1
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 DOI: 10.1016/j.neuropharm.2024.110084] [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: 02/15/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Microglia play numerous important roles in brain development. From early embryonic stages through adolescence, these immune cells influence neuronal genesis and maturation, guide connectivity, and shape brain circuits. They also interact with other glial cells and structures, influencing the brain's supportive microenvironment. While this central role makes microglia essential, it means that early life perturbations to microglia can have widespread effects on brain development, potentially resulting in long-lasting behavioral impairments. Here, we will focus on the effects of early life psychosocial versus physiological stressors in rodent models. Psychosocial stress refers to perceived threats that lead to stress axes activation, including prenatal stress, or chronic postnatal stress, including maternal separation and resource scarcity. Physiological stress refers to physical threats, including maternal immune activation, postnatal infection, and traumatic brain injury. Differing sources of early life stress have varied impacts on microglia, and these effects are moderated by factors such as developmental age, brain region, and sex. Overall, these stressors appear to either 1) upregulate basal microglia numbers and activity throughout the lifespan, while possibly blunting their responsivity to subsequent stressors, or 2) shift the developmental curve of microglia, resulting in differential timing and function, impacting the critical periods they govern. Either could contribute to behavioral dysfunctions that occur after the resolution of early life stress. Exploring how different stressors impact microglia, as well as how multiple stressors interact to alter microglia's developmental functions, could deepen our understanding of how early life stress changes the brain's developmental trajectory. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Psychology, Ohio State University, Columbus, OH, USA.
| | - Kathryn M Lenz
- Department of Psychology, Ohio State University, Columbus, OH, USA; Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Ohio State University, Columbus, OH, USA
| |
Collapse
|
2
|
Nikodemova M, Oberto JR, Kaye EL, Berschel MR, Michaelson AL, Watters JJ, Mitchell GS. Acute postnatal inflammation alters adult microglial responses to LPS that are sex-, region- and timing of postnatal inflammation-dependent. J Neuroinflammation 2024; 21:256. [PMID: 39390483 PMCID: PMC11465935 DOI: 10.1186/s12974-024-03245-x] [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: 06/11/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Adverse events in early life can have impact lasting into adulthood. We investigated the long-term effects of systemic inflammation during postnatal development on adult microglial responses to lipopolysaccharide (LPS) in two CNS regions (cortex, cervical spinal cord) in male and female rats. METHODS Inflammation was induced in Sprague-Dawley rats by LPS (1 mg/kg) administered intraperitoneally during postnatal development at P7, P12 or P18. As adults (12 weeks of age), the rats received a second LPS dose (1 mg/kg). Control rats received saline. Microglia were isolated 3 h post-LPS followed by gene expression analysis via qRT-PCR for pro-inflammatory (IL-6, iNOS, Ptgs2, C/EBPb, CD14, CXCL10), anti-inflammatory (CD68, Arg-1), and homeostatic genes (P2Y12, Tmemm119). CSF-1 and CX3CL1 mRNAs were analyzed in microglia-free homogenates. RESULTS Basal gene expression in adult microglia was largely unaffected by postnatal inflammation. Adult cortical microglial pro-inflammatory gene responses to LPS were either unchanged or attenuated in rats exposed to LPS during postnatal development. Ptgs2, C/EBPb, CXCL10 and Arg-1 were the most affected genes, with expression significantly downregulated vs. rats without postnatal LPS. Spinal microglia were affected most by LPS at P18, with mixed and sometimes opposing effects on proinflammatory genes in males vs. females. Overall, male cortical vs. spinal microglia were more affected by postnatal LPS. Females were affected in both cortex and spinal cord, but the effect was dependent on timing of postnatal LPS. Overall, inflammatory challenge at P18 had greater effect on adult microglia vs. challenge at P12 or P7. CONCLUSIONS Long-lasting effects of postnatal inflammation on adult microglia depend on postnatal timing, CNS region and sex.
Collapse
Affiliation(s)
- Maria Nikodemova
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| | - Jose R Oberto
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Ethan L Kaye
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Mackenzie R Berschel
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Alysha L Michaelson
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Jyoti J Watters
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53706, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
3
|
Hu A, Zaongo SD, Harypursat V, Wang X, Ouyang J, Chen Y. HIV-associated neurocognitive disorder: key implications of the microbiota-gut-brain axis. Front Microbiol 2024; 15:1428239. [PMID: 39155987 PMCID: PMC11327151 DOI: 10.3389/fmicb.2024.1428239] [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: 05/21/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
HIV-associated neurocognitive disorder (HAND) is now recognized to be relatively common in people living with HIV (PLWH), and remains a common cause of cognitive impairment. Unfortunately, the fundamental pathogenic processes underlying this specific outcome of HIV infection have not as yet been fully elucidated. With increased interest in research related to the microbiota-gut-brain axis, the gut-brain axis has been shown to play critical roles in regulating central nervous system disorders such as Alzheimer's disease and Parkinson's disease. PLWH are characterized by a particular affliction, referred to as gut-associated dysbiosis syndrome, which provokes an alteration in microbial composition and diversity, and of their associated metabolite composition within the gut. Interestingly, the gut microbiota has also been recognized as a key element, which both positively and negatively influences human brain health, including the functioning and development of the central nervous system (CNS). In this review, based on published evidence, we critically discuss the relevant interactions between the microbiota-gut-brain axis and the pathogenesis of HAND in the context of HIV infection. It is likely that HAND manifestation in PLWH mainly results from (i) gut-associated dysbiosis syndrome and a leaky gut on the one hand and (ii) inflammation on the other hand. In other words, the preceding features of HIV infection negatively alter the composition of the gut microbiota (microbes and their associated metabolites) and promote proinflammatory immune responses which singularly or in tandem damage neurons and/or induce inadequate neuronal signaling. Thus, HAND is fairly prevalent in PLWH. This work aims to demonstrate that in the quest to prevent and possibly treat HAND, the gut microbiota may ultimately represent a therapeutically targetable "host factor."
Collapse
Affiliation(s)
- Aizhen Hu
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Silvere D. Zaongo
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Vijay Harypursat
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Xin Wang
- Phase I Clinical Trial Center, Chonggang General Hospital, Chongqing, China
| | - Jing Ouyang
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Yaokai Chen
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
4
|
Cheng J, Yuan L, Yu S, Gu B, Luo Q, Wang X, Zhao Y, Gai C, Li T, Liu W, Wang Z, Liu D, Ho RCM, Ho CSH. Programmed cell death factor 4-mediated hippocampal synaptic plasticity is involved in early life stress and susceptibility to depression. Behav Brain Res 2024; 468:115028. [PMID: 38723677 DOI: 10.1016/j.bbr.2024.115028] [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: 02/22/2024] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Early life stress (ELS) increases the risk of depression later in life. Programmed cell death factor 4 (PDCD4), an apoptosis-related molecule, extensively participates in tumorigenesis and inflammatory diseases. However, its involvement in a person's susceptibility to ELS-related depression is unknown. To examine the effects and underlying mechanisms of PDCD4 on ELS vulnerability, we used a "two-hit" stress mouse model: an intraperitoneal injection of lipopolysaccharide (LPS) into neonatal mice was performed on postnatal days 7-9 (P7-P9) and inescapable foot shock (IFS) administration in adolescent was used as a later-life challenge. Our study shows that compared with mice that were only exposed to the LPS or IFS, the "two-hit" stress mice developed more severe depression/anxiety-like behaviors and social disability. We detected the levels of PDCD4 in the hippocampus of adolescent mice and found that they were significantly increased in "two-hit" stress mice. The results of immunohistochemical staining and Sholl analysis showed that the number of microglia in the hippocampus of "two-hit" stress mice significantly increased, with morphological changes, shortened branches, and decreased numbers. However, knocking down PDCD4 can prevent the number and morphological changes of microglia induced by ELS. In addition, we confirmed through the Golgi staining and immunohistochemical staining results that knocking down PDCD4 can ameliorate ELS-induced synaptic plasticity damage. Mechanically, the knockdown of PDCD4 exerts neuroprotective effects, possibly via the mediation of BDNF/AKT/CREB signaling. Combined, these results suggest that PDCD4 may play an important role in the ELS-induced susceptibility to depression and, thus, may become a therapeutic target for depressive disorders.
Collapse
Affiliation(s)
- Jiao Cheng
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Lin Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250012, PR China
| | - Shuwen Yu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Bing Gu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Xixi Wang
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Chengcheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Tingting Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Weiyang Liu
- Jinan Xicheng Experimental High School, Dezhou Road, Jinan, Shandong 1999, PR China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| | - Roger C M Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Institute of Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
| | - Cyrus S H Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
5
|
Biswas B, Eapen V, Morris MJ, Jones NM. Combined Effect of Maternal Separation and Early-Life Immune Activation on Brain and Behaviour of Rat Offspring. Biomolecules 2024; 14:197. [PMID: 38397434 PMCID: PMC10886936 DOI: 10.3390/biom14020197] [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: 11/17/2023] [Revised: 01/14/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Adversity during early life, a critical period for brain development, increases vulnerability and can have a lasting impact on the brain and behaviour of a child. However, the long-term effects of cumulative early-life stressors on brain and behaviour are not well known. We studied a 2-hit rat model of early-life adversity using maternal separation (MS) and immune activation (lipopolysaccharide (LPS)). Rat pups underwent MS for 15 (control) or 180 (MS) minutes per day from postnatal day (P)2-14 and were administered saline or LPS (intraperitoneal) on P3. Open-field (OFT) and object-place recognition tests were performed on rat offspring at P33-35 and P42-50, respectively. The pre-frontal cortex (PFC) and hippocampus were removed at the experimental endpoint (P52-55) for mRNA expression. MS induced anxiety-like behaviour in OFT in male and reduced locomotor activity in both male and female offspring. LPS induced a subtle decline in memory in the object-place recognition test in male offspring. MS increased glial fibrillary acidic protein (GFAP) and brain-derived neurotrophic factor expression in PFC and ionised calcium-binding adapter molecule-1 expression in male hippocampus. MS and LPS resulted in distinct behavioural phenotypes in a sex-specific manner. The combination of MS and LPS had a synergistic effect on the anxiety-like behaviour, locomotor activity, and GFAP mRNA expression outcomes.
Collapse
Affiliation(s)
- Bharti Biswas
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (B.B.); (V.E.)
- School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Valsamma Eapen
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia; (B.B.); (V.E.)
| | - Margaret J. Morris
- School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Nicole M. Jones
- School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW 2052, Australia
| |
Collapse
|
6
|
Wang L, Sha H, He X, Xie Y, Deng J, Chen J, Li G, Yang J. Neonatal IL-4 Over-Exposure is Accompanied by Macrophage Accumulation in Dura Mater After Instant Anti-inflammatory Cytokine Response in CSF. Cell Mol Neurobiol 2024; 44:18. [PMID: 38315435 PMCID: PMC10844484 DOI: 10.1007/s10571-023-01451-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/28/2023] [Indexed: 02/07/2024]
Abstract
Multiple studies have shown that clinical events resulting into neonatal IL-4 over-exposure, such as asthma in early life and food allergy, were associated with brain damage and that the neuroinflammation induced by them might lead to cognitive impairments, anxiety-/depressive-like behaviors. IL-4 is the most major elevated cytokine in periphery when these clinical events occur and peripheral IL-4 level positively correlates with the severity of those events. Our previous studies have verified that neonatal IL-4 over-exposure induced a delayed neuroinflammatory damage in rodents, which might have adverse implications for brain development and cognition. Neuroinflammation in brain parenchyma is often accompanied by changes in CSF cytokines levels. However, whether the cytokines levels in CSF change after neonatal IL-4 over-exposure is unknown. Here, we found a delayed pro-inflammatory cytokines response (higher IL-6, IL-1β and, TNF levels) in both hippocampus and CSF after an instant anti-inflammatory cytokine response in IL-4 over-exposed rats. Moreover, the pro-inflammatory cytokines response appeared earlier in CSF than in hippocampus. The level of each of the pro-inflammatory cytokines in CSF positively correlated with that in hippocampus at the age of postnatal day 42. More microglia numbers/activation and higher M-CSF level in the hippocampus in IL-4 over-exposed rats were also observed. Furthermore, there were more macrophages with inflammatory activation in dural mater of IL-4 over-exposed rats. In sum, neonatal IL-4 over-exposure in rats induces delayed inflammation in CSF, suggesting CSF examination may serve as a potential method in predicting delayed neuroinflammation in brain following neonatal IL-4 over-exposure.
Collapse
Affiliation(s)
- Ling Wang
- Grade 2019, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Haoran Sha
- Grade 2020, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoyi He
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Higher Education Mega Center, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Yinyin Xie
- Grade 2018, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Jiapeng Deng
- Grade 2018, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Jiexuan Chen
- Grade 2020, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Guoying Li
- Guangdong Medical Association, Guangzhou, 510180, Guangdong, China.
| | - Junhua Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Higher Education Mega Center, Guangzhou, 510006, Guangdong, People's Republic of China.
