by gut microbiota. How gut microbiota influence microglia-mediated sensitivity to stress remains unclear. Here we explored in an animal model of depression whether disruption of the gut microbiome primes hippocampal microglia, thereby impairing neurogenesis and sensitizing to stress. METHODS Male C57BL/6J mice were exposed to chronic unpredictable mild stress (CUMS) for 4 weeks, and effects on gut microbiota were assessed using 16S rRNA sequencing. Fecal microbiota was transplanted from control or CUMS mice into naïve animals. The depression-like behaviors of recipients were evaluated in a forced swimming test and sucrose preference test. The morphology and phenotype of microglia in the hippocampus of recipients were examined using immunohistochemistry, quantitative PCR, and enzyme-linked immunosorbent assays. The recipients were treated with lipopolysaccharide or chronic stress exposure, and effects were evaluated on behavior, microglial responses and hippocampal neurogenesis. Finally, we explored the ability of minocycline to reverse the effects of CUMS on hippocampal neurogenesis and stress sensitivity in recipients. RESULTS CUMS altered the gut microbiome, leading to higher relative abundance of some bacteria (Helicobacter, Bacteroides, and Desulfovibrio) and lower relative abundance of some bacteria (Lactobacillus, Bifidobacterium, and Akkermansia). Fecal microbiota transplantation from CUMS mice to naïve animals induced microglial priming in the dentate gyrus of recipients. This microglia showed hyper-ramified morphology, and became more sensitive to LPS challenge or chronic stress, which characterized by more significant morphological changes and inflammatory responses, as well as impaired hippocampal neurogenesis and increased depressive-like behaviors. Giving minocycline to recipients reversed these effects of fecal transplantation. CONCLUSIONS These findings suggest that gut microbiota from stressed animals can induce microglial priming in the dentate gyrus, which is associated with a hyper-immune response to stress and impaired hippocampal neurogenesis. Remodeling the gut microbiome or inhibiting microglial priming may be strategies to reduce sensitivity to stress.
Collapse
Affiliation(s)
- Hui He
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Haili He
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Li Mo
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Zili You
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
5
|
Nuzum ND, Deady C, Kittel-Schneider S, Cryan JF, O'Mahony SM, Clarke G. More than just a number: the gut microbiota and brain function across the extremes of life. Gut Microbes 2024; 16:2418988. [PMID: 39567371 PMCID: PMC11583591 DOI: 10.1080/19490976.2024.2418988] [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: 05/28/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024] Open
Abstract
Understanding the interrelationship between the gut microbiota and host physiology, although still in its relative infancy, has taken important steps forward over the past decade. In the context of brain disorders including those characterized by neurodevelopmental and neurodegenerative changes there have been important advances. However, initially research involved correlational analyses, had limited translational scope, and lacked functional assessments. Thus, largescale longitudinal clinical investigations that assess causation and underlying mechanisms via in depth analysis methods are needed. In neurodegeneration research, strong causal evidence now links the gut microbiome to Alzheimer's (AD), and Parkinson's Disease (PD), as supported by human-to-animal transplantation studies. Longitudinal interventions are being conducted in AD, PD, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. Neurodevelopmental research has also seen a boon in microbiome-related clinical research including in autism, Attention-deficit/hyperactivity disorder, and schizophrenia, which is confirming prior animal model work regarding the key time-windows in the gut microbiome important for infant cognition. While recent research advances represent important progress, fundamental knowledge gaps and obstacles remain. Knowing how and why the gut microbiome changes at the extremes of life will develop our mechanistic understanding and help build the evidence base as we strive toward counteracting microbial missteps with precision therapeutic interventions.
