1
|
Otero AM, Connolly MG, Gonzalez-Ricon RJ, Wang SS, Allen JM, Antonson AM. Influenza A virus during pregnancy disrupts maternal intestinal immunity and fetal cortical development in a dose- and time-dependent manner. Mol Psychiatry 2025; 30:13-28. [PMID: 38961232 PMCID: PMC11649561 DOI: 10.1038/s41380-024-02648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
Epidemiological studies link exposure to viral infection during pregnancy, including influenza A virus (IAV) infection, with increased incidence of neurodevelopmental disorders (NDDs) in offspring. Models of maternal immune activation (MIA) using viral mimetics demonstrate that activation of maternal intestinal T helper 17 (TH17) cells, which produce effector cytokine interleukin (IL)-17, leads to aberrant fetal brain development, such as neocortical malformations. Fetal microglia and border-associated macrophages (BAMs) also serve as potential cellular mediators of MIA-induced cortical abnormalities. However, neither the inflammation-induced TH17 cell pathway nor fetal brain-resident macrophages have been thoroughly examined in models of live viral infection during pregnancy. Here, we inoculated pregnant mice with two infectious doses of IAV and evaluated peak innate and adaptive immune responses in the dam and fetus. While respiratory IAV infection led to dose-dependent maternal colonic shortening and microbial dysregulation, there was no elevation in intestinal TH17 cells nor IL-17. Systemically, IAV resulted in consistent dose- and time-dependent increases in IL-6 and IFN-γ. Fetal cortical abnormalities and global changes in fetal brain transcripts were observable in the high-but not the moderate-dose IAV group. Profiling of fetal microglia and BAMs revealed dose- and time-dependent differences in the numbers of meningeal but not choroid plexus BAMs, while microglial numbers and proliferative capacity of Iba1+ cells remained constant. Fetal brain-resident macrophages increased phagocytic CD68 expression, also in a dose- and time-dependent fashion. Taken together, our findings indicate that certain features of MIA are conserved between mimetic and live virus models, while others are not. Overall, we provide consistent evidence of an infection severity threshold for downstream maternal inflammation and fetal cortical abnormalities, which recapitulates a key feature of the epidemiological data and further underscores the importance of using live pathogens in NDD modeling to better evaluate the complete immune response and to improve translation to the clinic.
Collapse
Affiliation(s)
- Ashley M Otero
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Meghan G Connolly
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | - Selena S Wang
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jacob M Allen
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Adrienne M Antonson
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
2
|
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] [Grants] [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
|
3
|
Zhou X, Guo Z, Ling Y, Teng W, Cui J, Yan Z, Hou X, Cen W, Long N, Li W, Yang H, Chu L. Causal effect of air pollution on the risk of brain health and potential mediation by gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117080. [PMID: 39332203 DOI: 10.1016/j.ecoenv.2024.117080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVE Epidemiologic investigations have examined the correlation between air pollution and neurologic disorders and neuroanatomic structures. Increasing evidence underscores the profound influence of the gut microbiota on brain health. However, the existing evidence is equivocal, and a causal link remains uncertain. This study aimed: to determine if there is a causal connection between four key air pollutants, and 42 neurologic diseases, and 1325 distinct brain structures; and to explore the potential role of the gut microbiota in mediating these associations. METHODS Univariable Mendelian randomization (UVMR) and multivariable Mendelian randomization (MVMR) models were deployed to estimate the causal impact of air pollutants (including particulate matter [PM] with aerodynamic diameters <2.5 μm [PM2.5], and <10 μm [PM10]; PM2.5 absorbance; and nitrogen oxides [NOx]) on brain health through various Mendelian randomization methodologies. Lastly, the mediating role of the gut microbiome in the connections between the identified pollutants and neurologic diseases and brain structures was systematically examined. RESULTS The potential causal associations of PM2.5, PM2.5 absorbance, PM10, and exposure to NOx, with the risks of intracerebral hemorrhage, hippocampal perivascular spaces, large artery strokes, generalized epilepsy with tonic-clonic seizures, Alzheimer's disease, multiple sclerosis, anorexia nervosa, post-traumatic stress disorder (PTSD), and 420 brain structures, were investigated by UVMR analysis. Following adjustment for air pollutants by MVMR analysis, the genetic correlations between PM10 exposure and PTSD and multiple sclerosis remained significant and robust. Importantly, we observed that phylum Lentisphaerae may mediate the association between PM10 and multiple sclerosis. Additionally, PM2.5 absorbance with a greater risk of reduced thickness in the left anterior transverse temporal gyrus of Heschl and a decreased area in the right sulcus intermedius primus of Jensen, mediated by genus Senegalimassilia and genus Lachnospiraceae UCG010, respectively. Finally, we provided evidence that Clostridium innocuum and genus Ruminococcus2 may partly mediate the causal effect of NOx on altered thickness in the left transverse temporal cortex and area in the right sulcus intermedius primus of Jensen, respectively. CONCLUSION This study established a genetic connection between air pollution and brain health, implicating the gut microbiota as a potential mediator in the relationship between air pollution, neurologic disorders, and altered brain structures.