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Higher Education Mega Center, Guangzhou, 510006, Guangdong, People's Republic of China.
| |
Collapse
|
7
|
Tian X, Dong W, Zhou W, Yan Y, Lu L, Mi J, Cao Y, Sun Y, Zeng X. The polysaccharides from the fruits of Lycium barbarum ameliorate high-fat and high-fructose diet-induced cognitive impairment via regulating blood glucose and mediating gut microbiota. Int J Biol Macromol 2024; 258:129036. [PMID: 38151081 DOI: 10.1016/j.ijbiomac.2023.129036] [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: 09/29/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023]
Abstract
High-fat and high-fructose diet (HFFD) consumption can induce cognitive dysfunction and gut microbiota disorder. In the present study, the effects of the polysaccharides from the fruits of Lycium barbarum L. (LBPs) on HFFD-induced cognitive deficits and gut microbiota dysbiosis were investigated. The results showed that intervention of LBPs (200 mg/kg/day) for 14 weeks could significantly prevent learning and memory deficits in HFFD-fed mice, evidenced by a reduction of latency and increment of crossing parameters of platform quadrant in Morris water maze test. Moreover, oral administration of LBPs enhanced the expression of postsynaptic density protein 95 and brain-derived neurotrophic factor and reduced the activation of glial cells in hippocampus. Besides, LBPs treatment enriched the relative abundances of Allobaculum and Lactococcus and reduced the relative abundance of Proteobacteria in gut bacterial community of HFFD-fed mice, accompanied by increased levels of short-chain fatty acids (SCFAs) as well as expression of associated G protein-coupled receptors. Furthermore, LBPs intervention prevented insulin resistance, obesity and colonic inflammation. Finally, a significant correlation was observed among neuroinflammation associated parameters, gut microbiota and SCFAs through Pearson correlation analysis. Collectively, these findings suggested that the regulation of gut microbiota might be the potential mechanism of LBPs on preventing cognitive dysfunction induced by HFFD.
Collapse
Affiliation(s)
- Xinyi Tian
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Dong
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangting Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yamei Yan
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, Ningxia, China; National Wolfberry Engineering Research Center, Yinchuan 750002, Ningxia, China
| | - Lu Lu
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, Ningxia, China; National Wolfberry Engineering Research Center, Yinchuan 750002, Ningxia, China
| | - Jia Mi
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, Ningxia, China; National Wolfberry Engineering Research Center, Yinchuan 750002, Ningxia, China
| | - Youlong Cao
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, Ningxia, China; National Wolfberry Engineering Research Center, Yinchuan 750002, Ningxia, China
| | - Yi Sun
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
8
|
Zaongo SD, Harypursat V, Rashid F, Dahourou DL, Ouedraogo AS, Chen Y. Influence of HIV infection on cognition and overall intelligence in HIV-infected individuals: advances and perspectives. Front Behav Neurosci 2023; 17:1261784. [PMID: 37953826 PMCID: PMC10637382 DOI: 10.3389/fnbeh.2023.1261784] [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: 08/03/2023] [Accepted: 10/10/2023] [Indexed: 11/14/2023] Open
Abstract
It is now well understood that HIV-positive individuals, even those under effective ART, tend to develop a spectrum of cognitive, motor, and/or mood conditions which are contemporarily referred to as HIV-associated neurocognitive disorder (HAND), and which is directly related to HIV-1 infection and HIV-1 replication in the central nervous system (CNS). As HAND is known to induce difficulties associated with attention, concentration, and memory, it is thus legitimate and pertinent to speculate upon the possibility that HIV infection may well influence human cognition and intelligence. We therefore propose herein to review the concept of intelligence, the concept of cells of intelligence, the influence of HIV on these particular cells, and the evidence pointing to differences in observed intelligence quotient (IQ) scores between HIV-positive and HIV-negative individuals. Additionally, cumulative research evidence continues to draw attention to the influence of the gut on human intelligence. Up to now, although it is known that HIV infection profoundly alters both the composition and diversity of the gut microbiota and the structural integrity of the gut, the influence of the gut on intelligence in the context of HIV infection remains poorly described. As such, we also provide herein a review of the different ways in which HIV may influence human intelligence via the gut-brain axis. Finally, we provide a discourse on perspectives related to HIV and human intelligence which may assist in generating more robust evidence with respect to this issue in future studies. Our aim is to provide insightful knowledge for the identification of novel areas of investigation, in order to reveal and explain some of the enigmas related to HIV infection.
Collapse
Affiliation(s)
- Silvere D. Zaongo
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Vijay Harypursat
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Farooq Rashid
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Désiré Lucien Dahourou
- Département Biomédical/Santé Publique, Institut de Recherche en Sciences de la Santé/CNRST, Ouagadougou, Burkina Faso
| | - Abdoul-Salam Ouedraogo
- Centre Muraz, Bobo-Dioulasso, Burkina Faso
- Department of Bacteriology and Virology, Souro Sanou University Hospital, Bobo-Dioulasso, Burkina Faso
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
9
|
Morin C, Bokobza C, Fleiss B, Hill-Yardin EL, Van Steenwinckel J, Gressens P. Preterm Birth by Cesarean Section: The Gut-Brain Axis, a Key Regulator of Brain Development. Dev Neurosci 2023; 46:179-187. [PMID: 37717575 DOI: 10.1159/000534124] [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: 07/13/2022] [Accepted: 09/11/2023] [Indexed: 09/19/2023] Open
Abstract
Understanding the long-term functional implications of gut microbial communities during the perinatal period is a bourgeoning area of research. Numerous studies have revealed the existence of a "gut-brain axis" and the impact of an alteration of gut microbiota composition in brain diseases. Recent research has highlighted how gut microbiota could affect brain development and behavior. Many factors in early life such as the mode of delivery or preterm birth could lead to disturbance in the assembly and maturation of gut microbiota. Notably, global rates of cesarean sections (C-sections) have increased in recent decades and remain important when considering premature delivery. Both preterm birth and C-sections are associated with an increased risk of neurodevelopmental disorders such as autism spectrum disorders, with neuroinflammation a major risk factor. In this review, we explore links between preterm birth by C-sections, gut microbiota alteration, and neuroinflammation. We also highlight C-sections as a risk factor for developmental disorders due to alterations in the microbiome.
Collapse
Affiliation(s)
- Cécile Morin
- Université Paris Cité, Inserm, NeuroDiderot, Paris, France
- Hôpital Robert Debré, Assistance Publique, Hôpitaux de Paris (APHP), Paris, France
| | - Cindy Bokobza
- Université Paris Cité, Inserm, NeuroDiderot, Paris, France
| | - Bobbi Fleiss
- Université Paris Cité, Inserm, NeuroDiderot, Paris, France
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Victoria, Australia
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Victoria, Australia
| | | | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, Paris, France
- Hôpital Robert Debré, Assistance Publique, Hôpitaux de Paris (APHP), Paris, France
| |
Collapse
|
10
|
Stupak A, Kwaśniewski W. Evaluating Current Molecular Techniques and Evidence in Assessing Microbiome in Placenta-Related Health and Disorders in Pregnancy. Biomolecules 2023; 13:911. [PMID: 37371491 PMCID: PMC10296270 DOI: 10.3390/biom13060911] [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: 03/24/2023] [Revised: 05/25/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
The microbiome is of great interest due to its potential influence on the occurrence and treatment of some human illnesses. It may be regarded as disruptions to the delicate equilibrium that humans ordinarily maintain with their microorganisms or the microbiota in their environment. The focus of this review is on the methodologies and current understanding of the functional microbiome in pregnancy outcomes. We present how novel techniques bring new insights to the contemporary field of maternal-fetal medicine with a critical analysis. The maternal microbiome in late pregnancy has been extensively studied, although data on maternal microbial changes during the first trimester are rare. Research has demonstrated that, in healthy pregnancies, the origin of the placental microbiota is oral (gut) rather than vaginal. Implantation, placental development, and maternal adaptation to pregnancy are complex processes in which fetal and maternal cells interact. Microbiome dysbiosis or microbial metabolites are rising as potential moderators of antenatal illnesses related to the placenta, such as fetal growth restriction, preeclampsia, and others, including gestational diabetes and preterm deliveries. However, because of the presence of antimicrobial components, it is likely that the bacteria identified in placental tissue are (fragments of) bacteria that have been destroyed by the placenta's immune cells. Using genomic techniques (metagenomics, metatranscriptomics, and metaproteomics), it may be possible to predict some properties of a microorganism's genome and the biochemical (epigenetic DNA modification) and physical components of the placenta as its environment. Despite the results described in this review, this subject needs further research on some major and crucial aspects. The phases of an in utero translocation of the maternal gut microbiota to the fetus should be explored. With a predictive knowledge of the impacts of the disturbance on microbial communities that influence human health and the environment, genomics may hold the answer to the development of novel therapies for the health of pregnant women.
Collapse
Affiliation(s)
- Aleksandra Stupak
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, Staszica Str. 16, 20-081 Lublin, Poland
| | - Wojciech Kwaśniewski
- Department of Gynecological Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland
| |
Collapse
|
11
|
Tejkalová H, Jakob L, Kvasnová S, Klaschka J, Sechovcová H, Mrázek J, Páleníček T, Fliegerová KO. The influence of antibiotic treatment on the behavior and gut microbiome of adult rats neonatally insulted with lipopolysaccharide. Heliyon 2023; 9:e15417. [PMID: 37123951 PMCID: PMC10130227 DOI: 10.1016/j.heliyon.2023.e15417] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/02/2023] Open
Abstract
The present study investigated whether neonatal exposure to the proinflammatory endotoxin lipopolysaccharide (LPS) followed by an antibiotic (ATB)-induced dysbiosis in early adulthood could induce neurodevelopmental disorders-like behavioral changes in adult male rats. Combining these two stressors resulted in decreased weight gain, but no significant behavioral abnormalities were observed. LPS treatment resulted in adult rats' hypoactivity and induced anxiety-like behavior in the social recognition paradigm, but these behavioral changes were not exacerbated by ATB-induced gut dysbiosis. ATB treatment seriously disrupted the gut bacterial community, but dysbiosis did not affect locomotor activity, social recognition, and acoustic reactivity in adult rats. Fecal bacterial community analyses showed no differences between the LPS challenge exposed/unexposed rats, while the effect of ATB administration was decisive regardless of prior LPS exposure. ATB treatment resulted in significantly decreased bacterial diversity, suppression of Clostridiales and Bacteroidales, and increases in Lactobacillales, Enterobacteriales, and Burkholderiales. The persistent effect of LPS on some aspects of behavior suggests a long-term effect of early toxin exposure that was not observed in ATB-treated animals. However, an anti-inflammatory protective effect of ATB cannot be assumed because of the increased abundance of pro-inflammatory, potentially pathogenic bacteria (Proteus, Suttrella) and the elimination of the bacterial families Ruminococcaceae and Lachnospiraceae, which are generally considered beneficial for gut health.
Collapse
Affiliation(s)
- Hana Tejkalová
- National Institute of Mental Health; Klecany, Czech Republic
| | - Lea Jakob
- National Institute of Mental Health; Klecany, Czech Republic
- 3rd Faculty of Medicine, Charles University, Czech Republic
- Corresponding author. National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic,
| | - Simona Kvasnová
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Czech Republic
| | - Jan Klaschka
- Institute of Computer Science of the Czech Academy of Sciences, Czech Republic
| | - Hana Sechovcová
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Czech Republic
- Czech University of Life Sciences in Prague, Czech Republic
| | - Jakub Mrázek
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Czech Republic
| | - Tomáš Páleníček
- National Institute of Mental Health; Klecany, Czech Republic
- 3rd Faculty of Medicine, Charles University, Czech Republic
| | | |
Collapse
|
12
|
Breach MR, Lenz KM. Sex Differences in Neurodevelopmental Disorders: A Key Role for the Immune System. Curr Top Behav Neurosci 2023; 62:165-206. [PMID: 35435643 PMCID: PMC10286778 DOI: 10.1007/7854_2022_308] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sex differences are prominent defining features of neurodevelopmental disorders. Understanding the sex biases in these disorders can shed light on mechanisms leading to relative risk and resilience for the disorders, as well as more broadly advance our understanding of how sex differences may relate to brain development. The prevalence of neurodevelopmental disorders is increasing, and the two most common neurodevelopmental disorders, Autism Spectrum Disorder (ASD) and Attention-Deficit/Hyperactivity Disorder (ADHD) exhibit male-biases in prevalence rates and sex differences in symptomology. While the causes of neurodevelopmental disorders and their sex differences remain to be fully understood, increasing evidence suggests that the immune system plays a critical role in shaping development. In this chapter we discuss sex differences in prevalence and symptomology of ASD and ADHD, review sexual differentiation and immune regulation of neurodevelopment, and discuss findings from human and rodent studies of immune dysregulation and perinatal immune perturbation as they relate to potential mechanisms underlying neurodevelopmental disorders. This chapter will give an overview of how understanding sex differences in neuroimmune function in the context of neurodevelopmental disorders could lend insight into their etiologies and better treatment strategies.
Collapse
Affiliation(s)
- Michaela R Breach
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
13
|
Kaki S, DeRosa H, Timmerman B, Brummelte S, Hunter RG, Kentner AC. Developmental Manipulation-Induced Changes in Cognitive Functioning. Curr Top Behav Neurosci 2023; 63:241-289. [PMID: 36029460 PMCID: PMC9971379 DOI: 10.1007/7854_2022_389] [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] [Indexed: 10/15/2022]
Abstract
Schizophrenia is a complex neurodevelopmental disorder with as-yet no identified cause. The use of animals has been critical to teasing apart the potential individual and intersecting roles of genetic and environmental risk factors in the development of schizophrenia. One way to recreate in animals the cognitive impairments seen in people with schizophrenia is to disrupt the prenatal or neonatal environment of laboratory rodent offspring. This approach can result in congruent perturbations in brain physiology, learning, memory, attention, and sensorimotor domains. Experimental designs utilizing such animal models have led to a greatly improved understanding of the biological mechanisms that could underlie the etiology and symptomology of schizophrenia, although there is still more to be discovered. The implementation of the Research and Domain Criterion (RDoC) has been critical in taking a more comprehensive approach to determining neural mechanisms underlying abnormal behavior in people with schizophrenia through its transdiagnostic approach toward targeting mechanisms rather than focusing on symptoms. Here, we describe several neurodevelopmental animal models of schizophrenia using an RDoC perspective approach. The implementation of animal models, combined with an RDoC framework, will bolster schizophrenia research leading to more targeted and likely effective therapeutic interventions resulting in better patient outcomes.