Collapse
Affiliation(s)
- Nathan D Nuzum
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Sarah Kittel-Schneider
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Siobhain M O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| |
Collapse
|
6
|
Grabrucker S, Marizzoni M, Silajdžić E, Lopizzo N, Mombelli E, Nicolas S, Dohm-Hansen S, Scassellati C, Moretti DV, Rosa M, Hoffmann K, Cryan JF, O’Leary OF, English JA, Lavelle A, O’Neill C, Thuret S, Cattaneo A, Nolan YM. Microbiota from Alzheimer's patients induce deficits in cognition and hippocampal neurogenesis. Brain 2023; 146:4916-4934. [PMID: 37849234 PMCID: PMC10689930 DOI: 10.1093/brain/awad303] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/29/2023] [Accepted: 08/19/2023] [Indexed: 10/19/2023] Open
Abstract
Alzheimer's disease is a complex neurodegenerative disorder leading to a decline in cognitive function and mental health. Recent research has positioned the gut microbiota as an important susceptibility factor in Alzheimer's disease by showing specific alterations in the gut microbiome composition of Alzheimer's patients and in rodent models. However, it is unknown whether gut microbiota alterations are causal in the manifestation of Alzheimer's symptoms. To understand the involvement of Alzheimer's patient gut microbiota in host physiology and behaviour, we transplanted faecal microbiota from Alzheimer's patients and age-matched healthy controls into microbiota-depleted young adult rats. We found impairments in behaviours reliant on adult hippocampal neurogenesis, an essential process for certain memory functions and mood, resulting from Alzheimer's patient transplants. Notably, the severity of impairments correlated with clinical cognitive scores in donor patients. Discrete changes in the rat caecal and hippocampal metabolome were also evident. As hippocampal neurogenesis cannot be measured in living humans but is modulated by the circulatory systemic environment, we assessed the impact of the Alzheimer's systemic environment on proxy neurogenesis readouts. Serum from Alzheimer's patients decreased neurogenesis in human cells in vitro and were associated with cognitive scores and key microbial genera. Our findings reveal for the first time, that Alzheimer's symptoms can be transferred to a healthy young organism via the gut microbiota, confirming a causal role of gut microbiota in Alzheimer's disease, and highlight hippocampal neurogenesis as a converging central cellular process regulating systemic circulatory and gut-mediated factors in Alzheimer's.
Collapse
Affiliation(s)
- Stefanie Grabrucker
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Moira Marizzoni
- Biological Psychiatry Unit, IRCCS Fatebenefratelli, Brescia, Italy
- Laboratory of Neuroimaging and Alzheimer’s Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Edina Silajdžić
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, King’s College London, SE5 9NU London, UK
| | - Nicola Lopizzo
- Biological Psychiatry Unit, IRCCS Fatebenefratelli, Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Elisa Mombelli
- Biological Psychiatry Unit, IRCCS Fatebenefratelli, Brescia, Italy
| | - Sarah Nicolas
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Sebastian Dohm-Hansen
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
- INFANT Research Centre, University College Cork, T12 DC4A Cork, Ireland
| | | | | | - Melissa Rosa
- Biological Psychiatry Unit, IRCCS Fatebenefratelli, Brescia, Italy
| | - Karina Hoffmann
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, King’s College London, SE5 9NU London, UK
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Olivia F O’Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Jane A English
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- INFANT Research Centre, University College Cork, T12 DC4A Cork, Ireland
| | - Aonghus Lavelle
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Cora O’Neill
- APC Microbiome Ireland, University College Cork, Ireland
- School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, T12 YT20 Cork, Ireland
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, King’s College London, SE5 9NU London, UK
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Fatebenefratelli, Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| |
Collapse
|
7
|
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
|
8
|
Pate T, Anthony DC, Radford-Smith DE. cFOS expression in the prefrontal cortex correlates with altered cerebral metabolism in developing germ-free mice. Front Mol Neurosci 2023; 16:1155620. [PMID: 37152431 PMCID: PMC10157641 DOI: 10.3389/fnmol.2023.1155620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction The microbiota plays a critical role in modulating various aspects of host physiology, particularly through the microbiota-gut-brain (MGB) axis. However, the mechanisms that transduce and affect gut-to-brain communication are still not well understood. Recent studies have demonstrated that dysbiosis of the microbiome is associated with anxiety and depressive symptoms, which are common complications of metabolic syndrome. Germ-free (GF) animal models offer a valuable tool for studying the causal effects of microbiota on the host. Methods We employed gene expression and nuclear magnetic resonance (NMR)-based metabolomic techniques to investigate the relationships between brain plasticity and immune gene expression, peripheral immunity, and cerebral and liver metabolism in GF and specific pathogen-free (SPF) mice. Results Our principal findings revealed that brain acetate (p = 0.012) was significantly reduced in GF relative to SPF mice, whereas glutamate (p = 0.0013), glutamine (p = 0.0006), and N-acetyl aspartate (p = 0.0046) metabolites were increased. Notably, cFOS mRNA expression, which was significantly decreased in the prefrontal cortex of GF mice relative to SPF mice (p = 0.044), correlated with the abundance of a number of key brain metabolites altered by the GF phenotype, including glutamate and glutamine. Discussion These results highlight the connection between the GF phenotype, altered brain metabolism, and immediate-early gene expression. The study provides insight into potential mechanisms by which microbiota can regulate neurotransmission through modulation of the host's brain and liver metabolome, which may have implications for stress-related psychiatric disorders such as anxiety.