Collapse
Affiliation(s)
- Xingwang Zhou
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Zhengshan Guo
- The Institute of Public Administration, Southwest University of Finance and Economics, Chengdu, Sichuan, PR China
| | - Yuanguo Ling
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Wei Teng
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Junshuan Cui
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Zhangwei Yan
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Xianwen Hou
- Department of Neurosurgery, Qianxi People's Hospital, Qianxi, Guizhou, PR China
| | - Wu Cen
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Niya Long
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Wenyan Li
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Hua Yang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Liangzhao Chu
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, PR China.
| |
Collapse
|
4
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [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: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
5
|
Love C, Sominsky L, O'Hely M, Berk M, Vuillermin P, Dawson SL. Prenatal environmental risk factors for autism spectrum disorder and their potential mechanisms. BMC Med 2024; 22:393. [PMID: 39278907 PMCID: PMC11404034 DOI: 10.1186/s12916-024-03617-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 09/05/2024] [Indexed: 09/18/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that is globally increasing in prevalence. The rise of ASD can be partially attributed to diagnostic expansion and advocacy efforts; however, the interplay between genetic predisposition and modern environmental exposures is likely driving a true increase in incidence. A range of evidence indicates that prenatal exposures are critical. Infection during pregnancy, gestational diabetes, and maternal obesity are established risk factors for ASD. Emerging areas of research include the effects of maternal use of selective serotonin reuptake inhibitors, antibiotics, and exposure to toxicants during pregnancy on brain development and subsequent ASD. The underlying pathways of these risk factors remain uncertain, with varying levels of evidence implicating immune dysregulation, mitochondrial dysfunction, oxidative stress, gut microbiome alterations, and hormonal disruptions. This narrative review assesses the evidence of contributing prenatal environmental factors for ASD and associated mechanisms as potential targets for novel prevention strategies.
Collapse
Affiliation(s)
- Chloe Love
- Child Health Research Unit, Barwon Health, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Luba Sominsky
- Child Health Research Unit, Barwon Health, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Martin O'Hely
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
- Murdoch Children's Research Institute, Parkville, Australia
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Peter Vuillermin
- Child Health Research Unit, Barwon Health, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
- Murdoch Children's Research Institute, Parkville, Australia
| | - Samantha L Dawson
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia.
- Murdoch Children's Research Institute, Parkville, Australia.
- Food and Mood Centre, Deakin University, Geelong, Australia.
| |
Collapse
|
6
|
Walker KA, Rhodes ST, Liberman DA, Gore AC, Bell MR. Microglial responses to inflammatory challenge in adult rats altered by developmental exposure to polychlorinated biphenyls in a sex-specific manner. Neurotoxicology 2024; 104:95-115. [PMID: 39038526 PMCID: PMC11548868 DOI: 10.1016/j.neuro.2024.07.009] [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: 02/01/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Polychlorinated biphenyls are ubiquitous environmental contaminants linkedc with peripheral immune and neural dysfunction. Neuroimmune signaling is critical to brain development and later health; however, effects of PCBs on neuroimmune processes are largely undescribed. This study extends our previous work in neonatal or adolescent rats by investigating longer-term effects of perinatal PCB exposure on later neuroimmune responses to an inflammatory challenge in adulthood. Male and female Sprague-Dawley rats were exposed to a low-dose, environmentally relevant, mixture of PCBs (Aroclors 1242, 1248, and 1254, 1:1:1, 20 μg / kg dam BW per gestational day) or oil control during gestation and via lactation. Upon reaching adulthood, rats were given a mild inflammatory challenge with lipopolysaccharide (LPS, 50 μg / kg BW, ip) or saline control and then euthanized 3 hours later for gene expression analysis or 24 hours later for immunohistochemical labeling of Iba1+ microglia. PCB exposure did not alter gene expression or microglial morphology independently, but instead interacted with the LPS challenge in brain region- and sex-specific ways. In the female hypothalamus, PCB exposure blunted LPS responses of neuroimmune and neuromodulatory genes without changing microglial morphology. In the female prefrontal cortex, PCBs shifted Iba1+ cells from reactive to hyperramified morphology in response to LPS. Conversely, in the male hypothalamus, PCBs shifted cell phenotypes from hyperramified to reactive morphologies in response to LPS. The results highlight the potential for long-lasting effects of environmental contaminants that are differentially revealed over a lifetime, sometimes only after a secondary challenge. These neuroimmune endpoints are possible mechanisms for PCB effects on a range of neural dysfunction in adulthood, including mental health and neurodegenerative disorders. The findings suggest possible interactions with other environmental challenges that also influence neuroimmune systems.