Collapse
Affiliation(s)
- Sahith Kaki
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Holly DeRosa
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
- University of Massachusetts Boston, Boston, MA, USA
| | - Brian Timmerman
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University, Detroit, MI, USA
| | | | - Amanda C Kentner
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
14
|
Lima MN, Barbosa-Silva MC, Maron-Gutierrez T. Microglial Priming in Infections and Its Risk to Neurodegenerative Diseases. Front Cell Neurosci 2022; 16:878987. [PMID: 35783096 PMCID: PMC9240317 DOI: 10.3389/fncel.2022.878987] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/26/2022] [Indexed: 11/29/2022] Open
Abstract
Infectious diseases of different etiologies have been associated with acute and long-term neurological consequences. The primary cause of these consequences appears to be an inflammatory process characterized primarily by a pro-inflammatory microglial state. Microglial cells, the local effectors' cells of innate immunity, once faced by a stimulus, alter their morphology, and become a primary source of inflammatory cytokines that increase the inflammatory process of the brain. This inflammatory scenario exerts a critical role in the pathogenesis of neurodegenerative diseases. In recent years, several studies have shown the involvement of the microglial inflammatory response caused by infections in the development of neurodegenerative diseases. This has been associated with a transitory microglial state subsequent to an inflammatory response, known as microglial priming, in which these cells are more responsive to stimuli. Thus, systemic inflammation and infections induce a transitory state in microglia that may lead to changes in their state and function, making priming them for subsequent immune challenges. However, considering that microglia are long-lived cells and are repeatedly exposed to infections during a lifetime, microglial priming may not be beneficial. In this review, we discuss the relationship between infections and neurodegenerative diseases and how this may rely on microglial priming.
Collapse
Affiliation(s)
- Maiara N. Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Maria C. Barbosa-Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Yi SY, Pirasteh A, Wang J, Bradshaw T, Jeffery JJ, Barnett BR, Stowe NA, McMillan AB, Vivas EI, Rey FE, Yu JPJ. 18F-SynVesT-1 PET/MR Imaging of the Effect of Gut Microbiota on Synaptic Density and Neurite Microstructure: A Preclinical Pilot Study. FRONTIERS IN RADIOLOGY 2022; 2:895088. [PMID: 37492655 PMCID: PMC10365022 DOI: 10.3389/fradi.2022.895088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/04/2022] [Indexed: 07/27/2023]
Abstract
The gut microbiome profoundly influences brain structure and function. The gut microbiome is hypothesized to play a key role in the etiopathogenesis of neuropsychiatric and neurodegenerative illness; however, the contribution of an intact gut microbiome to quantitative neuroimaging parameters of brain microstructure and function remains unknown. Herein, we report the broad and significant influence of a functional gut microbiome on commonly employed neuroimaging measures of diffusion tensor imaging (DTI), neurite orientation dispersion and density (NODDI) imaging, and SV2A 18F-SynVesT-1 synaptic density PET imaging when compared to germ-free animals. In this pilot study, we demonstrate that mice, in the presence of a functional gut microbiome, possess higher neurite density and orientation dispersion and decreased synaptic density when compared to age- and sex-matched germ-free mice. Our results reveal the region-specific structural influences and synaptic changes in the brain arising from the presence of intestinal microbiota. Further, our study highlights important considerations for the development of quantitative neuroimaging biomarkers for precision imaging in neurologic and psychiatric illness.
Collapse
Affiliation(s)
- Sue Y. Yi
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin–Madison, Madison, WI, United States
| | - Ali Pirasteh
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - James Wang
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Tyler Bradshaw
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Justin J. Jeffery
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Brian R. Barnett
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin–Madison, Madison, WI, United States
| | - Nicholas A. Stowe
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Alan B. McMillan
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Eugenio I. Vivas
- Department of Bacteriology, University of Wisconsin–Madison, Madison, WI, United States
- Gnotobiotic Animal Core Facility, Biomedical Research Model Services, University of Wisconsin–Madison, Madison, WI, United States
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin–Madison, Madison, WI, United States
| | - John-Paul J. Yu
- Neuroscience Training Program, Wisconsin Institutes for Medical Research, University of Wisconsin–Madison, Madison, WI, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
16
|
Bodnar TS, Lee C, Wong A, Rubin I, Parfrey LW, Weinberg J. Evidence for long-lasting alterations in the fecal microbiota following prenatal alcohol exposure. Alcohol Clin Exp Res 2022; 46:542-555. [PMID: 35102585 PMCID: PMC9238389 DOI: 10.1111/acer.14784] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/23/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND There is growing evidence that the gut microbiota can be shaped by early-life experiences/exposures, with long-term consequences for brain, behavior, and health. Changes in the gut microbiota have also been identified in neurodevelopmental disorders including Autism Spectrum Disorder and schizophrenia. In contrast, no studies to date have investigated whether the gut microbiota is altered in individuals with Fetal Alcohol Spectrum Disorder (FASD), the neurodevelopmental disorder that results from prenatal alcohol exposure (PAE). The current study was designed to assess the impact of PAE on the fecal microbiota. METHODS We used a rodent model in which pregnant Sprague-Dawley rats were provided with an EtOH-containing diet or a control diet throughout gestation. Fecal samples were collected from adult male and female animals and 16s rRNA sequencing was performed. RESULTS Overall, PAE rats showed greater richness of bacterial species, with community structure investigations demonstrating distinct clustering by prenatal treatment. In addition, prenatal treatment and sex-specific alterations were observed for many specific microbes. For example, in males, Bacteroides and Bifidobacterium, and in females, Faecalitalea and Proteus, differed in abundance between PAE and control rats. CONCLUSIONS Taken together, these results show for the first time that PAE has a long-lasting and sex-specific impact on the fecal microbiota. Further research is needed that considers fetal microbiota in the development of new interventions in FASD.
Collapse
Affiliation(s)
- Tamara S. Bodnar
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada, V6T 1Z3
| | - Christopher Lee
- Department of Microbiology and Immunology, 2185 E Mall, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Athena Wong
- Department of Biology, 6270 University Blvd, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Ilan Rubin
- Department of Zoology, 6270 University Blvd, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Laura Wegener Parfrey
- Department of Botany and Biodiversity Research Centre, University of British Columbia, 109 – 2212 Main Mall, Vancouver, BC, Canada, V6T 1Z4
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada, V6T 1Z3
| |
Collapse
|
17
|
Adcock SJJ. Early Life Painful Procedures: Long-Term Consequences and Implications for Farm Animal Welfare. FRONTIERS IN ANIMAL SCIENCE 2021. [DOI: 10.3389/fanim.2021.759522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Farm animals routinely undergo painful husbandry procedures early in life, including disbudding and castration in calves and goat kids, tail docking and castration in piglets and lambs, and beak trimming in chicks. In rodents, inflammatory events soon after birth, when physiological systems are developing and sensitive to perturbation, can profoundly alter phenotypic outcomes later in life. This review summarizes the current state of research on long-term phenotypic consequences of neonatal painful procedures in rodents and farm animals, and discusses the implications for farm animal welfare. Rodents exposed to early life inflammation show a hypo-/hyper-responsive profile to pain-, fear-, and anxiety-inducing stimuli, manifesting as an initial attenuation in responses that transitions into hyperresponsivity with increasing age or cumulative stress. Neonatal inflammation also predisposes rodents to cognitive, social, and reproductive deficits, and there is some evidence that adverse effects may be passed to offspring. The outcomes of neonatal inflammation are modulated by injury etiology, age at the time of injury and time of testing, sex, pain management, and rearing environment. Equivalent research examining long-term phenotypic consequences of early life painful procedures in farm animals is greatly lacking, despite obvious implications for welfare and performance. Improved understanding of how these procedures shape phenotypes will inform efforts to mitigate negative outcomes through reduction, replacement, and refinement of current practices.
Collapse
|
18
|
Faruqui NA, Prium DH, Mowna SA, Ullah MA, Araf Y, Sarkar B, Zohora US, Rahman MS. Gut microorganisms and neurological disease perspectives. FUTURE NEUROLOGY 2021. [DOI: 10.2217/fnl-2020-0026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract of every healthy human consists of a unique set of gut microbiota that collectively harbors a diverse and complex community of over 100 trillion microorganisms, including bacteria, viruses, archaea, protozoa and fungi. Gut microbes have a symbiotic relationship with our body. The composition of the microbiota is shaped early in life by gut maturation, which is influenced by several factors. Intestinal bacteria are crucial in maintaining immune and metabolic homeostasis and protecting against pathogens. Dysbiosis of gut microbiota is associated not only with intestinal disorders but also with extraintestinal diseases such as metabolic and neurological disorders. In this review, the authors examine different studies that have revealed the possible hypotheses and links in the development of neurological disorders associated with the gut microbiome.
Collapse
Affiliation(s)
- Nairita Ahsan Faruqui
- Department of Mathematics and Natural Sciences, Biotechnology Program, School of Data & Sciences, BRAC University, Dhaka, Bangladesh
| | - Durdana Hossain Prium
- Department of Mathematics and Natural Sciences, Biotechnology Program, School of Data & Sciences, BRAC University, Dhaka, Bangladesh
| | - Sadrina Afrin Mowna
- Department of Mathematics and Natural Sciences, Biotechnology Program, School of Data & Sciences, BRAC University, Dhaka, Bangladesh
| | - Md. Asad Ullah
- Department of Biotechnology & Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering & Biotechnology, School of Life Sciences, Shahjalal University of Science & Technology, Sylhet, Bangladesh
| | - Bishajit Sarkar
- Department of Biotechnology & Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Dhaka, Bangladesh
| | - Umme Salma Zohora
- Department of Biotechnology & Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Dhaka, Bangladesh
| | - Mohammad Shahedur Rahman
- Department of Biotechnology & Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Dhaka, Bangladesh
| |
Collapse
|
19
|
O'Connor TG, Ciesla AA. Maternal Immune Activation Hypotheses for Human Neurodevelopment: Some Outstanding Questions. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:471-479. [PMID: 34688920 PMCID: PMC9021321 DOI: 10.1016/j.bpsc.2021.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
The Maternal Immune Activation (MIA) hypothesis is a leading model for understanding prenatal influences on individual differences in, and clinical syndromes of, neurodevelopment. Experimental animal and human research has proliferated in recent years, and there is now a sizable research base. Several meta-analyses demonstrate general support for an association between prenatal immune activation and neurodevelopment in human research. However, questions remain about the nature of the immune activation, the network of underlying mechanisms involved, and the breadth of impact across behavioral phenotypes. Complementing recent reviews of results, the current review places particular emphasis on how advances in understanding mechanisms may be improved with greater attention to addressing the methodological variation and limitations of existing studies, and identifies areas for further clinical research.
Collapse
Affiliation(s)
- Thomas G O'Connor
- Department of Psychiatry, University of Rochester; Department of Psycholog, University of Rochestery; Department of Neuroscience, University of Rochester; Department of Obstetrics and Gynecology, University of Rochester; Wynne Center for Family Research, University of Rochester.
| | | |
Collapse
|
20
|
Bairwa SC, Shaw CA, Kuo M, Yoo J, Tomljenovic L, Eidi H. Cytokines profile in neonatal and adult wild-type mice post-injection of U. S. pediatric vaccination schedule. Brain Behav Immun Health 2021; 15:100267. [PMID: 34589773 PMCID: PMC8474652 DOI: 10.1016/j.bbih.2021.100267] [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: 01/08/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 11/09/2022] Open
Abstract
Introduction A recent study from our laboratory demonstrated a number of neurobehavioral abnormalities in mice colony injected with a mouse-weight equivalent dose of all vaccines that are administered to infants in their first 18 months of life according to the U. S. pediatric vaccination schedule. Cytokines have been studied extensively as blood immune and inflammatory biomarkers, and their association with neurodevelopmental disorders. Given the importance of cytokines in early neurodevelopment, we aimed to investigate the potential post-administration effects of the U. S. pediatric vaccines on circulatory cytokines in a mouse model. In the current study, cytokines have been assayed at early and late time points in mice vaccinated early in postnatal life and compared with placebo controls. Materials and methods Newborn mouse pups were divided into three groups: i) vaccine (V1), ii) vaccine × 3 (V3) and iii) placebo control. V1 group was injected with mouse weight-equivalent of the current U. S. pediatric vaccine schedule. V3 group was injected with same vaccines but at triple the dose and the placebo control was injected with saline. Pups were also divided according to the sampling age into two main groups: acute- and chronic-phase group. Blood samples were collected at postnatal day (PND) 23, two days following vaccine schedule for the acute-phase group or at 67 weeks post-vaccination for the chronic-phase groups. Fifteen cytokines were analyzed: GM-CSF, IFN-γ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12p70, IL-13, IL-17A, MCP-1, TNF-α, and VEGF-A. Wilcoxon Rank Sum test or unpaired Student's t-test was performed where applicable. Results IL-5 levels in plasma were significantly elevated in the V1 and V3 group compared with the control only in the acute-phase group. The elevation of IL-5 levels in the two vaccine groups were significant irrespective of whether the sexes were combined or analyzed separately. Other cytokines (VEGF-A, TNF-α, IL-10, MCP-1, GM-CSF, IL-6, and IL-13) were also impacted, although to a lesser extent and in a sex-dependent manner. In the acute-phase group, females showed a significant increase in IL-10 and MCP-1 levels and a decrease in VEGF-A levels in both V1 and V3 group compared to controls. In the acute-phase, a significant increase in MCP-1 levels in V3 group and CM-CSF levels in V1 and V3 group and decrease in TNF-α levels in V1 group were observed in treated males as compared with controls. In chronic-phase females, levels of VEGF-A in V1 and V3 group, TNF-α in V3 group, and IL-13 in V1 group were significantly decreased in contrast with controls. In chronic-phase males, TNF-α levels were significantly increased in V1 group and IL-6 levels decreased in V3 group in comparison to controls. The changes in levels of most tested cytokines were altered between the early and the late postnatal assays. Conclusions IL-5 levels significantly increased in the acute-phase of the treatment in the plasma of both sexes that were subjected to V1 and V3 injections. These increases had diminished by the second test assayed at week 67. These results suggest that a profound, albeit transient, effect on cytokine levels may be induced by the whole vaccine administration supporting our recently published observations regarding the behavioral abnormalities in the same mice. These observations support the view that the administration of whole pediatric vaccines in a neonatal period may impact at least short-term CNS functions in mice.