Collapse
|
9
|
Reemst K, Broos JY, Abbink MR, Cimetti C, Giera M, Kooij G, Korosi A. Early-life stress and dietary fatty acids impact the brain lipid/oxylipin profile into adulthood, basally and in response to LPS. Front Immunol 2022; 13:967437. [PMID: 36131915 PMCID: PMC9484596 DOI: 10.3389/fimmu.2022.967437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/04/2022] [Indexed: 01/06/2023] Open
Abstract
Brain lipid dysregulation is a hallmark of depression and Alzheimer's disease, also marked by chronic inflammation. Early-life stress (ELS) and dietary intake of polyunsaturated fatty acids (PUFAs) are risk factors for these pathologies and are known to impact inflammatory processes. However, if these early-life factors alter brain lipid homeostasis on the long-term and thereby contribute to this risk remains to be elucidated. We have recently shown that an early diet enriched in omega(ω)-3 PUFAs protected against the long-term negative effects of ELS on cognition and neuroinflammation. Here, we aim to understand if modulation of brain lipid and oxylipin profiles contributes to the detrimental effects of ELS and the protective ones of the diet. We therefore studied if and how ELS and early dietary PUFAs modulate the brain lipid and oxylipin profile, basally as well as in response to an inflammatory challenge, to unmask possible latent effects. Male mice were exposed to ELS via the limited bedding and nesting paradigm, received an early diet with high or low ω6/ω3 ratio (HRD and LRD) and were injected with saline or lipopolysaccharide (LPS) in adulthood. Twenty-four hours later plasma cytokines (Multiplex) and hypothalamic lipids and oxylipins (liquid chromatography tandem mass spectrometry) were measured. ELS exacerbated the LPS-induced increase in IL-6, CXCL1 and CCL2. Both ELS and diet affected the lipid/oxylipin profile long-term. For example, ELS increased diacylglycerol and LRD reduced triacylglycerol, free fatty acids and ceramides. Importantly, the ELS-induced alterations were strongly influenced by the early diet. For example, the ELS-induced decrease in eicosapentaenoic acid was reversed when fed LRD. Similarly, the majority of the LPS-induced alterations were distinct for control and ELS exposed mice and unique for mice fed with LRD or HRD. LPS decreased ceramides and lysophosphotidylcholine, increased hexosylceramides and prostaglandin E2, reduced triacylglycerol species and ω6-derived oxylipins only in mice fed LRD and ELS reduced the LPS-induced increase in phosphatidylcholine. These data give further insights into the alterations in brain lipids and oxylipins that might contribute to the detrimental effects of ELS, to the protective ones of LRD and the possible early-origin of brain lipid dyshomeostasis characterizing ELS-related psychopathologies.