Collapse
Affiliation(s)
- S C Bairwa
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - C A Shaw
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada.,Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada.,Program in Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - M Kuo
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - J Yoo
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - L Tomljenovic
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - H Eidi
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada.,French Agency for Veterinary Medicinal Products (ANMV) - French Agency for Food, Environmental and Occupational Health Safety (ANSES), Fougères, France
| |
Collapse
|
21
|
Gomes CI, Barr GA. Local injury and systemic infection in infants alter later nociception and pain affect during early life and adulthood. Brain Behav Immun Health 2021; 9:100175. [PMID: 34589906 PMCID: PMC8474633 DOI: 10.1016/j.bbih.2020.100175] [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: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 10/25/2022] Open
Abstract
Newborns in intensive care are regularly exposed to minor painful procedures at developmental time points when noxious stimulation would be normally absent. Pain from these interventions is inconsistently treated and often exists concurrently with systemic infection, a common comorbidity of prematurity. Our understanding of the independent and combined effects of early painful experiences and infection on pain response is incomplete. The main goals of this research therefore were to understand how pain and infection experienced early in life influence future nociceptive and affective responses to painful stimuli. Rat pups were infected with E-coli on postnatal day 2 (PN2) and had left hind paw injury with carrageenan on PN3. Standard thermal tests for acute pain, formalin tests for inflammatory pain, and conditioned place aversion testing were performed at different ages to assess the nociceptive and affective components of the pain response. Early E-coli infection and early inflammatory injury with carrageenan both independently increased pain scores following hind paw reinjury with formalin on PN8, with effects persisting into adulthood in the carrageenan exposed group. When experienced concurrently, early E-coli infection and carrageenan exposure also increased conditioned aversion to pain in adults. Effect of sex was significant only in formalin testing, with males showing higher pain scores in infancy and females showing higher pain scores as adults. These findings demonstrate that infection experienced early in life can alter both the nociceptive and affective components of the pain response and that there is a cumulative effect of local and systemic pro-inflammatory processes on the aversive component of pain.
Collapse
Affiliation(s)
- Carly I Gomes
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gordon A Barr
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, 3615 Civic Center Boulevard, Philadelphia, PA, 19104, USA.,Department of Psychology, University of Pennsylvania, 425 S. University Avenue, Stephen A. Levin Building, Philadelphia, PA, USA
| |
Collapse
|
22
|
Smith BL. Improving translational relevance: The need for combined exposure models for studying prenatal adversity. Brain Behav Immun Health 2021; 16:100294. [PMID: 34589787 PMCID: PMC8474200 DOI: 10.1016/j.bbih.2021.100294] [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: 05/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Prenatal environmental adversity is a risk factor for neurodevelopmental disorders (NDDs), with the neuroimmune environment proposed to play a role in this risk. Adverse maternal exposures are associated with cognitive consequences in the offspring that are characteristics of NDDs and simultaneous neuroimmune changes that may underlie NDD risk. In both animal models and human studies the association between prenatal environmental exposure and NDD risk has been shown to be complex. Maternal overnutrition/obesity and opioid use are two different examples of complex exposure epidemics, each with their own unique comorbidities. This review will examine maternal obesity and maternal opioid use separately, illustrating the pervasive comorbidities with each exposure to argue a need for animal models of compound prenatal exposures. Many of these comorbidities can impact neuroimmune function, warranting systematic investigation of combined exposures to begin to understand this complexity. While traditional approaches in animal models have focused on modeling a single prenatal exposure or second exposure later in life, a translational approach would begin to incorporate the most prevalent co-occurring prenatal exposures. Long term follow-up in humans is extremely challenging, so animal models can provide timely insight into neurodevelopmental consequences of complex prenatal exposures. Animal models that represent this translational context of comorbid exposures behind maternal obesity or comorbid exposures behind maternal opioid use may reveal potential synergistic neuroimmune interactions that contribute to cognitive consequences and NDD risk. Finally, translational co-exposure models can identify concerning exposure combinations to guide treatment in complex cases, and identify high risk children starting in the prenatal period where early interventions improve prognosis.
Collapse
Affiliation(s)
- Brittany L. Smith
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
23
|
Snigdha S, Ha K, Tsai P, Dinan TG, Bartos JD, Shahid M. Probiotics: Potential novel therapeutics for microbiota-gut-brain axis dysfunction across gender and lifespan. Pharmacol Ther 2021; 231:107978. [PMID: 34492236 DOI: 10.1016/j.pharmthera.2021.107978] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/21/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
Probiotics are live microorganisms, which when administered in adequate amounts, present a health benefit for the host. While the beneficial effects of probiotics on gastrointestinal function are generally well recognized, new animal research and clinical studies have found that alterations in gut microbial communities can have a broad range of effects throughout the body. Non-intestinal sites impacted include the immune, endocrine, cardiovascular and the central nervous system (CNS). In particular, there has been a growing interest and appreciation about the role that gut microbiota may play in affecting CNS-related function through the 'microbiota-gut-brain axis'. Emerging evidence suggests potential therapeutic benefits of probiotics in several CNS conditions, such as anxiety, depression, autism spectrum disorders and Parkinson's disease. There may also be some gender-specific variances in terms of probiotic mediated effects, with the gut microbiota shaping and being concurrently molded by the hormonal environment governing differences between the sexes. Probiotics may influence the ability of the gut microbiome to affect a variety of biological processes in the host, including neurotransmitter activity, vagal neurotransmission, generation of neuroactive metabolites and inflammatory response mediators. Some of these may engage in cross talk with host sex hormones, such as estrogens, which could be of relevance in relation to their effects on stress response and cognitive health. This raises the possibility of gender-specific variation with regards to the biological action of probiotics, including that on the endocrine and central nervous systems. In this review we aim to describe the current understanding in relation to the role and use of probiotics in microbiota-gut-brain axis-related dysfunction. Furthermore, we will address the conceptualization and classification of probiotics in the context of gender and lifespan as well as how restoring gut microbiota composition by clinical or dietary intervention can help in supporting health outcomes other than those related to the gastrointestinal tract. We also evaluate how these new learnings may impact industrial effort in probiotic research and the discovery and development of novel and more personalized, condition-specific, beneficial probiotic therapeutic agents.
Collapse
Affiliation(s)
| | - Kevin Ha
- MeriCal, 233 E Bristol St., Orange, CA, USA
| | - Paul Tsai
- MeriCal, 233 E Bristol St., Orange, CA, USA
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | | | | |
Collapse
|
24
|
Strzelewicz AR, Vecchiarelli HA, Rondón-Ortiz AN, Raneri A, Hill MN, Kentner AC. Interactive effects of compounding multidimensional stressors on maternal and male and female rat offspring outcomes. Horm Behav 2021; 134:105013. [PMID: 34171577 PMCID: PMC8403628 DOI: 10.1016/j.yhbeh.2021.105013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 10/21/2022]
Abstract
Exposure to adverse childhood experiences (ACEs) is a risk factor for the development of psychiatric disorders in addition to cardiovascular associated diseases. This risk is elevated when the cumulative burden of ACEs is increased. Laboratory animals can be used to model the changes (as well as the underlying mechanisms) that result in response to adverse events. In this study, using male and female Sprague Dawley rats, we examined the impact of increasing stress burden, utilizing both two adverse early life experiences (parental/offspring high fat diet + limited bedding exposure) and three adverse early life experiences (parental/offspring high fat diet + limited bedding exposure + neonatal inflammation), on maternal care quality and offspring behavior. Additionally, we measured hormones and hippocampal gene expression related to stress. We found that the adverse perinatal environment led to a compensatory increase in maternal care. Moreover, these dams had reduced maternal expression of oxytocin receptor, compared to standard housed dams, in response to acute stress on postnatal day (P)22. In offspring, the two-hit and three-hit models resulted in a hyperlocomotor phenotype and increased body weights. Plasma leptin and hippocampal gene expression of corticotropin releasing hormone (Chrh)1 and Crhr2 were elevated (males) while expression of oxytocin was reduced (females) following acute stress. On some measures (e.g., hyperlocomotion, leptin), the magnitude of change was lower in the three-hit compared to the two-hit model. This suggests that multiple early adverse events can have interactive, and often unpredictable, impacts, highlighting the importance of modeling complex interactions amongst stressors during development.
Collapse
Affiliation(s)
- Arielle R Strzelewicz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Haley A Vecchiarelli
- Divisions of Medical Sciences, University of Victoria, BC V8P 5C2, Canada; Neuroscience Graduate Program, Hotchkiss Brain Institute, Mathison Centre for Mental Health, Research and Education, Cumming School of Medicine, University of Calgary, AB T2N 1N4, Canada
| | - Alejandro N Rondón-Ortiz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Anthony Raneri
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Matthew N Hill
- Neuroscience Graduate Program, Hotchkiss Brain Institute, Mathison Centre for Mental Health, Research and Education, Cumming School of Medicine, University of Calgary, AB T2N 1N4, Canada
| | - Amanda C Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States.
| |
Collapse
|
25
|
Zhao X, Tran H, DeRosa H, Roderick RC, Kentner AC. Hidden talents: Poly (I:C)-induced maternal immune activation improves mouse visual discrimination performance and reversal learning in a sex-dependent manner. GENES BRAIN AND BEHAVIOR 2021; 20:e12755. [PMID: 34056840 DOI: 10.1111/gbb.12755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/13/2022]
Abstract
While there is a strong focus on the negative consequences of maternal immune activation (MIA) on developing brains, very little attention is directed towards potential advantages of early life challenges. In this study, we utilized a polyinosine-polycytidylic acid (poly(I:C)) MIA model to test visual pairwise discrimination (PD) and reversal learning (RL) in mice using touchscreen technology. Significant sex differences emerged in that MIA reduced the latency for males to make a correct choice in the PD task while females reached criterion sooner, made fewer errors, and utilized fewer correction trials in RL compared to saline controls. These surprising improvements were accompanied by the sex-specific upregulation of several genes critical to cognitive functioning, indicative of compensatory plasticity in response to MIA. In contrast, when exposed to a 'two-hit' stress model (MIA + loss of the social component of environmental enrichment [EE]), mice did not display anhedonia but required an increased number of PD and RL correction trials. These animals also had significant reductions of CamK2a mRNA in the prefrontal cortex. Appropriate functioning of synaptic plasticity, via mediators such as this protein kinase and others, are critical for behavioral flexibility. Although EE has been implicated in, delaying the appearance of symptoms associated with certain brain disorders, these findings are in line with evidence that it also makes individuals more vulnerable to its loss. Overall, with the right 'dose', early life stress exposure can confer at least some functional advantages, which are lost when the number or magnitude of these exposures become too great.
Collapse
Affiliation(s)
- Xin Zhao
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, Massachusetts, USA
| | - Hieu Tran
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, Massachusetts, USA
| | - Holly DeRosa
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, Massachusetts, USA
| | - Ryland C Roderick
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, Massachusetts, USA
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Nagu P, Parashar A, Behl T, Mehta V. Gut Microbiota Composition and Epigenetic Molecular Changes Connected to the Pathogenesis of Alzheimer's Disease. J Mol Neurosci 2021; 71:1436-1455. [PMID: 33829390 DOI: 10.1007/s12031-021-01829-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, and its pathogenesis is not fully known. Although there are several hypotheses, such as neuroinflammation, tau hyperphosphorylation, amyloid-β plaques, neurofibrillary tangles, and oxidative stress, none of them completely explain the origin and progression of AD. Emerging evidence suggests that gut microbiota and epigenetics can directly influence the pathogenesis of AD via their effects on multiple pathways, including neuroinflammation, oxidative stress, and amyloid protein. Various gut microbes such as Actinobacteria, Bacteroidetes, E. coli, Firmicutes, Proteobacteria, Tenericutes, and Verrucomicrobia are known to play a crucial role in the pathogenesis of AD. These microbes and their metabolites modulate various physiological processes that contribute to AD pathogenesis, such as neuroinflammation and other inflammatory processes, amyloid deposition, cytokine storm syndrome, altered BDNF and NMDA signaling, impairing neurodevelopmental processes. Likewise, epigenetic markers associated with AD mainly include histone modifications and DNA methylation, which are under the direct control of a variety of enzymes, such as acetylases and methylases. The activity of these enzymes is dependent upon the metabolites generated by the host's gut microbiome, suggesting the significance of epigenetics in AD pathogenesis. It is interesting to know that both gut microbiota and epigenetics are dynamic processes and show a high degree of variation according to diet, stressors, and environmental factors. The bidirectional relation between the gut microbiota and epigenetics suggests that they might work in synchrony to modulate AD representation, its pathogenesis, and progression. They both also provide numerous targets for early diagnostic biomarkers and for the development of AD therapeutics. This review discusses the gut microbiota and epigenetics connection in the pathogenesis of AD and aims to highlight vast opportunities for diagnosis and therapeutics of AD.
Collapse
Affiliation(s)
- Priyanka Nagu
- Department of Pharmaceutics, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.,Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India
| | - Arun Parashar
- Faculty of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Department of Pharmacology, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.
| |
Collapse
|
27
|
Neonatal proinflammatory challenge evokes a microglial response and affects the ratio between subtypes of GABAergic interneurons in the hippocampus of juvenile rats: sex-dependent and sex-independent effects. Brain Struct Funct 2021; 226:563-574. [DOI: 10.1007/s00429-020-02199-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
|
28
|
Bangma JT, Hartwell H, Santos HP, O'Shea TM, Fry RC. Placental programming, perinatal inflammation, and neurodevelopment impairment among those born extremely preterm. Pediatr Res 2021; 89:326-335. [PMID: 33184498 PMCID: PMC7658618 DOI: 10.1038/s41390-020-01236-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/02/2020] [Accepted: 10/08/2020] [Indexed: 01/30/2023]
Abstract
Individuals born extremely preterm are at significant risk for impaired neurodevelopment. After discharge from the neonatal intensive care, associations between the child's well-being and factors in the home and social environment become increasingly apparent. Mothers' prenatal health and socioeconomic status are associated with neurodevelopmental outcomes, and emotional and behavioral problems. Research on early life risk factors and on mechanisms underlying inter-individual differences in neurodevelopment later in life can inform the design of personalized approaches to prevention. Here, we review early life predictors of inter-individual differences in later life neurodevelopment among those born extremely preterm. Among biological mechanisms that mediate relationships between early life predictors and later neurodevelopmental outcomes, we highlight evidence for disrupted placental processes and regulated at least in part via epigenetic mechanisms, as well as perinatal inflammation. In relation to these mechanisms, we focus on four prenatal antecedents of impaired neurodevelopment, namely, (1) fetal growth restriction, (2) maternal obesity, (3) placental microorganisms, and (4) socioeconomic adversity. In the future, this knowledge may inform efforts to detect and prevent adverse outcomes in infants born extremely preterm. IMPACT: This review highlights early life risk factors and mechanisms underlying inter-individual differences in neurodevelopment later in life. The review emphasizes research on early life risk factors (fetal growth restriction, maternal obesity, placental microorganisms, and socioeconomic adversity) and on mechanisms (disrupted placental processes and perinatal inflammation) underlying inter-individual differences in neurodevelopment later in life. The findings highlighted here may inform efforts to detect and prevent adverse outcomes in infants born extremely preterm.
Collapse
Affiliation(s)
- Jacqueline T Bangma
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hadley Hartwell
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hudson P Santos
- Biobehavioral Laboratory, School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - T Michael O'Shea
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Biobehavioral Laboratory, School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
29
|
Savi FF, de Oliveira A, de Medeiros GF, Bozza FA, Michels M, Sharshar T, Dal-Pizzol F, Ritter C. What animal models can tell us about long-term cognitive dysfunction following sepsis: A systematic review. Neurosci Biobehav Rev 2020; 124:386-404. [PMID: 33309906 DOI: 10.1016/j.neubiorev.2020.12.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 01/28/2023]
Abstract
Survivors of sepsis often develop long-term cognitive impairments. This review aimed at exploring the results of the behavioral tools and tests which have been used to evaluate cognitive dysfunction in different animal models of sepsis. Two independent investigators searched for sepsis- and cognition-related keywords. 6323 publications were found, of which 355 were selected based on their title, and 226 of these were chosen based on manuscript review. LPS was used to induce sepsis in 171 studies, while CLP was used in 55 studies. Inhibitory avoidance was the most widely used method for assessing aversive memory, followed by fear conditioning and continuous multi-trial inhibitory avoidance. With regard to non-aversive memory, most studies used the water maze, open-field, object recognition, Y-maze, plus maze, and radial maze tests. Both CLP and LPS models of sepsis were effective in inducing short- and long-term behavioral impairment. Our findings help elucidate the mechanisms involved in the pathophysiology of sepsis-induced cognitive changes, as well as the available methods and tests used to study this in animal models.
Collapse
Affiliation(s)
- Felipe Figueredo Savi
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil
| | - Alexandre de Oliveira
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil
| | | | - Fernando Augusto Bozza
- Laboratório de Medicina Intensiva, Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Monique Michels
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil
| | - Tarek Sharshar
- Laboratoire de Neuropathologie Expérimentale, Institut Pasteur, Paris, France; Department of Neuro-Intensive Care Medicine, Sainte-Anne Hospital, Paris-Descartes University, Paris, France
| | - Felipe Dal-Pizzol
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil; Laboratoire de Neuropathologie Expérimentale, Institut Pasteur, Paris, France
| | - Cristiane Ritter
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil.
| |
Collapse
|
30
|
Veerasammy S, Van Steenwinckel J, Le Charpentier T, Seo JH, Fleiss B, Gressens P, Levison SW. Perinatal IL-1β-induced inflammation suppresses Tbr2 + intermediate progenitor cell proliferation in the developing hippocampus accompanied by long-term behavioral deficits. Brain Behav Immun Health 2020; 7:100106. [PMID: 34589867 PMCID: PMC8474668 DOI: 10.1016/j.bbih.2020.100106] [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: 07/07/2020] [Accepted: 07/12/2020] [Indexed: 12/12/2022] Open
Abstract
Meta-analyses have revealed associations between the incidence of maternal infections during pregnancy, premature birth, smaller brain volumes, and subsequent cognitive, motor and behavioral deficits as these children mature. Inflammation during pregnancy in rodents produces cognitive and behavioral deficits in the offspring that are similar to those reported in human studies. These deficits are accompanied by decreased neurogenesis and proliferation in the subgranular zone (SGZ) of the dentate gyrus (DG) of the hippocampus. As systemically administering interleukin-1 β (IL-1β) to neonatal mice recapitulates many of the brain abnormalities seen in premature babies including developmental delays, the goal of this study was to determine whether IL-1-mediated neuroinflammation would affect hippocampal growth during development to produce cognitive and behavioral abnormalities. For these studies, 10 ng/g IL-1β was administered twice daily to Swiss Webster mice during the first 5 days of life, which increased hippocampal levels of IL-1α and acutely reduced the proliferation of Tbr2+ neural progenitors in the DG. In vitro, both IL-1α and IL-1β produced G1/S cell cycle arrest that resulted in reduced progenitor cell proliferation within the transit amplifying progenitor cell cohort. By contrast, IL-1β treatment increased neural stem cell frequency. Upon terminating IL-1β treatment, the progenitor cell pool regained its proliferative capacity. An earlier study that used this in vivo model of perinatal inflammation showed that mice that received IL-1β as neonates displayed memory deficits which suggested abnormal hippocampal function. To evaluate whether other cognitive and behavioral traits associated with hippocampal function would also be altered, mice were tested in tasks designed to assess exploratory and anxiety behavior as well as working and spatial memory. Interestingly, mice that received IL-1β as neonates showed signs of anxiety in several behavioral assays during adolescence that were also evident in adulthood. Additionally, these mice did not display working memory deficits in adulthood, but they did display deficits in long-term spatial memory. Altogether, these data support the view that perinatal inflammation negatively affects the developing hippocampus by producing behavioral deficits that persist into adulthood. These data provide a new perspective into the origin of the cognitive and behavioral impairments observed in prematurely-born sick infants.
Collapse
Affiliation(s)
- Stephanie Veerasammy
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University, New Jersey Medical School, Cancer Center, 205 South Orange Avenue, Newark, NJ, 07103, USA
| | | | - Tifenn Le Charpentier
- Université de Paris, NeuroDiderot, Inserm, F-75019, Paris, France
- PremUP, F-75006, Paris, France
| | - Joon Ho Seo
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University, New Jersey Medical School, Cancer Center, 205 South Orange Avenue, Newark, NJ, 07103, USA
| | - Bobbi Fleiss
- Université de Paris, NeuroDiderot, Inserm, F-75019, Paris, France
- PremUP, F-75006, Paris, France
- School of Health and Biomedical Sciences, RMIT University, Bundoora, 3083, VIC, Australia
| | - Pierre Gressens
- Université de Paris, NeuroDiderot, Inserm, F-75019, Paris, France
- PremUP, F-75006, Paris, France
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London, SE1 7EH, UK
| | - Steven W. Levison
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University, New Jersey Medical School, Cancer Center, 205 South Orange Avenue, Newark, NJ, 07103, USA
| |
Collapse
|
31
|
Pajoohesh-Ganji A, Karl M, Garrison E, Osei-Bonsu NA, Clarkson-Paredes C, Ahn J, Miller RH. Developmental ablation of mature oligodendrocytes exacerbates adult CNS demyelination. Brain Behav Immun Health 2020; 7:100110. [PMID: 34589870 PMCID: PMC8474627 DOI: 10.1016/j.bbih.2020.100110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 07/19/2020] [Indexed: 11/28/2022] Open
Abstract
Multiple sclerosis (MS) is a CNS neurodegenerative autoimmune disease characterized by loss of oligodendrocytes and myelin in the brain and the spinal cord that results in localized functional deficits. Several risk factors have been associated with MS, however none fully explain the enhanced susceptibility seen in older individuals. Epidemiological data, based on geographical prevalence studies suggest that susceptibility is established early in life and frequently long before the diagnosis of disease raising the possibility that developmental events influence adult disease onset and progression. Here we test the hypothesis that selective loss of mature oligodendrocytes during postnatal development results in enhanced susceptibility to a demyelinating insult to the mature CNS. A transgenic mouse model was utilized to specifically induce apoptotic cell death in a subset of mature oligodendrocytes (MBP-iCP9) during the first 2 postnatal weeks followed by either a local LPC spinal cord injection or the induction of EAE in the adult animal. Immunostaining, immunoblotting, behavioral testing, and electron microscopy were utilized to examine the differences in the response between animals with developmental loss of oligodendrocytes and controls. We show that during development, oligodendrocyte apoptosis results in transient reductions in myelination and functional deficits that recover after 10–14 days. Compared to animals in which oligodendrocyte development was unperturbed, animals subjected to postnatal oligodendrocyte loss showed delayed recovery from an LPC lesion to the mature spinal cord. Unexpectedly, the induction and severity of MOG induced EAE was not significantly altered in animals following oligodendrocyte developmental loss even though there was a substantial increase in spinal cord tissue damage and CNS inflammation. It is unclear why the elevated glial responses seen in developmentally compromised animals were not reflected in enhanced functional deficits. These observations suggest that developmental loss of oligodendrocytes results in long lasting tissue changes that alter its response to subsequent insults and the capacity for repair in the adult. Developmental oligodendrocyte apoptosis induces transient myelin and functional loss. Developmental loss of oligodendrocytes compromises adult remyelination after LPC. Developmental loss of oligodendrocytes enhances CNS immune response in EAE. Elevated CNS glial reactivity does not alter course of EAE.
Collapse
Affiliation(s)
- Ahdeah Pajoohesh-Ganji
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, United States
| | - Molly Karl
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, United States
| | - Eric Garrison
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, United States
| | - Nana Adwoa Osei-Bonsu
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, United States
| | - Cheryl Clarkson-Paredes
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, United States
| | - Julie Ahn
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, United States
| | - Robert H Miller
- Department of Anatomy and Cell Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
32
|
Newville J, Maxwell JR, Kitase Y, Robinson S, Jantzie LL. Perinatal Opioid Exposure Primes the Peripheral Immune System Toward Hyperreactivity. Front Pediatr 2020; 8:272. [PMID: 32670993 PMCID: PMC7332770 DOI: 10.3389/fped.2020.00272] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/29/2020] [Indexed: 11/29/2022] Open
Abstract
The increased incidence of opioid use during pregnancy warrants investigation to reveal the impact of opioid exposure on the developing fetus. Exposure during critical periods of development could have enduring consequences for affected individuals. Particularly, evidence is mounting that developmental injury can result in immune priming, whereby subsequent immune activation elicits an exaggerated immune response. This maladaptive hypersensitivity to immune challenge perpetuates dysregulated inflammatory signaling and poor health outcomes. Utilizing an established preclinical rat model of perinatal methadone exposure, we sought to investigate the consequences of developmental opioid exposure on in vitro activation of peripheral blood mononuclear cells (PBMCs). We hypothesize that PBMCs from methadone-exposed rats would exhibit abnormal chemokine and cytokine expression at baseline, with exaggerated chemokine and cytokine production following immune stimulation compared to saline-exposed controls. On postnatal day (P) 7, pup PMBCs were isolated and cultured, pooling three pups per n. Following 3 and 24 h, the supernatant from cultured PMBCs was collected and assessed for inflammatory cytokine and chemokine expression at baseline or lipopolysaccharide (LPS) stimulation using multiplex electrochemiluminescence. Following 3 and 24 h, baseline production of proinflammatory chemokine and cytokine levels were significantly increased in methadone PBMCs (p < 0.0001). Stimulation with LPS for 3 h resulted in increased tumor necrosis factor (TNF-α) and C-X-C motif chemokine ligand 1 (CXCL1) expression by 3.5-fold in PBMCs from methadone-exposed PBMCs compared to PBMCs from saline-exposed controls (p < 0.0001). Peripheral blood mononuclear cell hyperreactivity was still apparent at 24 h of LPS stimulation, evidenced by significantly increased TNF-α, CXCL1, interleukin 6 (IL-6), and IL-10 production by methadone PMBCs compared to saline control PBMCs (p < 0.0001). Together, we provide evidence of increased production of proinflammatory molecules from methadone PBMCs at baseline, in addition to sustained hyperreactivity relative to saline-exposed controls. Exaggerated peripheral immune responses exacerbate inflammatory signaling, with subsequent consequences on many organ systems throughout the body, such as the developing nervous system. Enhanced understanding of these inflammatory mechanisms will allow for appropriate therapeutic development for infants who were exposed to opioids during development. Furthermore, these data highlight the utility of this in vitro PBMC assay technique for future biomarker development to guide specific treatment for patients exposed to opioids during gestation.