Collapse
Affiliation(s)
- Kitty Reemst
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park, Amsterdam, Netherlands
| | - Jelle Y. Broos
- Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Multiple Sclerosis (MS) Center Amsterdam, Amsterdam, Netherlands,Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Maralinde R. Abbink
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park, Amsterdam, Netherlands
| | - Chiara Cimetti
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park, Amsterdam, Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Gijs Kooij
- Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Multiple Sclerosis (MS) Center Amsterdam, Amsterdam, Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park, Amsterdam, Netherlands,*Correspondence: Aniko Korosi,
| |
Collapse
|
10
|
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
|
11
|
Zhao C, Sheng C, Zhou C. Fast Gelation of Poly(ionic liquid)-Based Injectable Antibacterial Hydrogels. Gels 2022; 8:52. [PMID: 35049587 PMCID: PMC8775204 DOI: 10.3390/gels8010052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 12/11/2022] Open
Abstract
Traditional antibacterial hydrogels have a broad-spectrum bactericidal effect and are widely used as wound dressings. However, the biological toxicity and drug resistance of these antibacterial hydrogels cannot meet the requirements of long-term clinical application. Imidazolium poly(ionic liquids) (PILs) are polymeric antibacterial agents exhibiting strong antibacterial properties, as they contain a strong positive charge. In this study, two imidazolium PILs, namely poly(N-butylimidazolium propiolic acid sodium) (PBP) and poly(N-(3,6-dioxaoctane) imidazolium propiolic acid sodium) (PDP), as high efficiency antibacterial agents, were synthesized by polycondensation reaction. Then, the PILs were compounded with polyethylene glycol (PEG) by a thiol-yne click reaction to prepare injectable antibacterial hydrogels. An in vitro assay showed that the injectable antibacterial hydrogels could not only quickly kill Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus), but also had low toxicity for human skin fibroblasts cells (HSFs) and human umbilical vein endothelial cells (HUVECs), respectively. Additionally, the lipopolysaccharide (LPS) inflammation model revealed that the injectable antibacterial hydrogels also had anti-inflammatory effects, which would be advantageous to accelerate wound healing.
Collapse
Affiliation(s)
- Che Zhao
- School of Aerospace and Mechanical Engineering, Changzhou Institute of Technology, Changzhou 213032, China;
| | - Chengju Sheng
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Chao Zhou
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China
| |
Collapse
|
12
|
Harris EP, McGovern AJ, Melo TG, Barron A, Nola YM, O'Leary OF. Juvenile Stress Exerts Sex-independent Effects on Anxiety, Antidepressant-like Behaviours and Dopaminergic Innervation of the Prelimbic Cortex in Adulthood and Does Not Alter Hippocampal Neurogenesis. Behav Brain Res 2021; 421:113725. [PMID: 34929235 DOI: 10.1016/j.bbr.2021.113725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/24/2021] [Accepted: 12/15/2021] [Indexed: 11/26/2022]
Abstract
Stress, particularly during childhood, is a major risk factor for the development of depression. Depression is twice as prevalent in women compared to men, which suggests that that biological sex also contributes to depression susceptibility. However, the neurobiology underpinning sex differences in the long-term consequences of childhood stress remains unknown. Thus, the aim of this study was to determine whether stress applied during the prepubertal juvenile period (postnatal day 27-29) in rats induces sex-specific changes in anxiety-like behaviour, anhedonia, and antidepressant-like behaviour in adulthood in males and females. The impact of juvenile stress on two systems in the brain associated with these behaviours and that develop during the juvenile period, the mesocorticolimbic dopaminergic system and hippocampal neurogenesis, were also investigated. Juvenile stress altered escape-oriented behaviours in the forced swim test in both sexes, decreased latency to drink a palatable substance in a novel environment in the novelty-induced hypophagia test in both sexes, and decreased open field supported rearing behavior in females. These behavioural changes were accompanied by stress-induced increases in tyrosine hydroxylase immunoreactivity in the prefrontal cortex of both sexes, but not other regions of the mesocorticolimbic dopaminergic system. Juvenile stress did not impact anhedonia in adulthood as measured by the saccharin preference test and had no effect hippocampal neurogenesis across the longitudinal axis of the hippocampus. These results suggest that juvenile stress has long-lasting impacts on antidepressant-like and reward-seeking behaviour in adulthood and these changes may be due to alterations to catecholaminergic innervation of the medial prefrontal cortex.
Collapse
Affiliation(s)
- Erin P Harris
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Andrew J McGovern
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Thieza G Melo
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Aaron Barron
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Yvonne M Nola
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|