Collapse
Affiliation(s)
- Jessie Newville
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jessie R. Maxwell
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
- Departments of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Yuma Kitase
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lauren L. Jantzie
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Kennedy Krieger Institute, Baltimore, MD, United States
| |
Collapse
|
33
|
Esshili A, Manitz MP, Freund N, Juckel G. Induction of inducible nitric oxide synthase expression in activated microglia and astrocytes following pre- and postnatal immune challenge in an animal model of schizophrenia. Eur Neuropsychopharmacol 2020; 35:100-110. [PMID: 32439226 DOI: 10.1016/j.euroneuro.2020.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 02/24/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022]
Abstract
In the central nervous system, activated microglia and astrocytes produce proinflammatory mediators such as inducible nitric oxide (iNOS) and cytokines. Uncontrolled release of these mediators induced by immune challenge can lead to increased vulnerability to complex brain disorders such as schizophrenia. In this study, BALB/c mice were injected intraperitoneally (i.p) with the viral mimetic polyriboinosinic-polyribocytidilic acid (poly(I:C)) or saline. At postnatal day 30 (PND0), the animals were sacrificed and the hippocampus, corpus callosum, striatum, cortex, fimbria and ventricle were immunostained for Iba-1, a microglial marker, glial fibrillary acidic protein (GFAP), an astrocyte marker, and iNOS, an activation marker for NO. Additionally, serum cytokine profiling (Interleukin-2 (IL-2), IL- 4, IL-6, interferon gamma (IFN-γ), tumour necrosis factor (TNF), IL-17A and IL-10) was determined using serum samples from poly(I:C)-treated and control mice. Our results demonstrated that poly(I:C) induced overactivation of differential proinflammatory responses in microglia and astrocytes, which could be strongly enhanced by a postnatal poly(I:C) administration before PND 30 in one part of the animals investigated. Specifically, there was significant iNOS upregulation in hippocampus, cortex and corpus callosum of poly(I:C)-affected off-springs. These inflammatory alterations were accompanied by increased circulating levels of the proinflammatory cytokines tumour necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). This study provides insight into the role of microglia and astrocytes in an animal model of schizophrenia and an understanding of the regulation of iNOS expression in glial cells and cytokine networks. This knowledge could help identify novel targets for anti-oxidative and anti-inflammatory therapeutic schizophrenia intervention.
Collapse
Affiliation(s)
- Awatef Esshili
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, LWL-University Hospital, Ruhr University Bochum, Bochum, Germany; Laboratoire de génétique, biodiversité et valorisation des bioressources, Institut supérieur de biotechnologie de Monastir, Université de Monastir, Tunisie
| | - Marie-Pierre Manitz
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, LWL-University Hospital, Ruhr University Bochum, Bochum, Germany
| | - Nadja Freund
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, LWL-University Hospital, Ruhr University Bochum, Bochum, Germany
| | - Georg Juckel
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, LWL-University Hospital, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
34
|
Oldenburg KS, O’Shea TM, Fry RC. Genetic and epigenetic factors and early life inflammation as predictors of neurodevelopmental outcomes. Semin Fetal Neonatal Med 2020; 25:101115. [PMID: 32444251 PMCID: PMC7363586 DOI: 10.1016/j.siny.2020.101115] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Among individuals born very preterm, perinatal inflammation, particularly if sustained or recurring, is highly likely to contribute to adverse neurodevelopmental outcomes, including cerebral white matter damage, cerebral palsy, cognitive impairment, attention-deficit/hyperactivity disorder, and autism spectrum disorder. Antecedents and correlates of perinatal inflammation include socioeconomic disadvantage, maternal obesity, maternal infections, fetal growth restriction, neonatal sepsis, necrotizing enterocolitis, and prolonged mechanical ventilation. Genetic factors can modify susceptibility to perinatal inflammation and to neurodevelopmental disorders. Preliminary evidence supports a role of epigenetic markers as potential mediators of the presumed effects of preterm birth and/or its consequences on neurodevelopment later in life. Further study is needed of factors such as sex, psychosocial stressors, and environmental exposures that could modify the relationship of early life inflammation to later neurodevelopmental impairments. Also needed are pharmacological and non-pharmacological interventions to attenuate inflammation towards the goal of improving the neurodevelopment of individuals born very preterm.
Collapse
Affiliation(s)
- Kirsi S. Oldenburg
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - T. Michael O’Shea
- Department of Pediatrics (Neonatology), University of North Carolina School of Medicine
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| |
Collapse
|
35
|
Alemany-Navarro M, Cruz R, Real E, Segalàs C, Bertolín S, Rabionet R, Carracedo Á, Menchón JM, Alonso P. Looking into the genetic bases of OCD dimensions: a pilot genome-wide association study. Transl Psychiatry 2020; 10:151. [PMID: 32424139 PMCID: PMC7235014 DOI: 10.1038/s41398-020-0804-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/23/2020] [Accepted: 04/08/2020] [Indexed: 12/19/2022] Open
Abstract
The multidimensional nature of obsessive-compulsive disorder (OCD) has been consistently reported. Clinical and biological characteristics have been associated with OCD dimensions in different ways. Studies suggest the existence of specific genetic bases for the different OCD dimensions. In this study, we analyze the genomic markers, genes, gene ontology and biological pathways associated with the presence of aggressive/checking, symmetry/order, contamination/cleaning, hoarding, and sexual/religious symptoms, as assessed via the Dimensional Yale-Brown Obsessive Compulsive Scale (DY-BOCS) in 399 probands. Logistic regression analyses were performed at the single-nucleotide polymorphism (SNP) level. Gene-based and enrichment analyses were carried out for common (SNPs) and rare variants. No SNP was associated with any dimension at a genome-wide level (p < 5 × 10-8). Gene-based analyses showed one gene to be associated with hoarding (SETD3, p = 1.89 × 10-08); a gene highly expressed in the brain and which plays a role in apoptotic processes and transcriptomic changes, and another gene associated with aggressive symptoms (CPE; p = 4.42 × 10-6), which is involved in neurotrophic functions and the synthesis of peptide hormones and neurotransmitters. Different pathways or biological processes were represented by genes associated with aggressive (zinc ion response and lipid metabolism), order (lipid metabolism), sexual/religious (G protein-mediated processes) and hoarding (metabolic processes and anion transport) symptoms after FDR correction; while no pathway was associated with contamination. Specific genomic bases were found for each dimension assessed, especially in the enrichment analyses. Further research with larger samples and different techniques, such as next-generation sequencing, are needed to better understand the differential genetics of OCD dimensions.
Collapse
Affiliation(s)
- María Alemany-Navarro
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain. .,OCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, Barcelona, Spain. .,Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain.
| | - Raquel Cruz
- grid.11794.3a0000000109410645Grupo de Medicina Xenómica, CIBERER, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Eva Real
- grid.418284.30000 0004 0427 2257Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.411129.e0000 0000 8836 0780OCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, Barcelona, Spain ,grid.413448.e0000 0000 9314 1427CIBERSAM (Centro de Investigación en Red de Salud Mental), Instituto de Salud Carlos III, Madrid, Spain
| | - Cinto Segalàs
- grid.418284.30000 0004 0427 2257Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.411129.e0000 0000 8836 0780OCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, Barcelona, Spain ,grid.413448.e0000 0000 9314 1427CIBERSAM (Centro de Investigación en Red de Salud Mental), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Bertolín
- grid.411129.e0000 0000 8836 0780OCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Raquel Rabionet
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain ,grid.5841.80000 0004 1937 0247Institut de Biomedicina de la Universitat de Barcelona (IBUB), CIBERER, and Dept. Genetics, Microbiology & Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Ángel Carracedo
- grid.11794.3a0000000109410645Grupo de Medicina Xenómica, CIBERER, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain ,Fundación Pública Galega de Medicina Xenómica, SERGAS, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Jose M. Menchón
- grid.418284.30000 0004 0427 2257Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.411129.e0000 0000 8836 0780OCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, Barcelona, Spain ,grid.5841.80000 0004 1937 0247Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain ,grid.413448.e0000 0000 9314 1427CIBERSAM (Centro de Investigación en Red de Salud Mental), Instituto de Salud Carlos III, Madrid, Spain
| | - Pino Alonso
- grid.411129.e0000 0000 8836 0780OCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, Barcelona, Spain ,grid.5841.80000 0004 1937 0247Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain ,grid.413448.e0000 0000 9314 1427CIBERSAM (Centro de Investigación en Red de Salud Mental), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
36
|
Patel CD, Backes IM, Taylor SA, Jiang Y, Marchant A, Pesola JM, Coen DM, Knipe DM, Ackerman ME, Leib DA. Maternal immunization confers protection against neonatal herpes simplex mortality and behavioral morbidity. Sci Transl Med 2020; 11:11/487/eaau6039. [PMID: 30971454 DOI: 10.1126/scitranslmed.aau6039] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/30/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022]
Abstract
Neonatal herpes simplex virus (nHSV) infections cause devastating morbidity and mortality in infants. Most nHSV cases are associated with primary maternal infection, consistent with the hypothesis that maternal immunity is protective. In humans, we found HSV-specific neutralizing antibodies in newborns of immune mothers, indicating that placentally transferred HSV-specific antibody is protective. Using a murine model, we showed that passive administration of HSV-specific antibody to dams prevented disseminated infection and mortality in pups. Maternal immunization with an HSV-2 replication-defective vaccine candidate, dl5-29, led to transfer of HSV-specific antibodies into neonatal circulation that protected against nHSV neurological disease and death. Furthermore, we observed considerable anxiety-like behavior in adult mice that had been infected with low doses of HSV as neonates, despite a notable lack of signs of infection. This phenotype suggests that nHSV infection can have an unsuspected and permanent impact on behavior. These behavioral sequelae of nHSV were prevented by maternal immunization with dl5-29, demonstrating an unexpected benefit of immunization. These findings also support the general concept that maternal immunization can prevent neurotropic neonatal infections and associated morbidity and mortality.
Collapse
Affiliation(s)
- Chaya D Patel
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA.,Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA
| | - Iara M Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA.,Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Sean A Taylor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Yike Jiang
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi B-6041, Belgium
| | - Jean M Pesola
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Donald M Coen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - David M Knipe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - David A Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA.
| |
Collapse
|
37
|
Jantzie LL, Maxwell JR, Newville JC, Yellowhair TR, Kitase Y, Madurai N, Ramachandra S, Bakhireva LN, Northington FJ, Gerner G, Tekes A, Milio LA, Brigman JL, Robinson S, Allan A. Prenatal opioid exposure: The next neonatal neuroinflammatory disease. Brain Behav Immun 2020; 84:45-58. [PMID: 31765790 PMCID: PMC7010550 DOI: 10.1016/j.bbi.2019.11.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/29/2019] [Accepted: 11/17/2019] [Indexed: 01/19/2023] Open
Abstract
The rates of opioid use disorder during pregnancy have more than quadrupled in the last decade, resulting in numerous infants suffering exposure to opioids during the perinatal period, a critical period of central nervous system (CNS) development. Despite increasing use, the characterization and definition of the molecular and cellular mechanisms of the long-term neurodevelopmental impacts of opioid exposure commencing in utero remains incomplete. Thus, in consideration of the looming public health crisis stemming from the multitude of infants with prenatal opioid exposure entering school age, we undertook an investigation of the effects of perinatal methadone exposure in a novel preclinical model. Specifically, we examined the effects of opioids on the developing brain to elucidate mechanisms of putative neural cell injury, to identify diagnostic biomarkers and to guide clinical studies of outcome and follow-up. We hypothesized that methadone would induce a pronounced inflammatory profile in both dams and their pups, and be associated with immune system dysfunction, sustained CNS injury, and altered cognition and executive function into adulthood. This investigation was conducted using a combination of cellular, molecular, biochemical, and clinically translatable biomarker, imaging and cognitive assessment platforms. Data reveal that perinatal methadone exposure increases inflammatory cytokines in the neonatal peripheral circulation, and reprograms and primes the immune system through sustained peripheral immune hyperreactivity. In the brain, perinatal methadone exposure not only increases chemokines and cytokines throughout a crucial developmental period, but also alters microglia morphology consistent with activation, and upregulates TLR4 and MyD88 mRNA. This increase in neuroinflammation coincides with reduced myelin basic protein and altered neurofilament expression, as well as reduced structural coherence and significantly decreased fractional anisotropy on diffusion tensor imaging. In addition to this microstructural brain injury, adult rats exposed to methadone in the perinatal period have significant impairment in associative learning and executive control as assessed using touchscreen technology. Collectively, these data reveal a distinct systemic and neuroinflammatory signature associated with prenatal methadone exposure, suggestive of an altered CNS microenvironment, dysregulated developmental homeostasis, complex concurrent neural injury, and imaging and cognitive findings consistent with clinical literature. Further investigation is required to define appropriate therapies targeted at the neural injury and improve the long-term outcomes for this exceedingly vulnerable patient population.
Collapse
Affiliation(s)
- Lauren L. Jantzie
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Neurology, Kennedy Krieger Institute, Baltimore, MD.,Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM.,Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM.,Correspondence: Lauren L. Jantzie, PhD, Johns Hopkins University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, 600 N. Wolfe Street, CMSC Building Room 6-104A, Baltimore, MD 21287,
| | - Jessie R. Maxwell
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM.,Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM
| | - Jessie C. Newville
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM.,Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM
| | - Tracylyn R. Yellowhair
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yuma Kitase
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nethra Madurai
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sindhu Ramachandra
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ludmila N. Bakhireva
- Substance Use Research and Education (SURE) Center, University of New Mexico College of Pharmacy, Albuquerque, NM
| | | | - Gwendolyn Gerner
- Department of Neuropsychology, Kennedy Krieger Institute, Baltimore, MD,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Aylin Tekes
- Division of Pediatric Radiology and Pediatric Neuroradiology, Russell Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lorraine A. Milio
- Department of Obstetrics & Gynecology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jonathan L. Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrea Allan
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM
| |
Collapse
|
38
|
Kerr N, Dietrich DW, Bramlett HM, Raval AP. Sexually dimorphic microglia and ischemic stroke. CNS Neurosci Ther 2019; 25:1308-1317. [PMID: 31747126 PMCID: PMC6887716 DOI: 10.1111/cns.13267] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/26/2022] Open
Abstract
Ischemic stroke kills more women compared with men thus emphasizing a significant sexual dimorphism in ischemic pathophysiological outcomes. However, the mechanisms behind this sexual dimorphism are yet to be fully understood. It is well established that cerebral ischemia activates a variety of inflammatory cascades and that microglia are the primary immune cells of the brain. After ischemic injury, microglia are activated and play a crucial role in progression and resolution of the neuroinflammatory response. In recent years, research has focused on the role that microglia play in this sexual dimorphism that exists in the response to central nervous system (CNS) injury. Evidence suggests that the molecular mechanisms leading to microglial activation and polarization of phenotypes may be influenced by sex, therefore causing a difference in the pro/anti‐inflammatory responses after CNS injury. Here, we review advances highlighting that sex differences in microglia are an important factor in the inflammatory responses that are seen after ischemic injury. We discuss the main differences between microglia in the healthy and diseased developing, adult, and aging brain. We also focus on the dimorphism that exists between males and females in microglial‐induced inflammation and energy metabolism after CNS injury. Finally, we describe how all of the current research and literature regarding sex differences in microglia contribute to the differences in poststroke responses between males and females.
Collapse
Affiliation(s)
- Nadine Kerr
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Dalton W Dietrich
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA.,Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
39
|
|
40
|
Biouss G, Antounians L, Li B, O'Connell JS, Seo S, Catania VD, Guadagno J, Rahman A, Zani-Ruttenstock E, Svergun N, Pierro A, Zani A. Experimental necrotizing enterocolitis induces neuroinflammation in the neonatal brain. J Neuroinflammation 2019; 16:97. [PMID: 31077225 PMCID: PMC6511222 DOI: 10.1186/s12974-019-1481-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/16/2019] [Indexed: 01/15/2023] Open
Abstract
Background Necrotizing enterocolitis (NEC) is an inflammatory gastrointestinal disease primarily affecting preterm neonates. Neonates with NEC suffer from a degree of neurodevelopmental delay that is not explained by prematurity alone. There is a need to understand the pathogenesis of neurodevelopmental delay in NEC. In this study, we assessed the macroscopic and microscopic changes that occur to brain cell populations in specific brain regions in a neonatal mouse model of NEC. Moreover, we investigated the role of intestinal inflammation as part of the mechanism responsible for the changes observed in the brain of pups with NEC. Methods Brains of mice were assessed for gross morphology and cerebral cortex thickness (using histology). Markers for mature neurons, oligodendrocytes, neural progenitor cells, microglia, and astrocytes were used to quantify their cell populations in different regions of the brain. Levels of cell apoptosis in the brain were measured by Western blotting and immunohistochemistry. Endoplasmic reticulum (ER) stress markers and levels of pro-inflammatory cytokines (in the ileum and brain) were measured by RT-qPCR and Western blotting. A Pearson test was used to correlate the levels of cytokines (ELISA) in the brain and ileum and to correlate activated microglia and astrocyte populations to the severity of NEC. Results NEC pups had smaller brain weights, higher brain-to-body weight ratios, and thinner cortices compared to control pups. NEC pups had increased levels of apoptosis and ER stress. In addition, NEC was associated with a reduction in the number of neurons, oligodendrocytes, and neural progenitors in specific regions of the brain. Levels of pro-inflammatory cytokines and the density of activated microglia and astrocytes were increased in the brain and positively correlated with the increase in the levels pro-inflammatory cytokines in the gut and the severity of NEC damage respectively. Conclusions NEC is associated with severe changes in brain morphology, a pro-inflammatory response in the brain that alters cell homeostasis and density of brain cell populations in specific cerebral regions. We show that the severity of neuroinflammation is associated with the severity of NEC. Our findings suggest that early intervention during NEC may reduce the chance of acute neuroinflammation and cerebral damage. Electronic supplementary material The online version of this article (10.1186/s12974-019-1481-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- George Biouss
- Developmental and Stem Cell Biology Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.,Translational Medicine Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.,Translational Medicine Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
| | - Joshua S O'Connell
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.,Translational Medicine Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
| | - Shogo Seo
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.,Translational Medicine Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
| | - Vincenzo D Catania
- Developmental and Stem Cell Biology Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Jennifer Guadagno
- Developmental and Stem Cell Biology Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Abidur Rahman
- Developmental and Stem Cell Biology Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
| | - Elke Zani-Ruttenstock
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.,Translational Medicine Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Nataliia Svergun
- Developmental and Stem Cell Biology Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.,Translational Medicine Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, PGCRL, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada. .,Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada. .,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Yellowhair TR, Noor S, Mares B, Jose C, Newville JC, Maxwell JR, Northington FJ, Milligan ED, Robinson S, Jantzie LL. Chorioamnionitis in Rats Precipitates Extended Postnatal Inflammatory Lymphocyte Hyperreactivity. Dev Neurosci 2019; 40:1-11. [PMID: 30921800 PMCID: PMC6765467 DOI: 10.1159/000497273] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/25/2019] [Indexed: 12/16/2022] Open
Abstract
Preterm birth is an important cause of perinatal brain injury (PBI). Neurological injury in extremely preterm infants often begins in utero with chorioamnionitis (CHORIO) or inflammation/infection of the placenta and concomitant placental insufficiency. Studies in humans have shown dysregulated inflammatory signaling throughout the placental-fetal brain axis and altered peripheral immune responses in children born preterm with cerebral palsy (CP). We hypothesized that peripheral immune responses would be altered in our well-established rat model of CP. Specifically, we proposed that isolated peripheral blood mononuclear cells (PBMCs) would be hyperresponsive to a second hit of inflammation throughout an extended postnatal time course. Pregnant Sprague-Dawley dams underwent a laparotomy on embryonic day 18 (E18) with occlusion of the uterine arteries (for 60 min) followed by intra-amniotic injection of lipopolysaccharide (LPS, 4 μg/sac) to induce injury in utero. Shams underwent laparotomy only, with equivalent duration of anesthesia. Laparotomies were then closed, and the rat pups were born at E22. PBMCs were isolated from pups on postnatal day 7 (P7) and P21, and subsequently stimulated in vitro with LPS for 3 or 24 h. A secreted inflammatory profile analysis of conditioned media was performed using multiplex electrochemiluminescent immunoassays, and the composition of inflammatory cells was assayed with flow cytometry (FC). Results indicate that CHORIO PBMCs challenged with LPS are hyperreactive and secrete significantly more tumor necrosis factor α (TNFα) and C-X-C chemokine ligand 1 at P7. FC confirmed increased intracellular TNFα in CHORIO pups at P7 following LPS stimulation, in addition to increased numbers of CD11b/c immunopositive myeloid cells. Notably, TNFα secretion was sustained until P21, with increased interleukin 6, concomitant with increased expression of integrin β1, suggesting both sustained peripheral immune hyperreactivity and a heightened activation state. Taken together, these data indicate that in utero injury primes the immune system and augments enhanced inflammatory signaling. The insidious effects of primed peripheral immune cells may compound PBI secondary to CHORIO and/or placental insufficiency, and thereby render the brain susceptible to future chronic neurological disease. Further understanding of inflammatory mechanisms in PBI may yield clinically important biomarkers and facilitate individualized repair strategies and treatments.
Collapse
Affiliation(s)
- Tracylyn R Yellowhair
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Shahani Noor
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Brittney Mares
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Clement Jose
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Jessie C Newville
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Jessie R Maxwell
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Frances J Northington
- Division of Newborn Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erin D Milligan
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lauren L Jantzie
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA,
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA,
- Division of Newborn Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,
| |
Collapse
|
42
|
Sylvia KE, Deyoe JE, Demas GE. Early-life sickness may predispose Siberian hamsters to behavioral changes following alterations of the gut microbiome in adulthood. Brain Behav Immun 2018; 73:571-583. [PMID: 29981426 PMCID: PMC6607895 DOI: 10.1016/j.bbi.2018.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 06/21/2018] [Accepted: 07/01/2018] [Indexed: 12/24/2022] Open
Abstract
Although it is well-established that the immune system plays an important role in the development of physiology and behavior, the gut microbiome has recently become of interest in the study of developmental origins of behavior. Studies suggest that the effects of early-life immune activation may not occur until a secondary stressor is introduced, though the precise nature and timing of the stressor may be critical in the response. Further, recent work suggests that the microbiome and the immune system develop in parallel, and therefore any perturbations to one of these systems early in life will likely affect the other. Here, we sought to determine whether early-life activation of the immune system had long-term consequences on how the gut microbiome responds to antibiotic treatment in adulthood and whether those changes influence adult same-sex social behavior. In order to test the hypothesis that an early-life immune challenge makes individuals more vulnerable to the effects of antibiotics, we mimicked an early-life infection by injecting pups at postnatal day 3 and 5 with lipopolysaccharide (LPS; cell wall component of gram-negative bacteria) or saline, and subsequently exposed the same animals to antibiotic treatment (known to influence microbial community composition and behavior) or water in adulthood. We tracked physiology across development, and paired males and females with a novel individual of the same age and sex in adulthood to score same-sex behavior (e.g., aggression, investigation, grooming) before antibiotic treatment, immediately following treatment, and after recovery from antibiotics. LPS-treated females exhibited impaired reproductive physiology and function in adulthood (e.g., smaller ovaries and abnormal estrous cycles), and female and male gut microbial communities were strongly affected by antibiotic treatment in adulthood, but only slightly affected by postnatal LPS alone. Interestingly, LPS-treated males exhibited more robust changes in their behavioral response following adult antibiotic treatment, including decreased investigation and increased grooming, suggestive of changes in anxiety-like behaviors. These data suggest that males may be more vulnerable than females to behavioral abnormalities after being predisposed to an immune challenge early in life. Collectively, these results provide novel evidence that some of the sex-specific behavioral consequences of an early-life immune challenge may not transpire until an individual is faced with a secondary challenge, and the context in which an individual is exposed can greatly influence the response.
Collapse
Affiliation(s)
- Kristyn E. Sylvia
- Corresponding author at: Department of Biology, Indiana University, 1001 E. 3 Street, Bloomington, IN 47405, USA. (K.E. Sylvia)
| | | | | |
Collapse
|
43
|
Wang X, Yang J, Xing Z, Zhang H, Wen Y, Qi F, Zuo Z, Xu J, Yao Z. IL-4 mediates the delayed neurobehavioral impairments induced by neonatal hepatitis B vaccination that involves the down-regulation of the IL-4 receptor in the hippocampus. Cytokine 2018; 110:137-149. [PMID: 29751176 DOI: 10.1016/j.cyto.2018.04.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 01/23/2023]
Abstract
We have previously verified that neonatal hepatitis B vaccination induced hippocampal neuroinflammation and behavior impairments in mice. However, the exact mechanism of these effects remain unclear. In this study, we observed that neonatal hepatitis B vaccination induced an anti-inflammatory cytokine response lasting for 4-5 weeks in both the serum and the hippocampus, primarily indicated by elevated IL-4 levels. Three weeks after the vaccination schedule, however, hepatitis B vaccine (HBV)-mice showed delayed hippocampal neuroinflammation. In periphery, IL-4 is the major cytokine induced by this vaccine. Correlation analyses showed a positive relationship in the IL-4 levels between serum and hippocampus in HBV-mice. Thus, we investigated whether neonatal over-exposure to systemic IL-4 influences brain and behavior. We observed that mice injected intraperitoneally with recombinant mouse IL-4 (mIL-4) during early life had similar neuroinflammation and cognition impairment similar to those induced by neonatal hepatitis B vaccination. Next, the mechanism underlying the effects of IL-4 on brain in mice was explored using a series of experiments. In brief, these experiments showed that IL-4 mediates the delayed neurobehavioral impairments induced by neonatal hepatitis B vaccination, which involves the permeability of neonatal blood-brain barrier and the down-regulation of IL-4 receptor. This finding suggests that clinical events concerning neonatal IL-4 over-exposure, including neonatal hepatitis B vaccination and allergic asthma in human infants, may have adverse implications for brain development and cognition.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Junhua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Zhiwei Xing
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Hongyang Zhang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Yaru Wen
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China.
| |
Collapse
|
44
|
Vojtechova I, Petrasek T, Maleninska K, Brozka H, Tejkalova H, Horacek J, Stuchlik A, Vales K. Neonatal immune activation by lipopolysaccharide causes inadequate emotional responses to novel situations but no changes in anxiety or cognitive behavior in Wistar rats. Behav Brain Res 2018; 349:42-53. [PMID: 29729302 DOI: 10.1016/j.bbr.2018.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 11/28/2022]
Abstract
Infection during the prenatal or neonatal stages of life is considered one of the major risk factors for the development of mental diseases such as schizophrenia or autism. However, the impacts of such an immune challenge on adult behavior are still not clear. In our study, we used a model of early postnatal immune activation by the application of bacterial endotoxin lipopolysaccharide (LPS) to rat pups at a dose of 2 mg/kg from postnatal day (PD) 5 to PD 9. In adulthood, the rats were tested in a battery of tasks probing various aspects of behavior: spontaneous activity (open field test), social behavior (social interactions and female bedding exploration), anxiety (elevated plus maze), cognition (active place avoidance in Carousel) and emotional response (ultrasonic vocalization recording). Moreover, we tested sensitivity to acute challenge with MK-801, a psychotomimetic drug. Our results show that the application of LPS led to increased self-grooming in the female bedding exploration test and inadequate emotional reactions in Carousel maze displayed by ultrasonic vocalizations. However, it did not have serious consequences on exploration, locomotion, social behavior or cognition. Furthermore, exposition to MK-801 did not trigger social or cognitive deficits in the LPS-treated rats. We conclude that the emotional domain is the most sensitive to the changes induced by neonatal immune activation in rats, including a disrupted response to novel and stressful situations in early adulthood (similar to that observed in human patients suffering from schizophrenia or autism), while other aspects of tested behavior remain unaffected.
Collapse
Affiliation(s)
- Iveta Vojtechova
- First Faculty of Medicine, Charles University, Katerinska 32, 12108, Prague 2, Czech Republic; Department of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic; National Institute of Mental Health, Topolova 748, 25067, Klecany, Czech Republic.
| | - Tomas Petrasek
- Department of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic; National Institute of Mental Health, Topolova 748, 25067, Klecany, Czech Republic.
| | - Kristyna Maleninska
- Department of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic.
| | - Hana Brozka
- Department of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic.
| | - Hana Tejkalova
- National Institute of Mental Health, Topolova 748, 25067, Klecany, Czech Republic.
| | - Jiri Horacek
- National Institute of Mental Health, Topolova 748, 25067, Klecany, Czech Republic.
| | - Ales Stuchlik
- Department of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic.
| | - Karel Vales
- National Institute of Mental Health, Topolova 748, 25067, Klecany, Czech Republic.
| |
Collapse
|
45
|
The Role of the Microglial Cx3cr1 Pathway in the Postnatal Maturation of Retinal Photoreceptors. J Neurosci 2018; 38:4708-4723. [PMID: 29669747 DOI: 10.1523/jneurosci.2368-17.2018] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 03/22/2018] [Accepted: 04/10/2018] [Indexed: 11/21/2022] Open
Abstract
Microglia are the resident immune cells of the CNS, and their response to infection, injury and disease is well documented. More recently, microglia have been shown to play a role in normal CNS development, with the fractalkine-Cx3cr1 signaling pathway of particular importance. This work describes the interaction between the light-sensitive photoreceptors and microglia during eye opening, a time of postnatal photoreceptor maturation. Genetic removal of Cx3cr1 (Cx3cr1GFP/GFP ) led to an early retinal dysfunction soon after eye opening [postnatal day 17 (P17)] and cone photoreceptor loss (P30 onward) in mice of either sex. This dysfunction occurred at a time when fractalkine expression was predominantly outer retinal, when there was an increased microglial presence near the photoreceptor layer and increased microglial-cone photoreceptor contacts. Photoreceptor maturation and outer segment elongation was coincident with increased opsin photopigment expression in wild-type retina, while this was aberrant in the Cx3cr1GFP/GFP retina and outer segment length was reduced. A beadchip array highlighted Cx3cr1 regulation of genes involved in the photoreceptor cilium, a key structure that is important for outer segment elongation. This was confirmed with quantitative PCR with specific cilium-related genes, Rpgr and Rpgrip1, downregulated at eye opening (P14). While the overall cilium structure was unaffected, expression of Rpgr, Rpgrip1, and centrin were restricted to more proximal regions of the transitional zone. This study highlighted a novel role for microglia in postnatal neuronal development within the retina, with loss of fractalkine-Cx3cr1 signaling leading to an altered distribution of cilium proteins, failure of outer segment elongation and ultimately cone photoreceptor loss.SIGNIFICANCE STATEMENT Microglia are involved in CNS development and disease. This work highlights the role of microglia in postnatal development of the light-detecting photoreceptor neurons within the mouse retina. Loss of the microglial Cx3cr1 signaling pathway resulted in specific alterations in the cilium, a key structure in photoreceptor outer segment elongation. The distribution of key components of the cilium transitional zone, Rpgr, Rpgrip1, and centrin, were altered in retinae lacking Cx3cr1 with reduced outer segment length and cone photoreceptor death observed at later postnatal ages. This work identifies a novel role for microglia in the postnatal maturation of retinal photoreceptors.
Collapse
|
46
|
Petitdant N, Lecomte A, Robidel F, Gamez C, Blazy K, Villégier AS. Alteration of adaptive behaviors of progeny after maternal mobile phone exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:10894-10903. [PMID: 29397508 DOI: 10.1007/s11356-017-1178-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 12/27/2017] [Indexed: 06/07/2023]
Abstract
Exposure of pregnant women to radiofrequency (RF) devices raises questions on their possible health consequences for their progeny. We examined the hazard threshold of gestational RF on the progeny's glial homeostasis, sensory-motor gating, emotionality, and novelty seeking and tested whether maternal immune activation would increase RF toxicity. Pregnant dams were daily restrained with loop antennas adjoining the abdomen (fetus body specific absorption rates (SAR): 0, 0.7, or 2.6 W/kg) and received three lipopolysaccharide (LPS) intra-peritoneal injections (0 or 80 μg/kg). Scores in the prepulse startle inhibition, fear conditioning, open field, and elevated plus maze were assessed at adolescence and adulthood. Glial fibrillary acidic protein (GFAP) and interleukines-1β (ILs) were quantified. LPS induced a SAR-dependent reduction of the prepulse startle inhibition in adults. Activity in the open field was reduced at 2.6 W/kg at adolescence. GFAP and ILs, emotional memory, and anxiety-related behaviors were not modified. These data support the hypothesis that maternal immune activation increased the developmental RF exposure-induced long-term neurobiological impairments. These data support the fact that fetuses who receive combined environmental exposures with RF need special attention for protection.
Collapse
Affiliation(s)
- Nicolas Petitdant
- Toxicology Unit, National Institute for Environmental Protection and Industrial Risks (INERIS), Verneuil-en-Halatte, France
- PériTox-INERIS Laboratory, UMR-I 01 Jules Verne University of Picardy, 80054, Amiens, France
| | - Anthony Lecomte
- Toxicology Unit, National Institute for Environmental Protection and Industrial Risks (INERIS), Verneuil-en-Halatte, France
- PériTox-INERIS Laboratory, UMR-I 01 Jules Verne University of Picardy, 80054, Amiens, France
| | - Franck Robidel
- Toxicology Unit, National Institute for Environmental Protection and Industrial Risks (INERIS), Verneuil-en-Halatte, France
- PériTox-INERIS Laboratory, UMR-I 01 Jules Verne University of Picardy, 80054, Amiens, France
| | - Christelle Gamez
- Toxicology Unit, National Institute for Environmental Protection and Industrial Risks (INERIS), Verneuil-en-Halatte, France
- PériTox-INERIS Laboratory, UMR-I 01 Jules Verne University of Picardy, 80054, Amiens, France
| | - Kelly Blazy
- Toxicology Unit, National Institute for Environmental Protection and Industrial Risks (INERIS), Verneuil-en-Halatte, France
- PériTox-INERIS Laboratory, UMR-I 01 Jules Verne University of Picardy, 80054, Amiens, France
| | - Anne-Sophie Villégier
- Toxicology Unit, National Institute for Environmental Protection and Industrial Risks (INERIS), Verneuil-en-Halatte, France.
- PériTox-INERIS Laboratory, UMR-I 01 Jules Verne University of Picardy, 80054, Amiens, France.
- Unité de Toxicologie Expérimentale, Parc Technologique ALATA, Institut National de l'Environnement Industriel et des Risques, BP no. 2, 60550, Verneuil-en-Halatte, France.
| |
Collapse
|
47
|
Sylvia KE, Demas GE. A gut feeling: Microbiome-brain-immune interactions modulate social and affective behaviors. Horm Behav 2018; 99:41-49. [PMID: 29427583 PMCID: PMC5880698 DOI: 10.1016/j.yhbeh.2018.02.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 02/02/2018] [Accepted: 02/04/2018] [Indexed: 02/07/2023]
Abstract
The expression of a wide range of social and affective behaviors, including aggression and investigation, as well as anxiety- and depressive-like behaviors, involves interactions among many different physiological systems, including the neuroendocrine and immune systems. Recent work suggests that the gut microbiome may also play a critical role in modulating behavior and likely functions as an important integrator across physiological systems. Microbes within the gut may communicate with the brain via both neural and humoral pathways, providing numerous avenues of research in the area of the gut-brain axis. We are now just beginning to understand the intricate relationships among the brain, microbiome, and immune system and how they work in concert to influence behavior. The effects of different forms of experience (e.g., changes in diet, immune challenge, and psychological stress) on the brain, gut microbiome, and the immune system have often been studied independently. Though because these systems do not work in isolation, it is essential to shift our focus to the connections among them as we move forward in our investigations of the gut-brain axis, the shaping of behavioral phenotypes, and the possible clinical implications of these interactions. This review summarizes the recent progress the field has made in understanding the important role the gut microbiome plays in the modulation of social and affective behaviors, as well as some of the intricate mechanisms by which the microbiome may be communicating with the brain and immune system.
Collapse
Affiliation(s)
- Kristyn E Sylvia
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA.
| | - Gregory E Demas
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA; Program in Neuroscience, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
48
|
Malan-Muller S, Valles-Colomer M, Raes J, Lowry CA, Seedat S, Hemmings SM. The Gut Microbiome and Mental Health: Implications for Anxiety- and Trauma-Related Disorders. ACTA ACUST UNITED AC 2018; 22:90-107. [DOI: 10.1089/omi.2017.0077] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Stefanie Malan-Muller
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Mireia Valles-Colomer
- Department of Microbiology and Immunology, Rega Institute, KU Leuven–University of Leuven, Leuven, Belgium
- VIB, Center for Microbiology, Leuven, Belgium
| | - Jeroen Raes
- Department of Microbiology and Immunology, Rega Institute, KU Leuven–University of Leuven, Leuven, Belgium
- VIB, Center for Microbiology, Leuven, Belgium
| | - Christopher A. Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-Core), Aurora, Colorado
- Department of Psychiatry, Neurology & Physical Medicine and Rehabilitation, Anschutz School of Medicine, University of Colorado, Aurora, Colorado
- VA Rocky Mountain Mental Illness Research, Education, and Clinical Center (MIRECC), Denver, Colorado
- Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Sian M.J. Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
49
|
Sylvia KE, Báez Ramos P, Demas GE. Sickness-induced changes in physiology do not affect fecundity or same-sex behavior. Physiol Behav 2018; 184:68-77. [PMID: 29127071 PMCID: PMC5753605 DOI: 10.1016/j.physbeh.2017.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/04/2017] [Accepted: 11/04/2017] [Indexed: 12/14/2022]
Abstract
Previous work in our lab has shown that early-life infection affects female reproductive physiology and function (i.e., smaller ovaries, abnormal estrous cycles) and alters investigation and aggression towards male conspecifics in a reproductive context. Although many studies have investigated the effects of postnatal immune challenge on physiological and behavioral development, fewer studies have examined whether these changes have ultimate effects on reproduction. In the current study, we paired Siberian hamsters (Phodopus sungorus) and simulated a bacterial infection in early life by administering lipopolysaccharide (LPS) to male and female pups on pnd3 and pnd5. In adulthood, hamsters were paired with novel individuals of the same sex, and we scored an array of social behaviors (e.g., investigation, aggression). We then paired animals with individuals of the opposite sex for 5 consecutive nights, providing them with the opportunity to mate. We found that females exhibited impaired reproductive physiology and function in adulthood (i.e., smaller ovaries and abnormal estrous cycles), similar to our previous work. However, both LPS-treated males and females exhibited similar same-sex social behavior when compared with saline-treated controls, they successfully mated, and there were no significant changes in fecundity. These data suggest that the physiological changes in response to neonatal immune challenge may not have long-term effects on reproductive success in a controlled environment. Collectively, the results of this study are particularly important when investigating the relationships between physiology and behavior within an ultimate context. Animals exposed to early-life stress may in fact be capable of compensating for changes in physiology in order to survive and reproduce in some contexts.
Collapse
Affiliation(s)
- Kristyn E Sylvia
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA.
| | - Patricia Báez Ramos
- Biology Department, University of Puerto Rico at Mayagüez, Mayagüez, PR 00681, USA
| | - Gregory E Demas
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
50
|
Weinhard L, d'Errico P, Leng Tay T. Headmasters: Microglial regulation of learning and memory in health and disease. AIMS MOLECULAR SCIENCE 2018. [DOI: 10.3934/molsci.2018.1.63] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|