Collapse
Affiliation(s)
- Katherine A Walker
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Simone T Rhodes
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Deborah A Liberman
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy and Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA.
| | - Margaret R Bell
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA; Division of Pharmacology and Toxicology, College of Pharmacy and Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
7
|
Santana-Coelho D, Pranske ZJ, Nolan SO, Hodges SL, Binder MS, Womble PD, Narvaiz DA, Muhammad I, Lugo JN. Neonatal immune stimulation results in sex-specific changes in ultrasonic vocalizations but does not affect seizure susceptibility in neonatal mice. Int J Dev Neurosci 2024; 84:381-391. [PMID: 38712612 DOI: 10.1002/jdn.10333] [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: 10/12/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/08/2024] Open
Abstract
Neuroinflammation during the neonatal period has been linked to disorders such as autism and epilepsy. In this study, we investigated the early life behavioral consequences of a single injection of lipopolysaccharide (LPS) at postnatal day 10 (PD10) in mice. To assess deficits in communication, we performed the isolation-induced ultrasonic vocalizations (USVs) test at PD12. To determine if early life immune stimulus could alter seizure susceptibility, latency to flurothyl-induced generalized seizures was measured at 4 hours (hrs), 2 days, or 5 days after LPS injections. LPS had a sex-dependent effect on USV number. LPS-treated male mice presented significantly fewer USVs than LPS-treated female mice. However, the number of calls did not significantly differ between control and LPS for either sex. In male mice, we found that downward, short, and composite calls were significantly more prevalent in the LPS treatment group, while upward, chevron, and complex calls were less prevalent than in controls (p < 0.05). Female mice that received LPS presented a significantly higher proportion of short, frequency steps, two-syllable, and composite calls in their repertoire when compared with female control mice (p < 0.05). Seizure latency was not altered by early-life inflammation at any of the time points measured. Our findings suggest that early-life immune stimulation at PD10 disrupts vocal development but does not alter the susceptibility to flurothyl-induced seizures during the neonatal period. Additionally, the effect of inflammation in the disruption of vocalization is sex-dependent.
Collapse
Affiliation(s)
| | - Zachary J Pranske
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Suzanne O Nolan
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | | | - Matthew S Binder
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Paige D Womble
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - David A Narvaiz
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Ilyasah Muhammad
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Joaquin N Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
- Institute of Biomedical Studets, Waco, Texas, USA
- Department of Biology, Baylor University, Waco, Texas, USA
| |
Collapse
|
8
|
Finch CE, Thorwald MA. Inhaled Pollutants of the Gero-Exposome and Later-Life Health. J Gerontol A Biol Sci Med Sci 2024; 79:glae107. [PMID: 38644649 PMCID: PMC11170295 DOI: 10.1093/gerona/glae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Indexed: 04/23/2024] Open
Abstract
Inhaled air pollutants (AirP) comprise extraordinarily diverse particles, volatiles, and gases from traffic, wildfire, cigarette smoke, dust, and various other sources. These pollutants contain numerous toxic components, which collectively differ in relative levels of components, but broadly share chemical classes. Exposure and health outcomes from AirP are complex, depending on pollutant source, duration of exposure, and socioeconomic status. We discuss examples in the current literature on organ responses to AirP, with a focus on lung, arteries, and brain. Some transcriptional responses are shared. It is well accepted that AirP contributes to Alzheimer's disease and other neurodegenerative conditions in the Gero-Exposome. However, we do not know which chemical compounds initiate these changes and how activation of these transcriptional pathways is further modified by genetics and prenatal development.
Collapse
Affiliation(s)
- Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Max A Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
9
|
Cacialli P, Ricci S, Servetto GP, Franceschini V, Ruiz-Zepeda F, Vigliaturo R. Altered Morpho-Functional Features of Neurogenesis in Zebrafish Embryos Exposed to Non-Combustion-Derived Magnetite. Int J Mol Sci 2024; 25:6459. [PMID: 38928164 PMCID: PMC11203806 DOI: 10.3390/ijms25126459] [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: 05/14/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Neurogenesis is the process by which new brain cells are formed. This crucial event emerges during embryonic life and proceeds in adulthood, and it could be influenced by environmental pollution. Non-combustion-derived magnetite represents a portion of the coarse particulate matter (PM) contributing to air and water pollution in urban settings. Studies on humans have reported that magnetite and other iron oxides have significant damaging effects at a central level, where these particles accumulate and promote oxidative stress. Similarly, magnetite nanoparticles can cross the placenta and damage the embryo brain during development, but the impact on neurogenesis is still unknown. Furthermore, an abnormal Fe cation concentration in cells and tissues might promote reactive oxygen species (ROS) generation and has been associated with multiple neurodegenerative conditions. In the present study, we used zebrafish as an in vivo system to analyze the specific effects of magnetite on embryonic neurogenesis. First, we characterized magnetite using mineralogical and spectroscopic analyses. Embryos treated with magnetite at sub-lethal concentrations showed a dose-response increase in ROS in the brain, which was accompanied by a massive decrease in antioxidant genes (sod2, cat, gsr, and nrf2). In addition, a higher number of apoptotic cells was observed in embryos treated with magnetite. Next, interestingly, embryos exposed to magnetite displayed a decrease in neural staminal progenitors (nestin, sox2, and pcna markers) and a neuronal marker (elavl3). Finally, we observed significative increases in apoeb (specific microglia marker) and interleukin-1b (il1b), confirming a status of inflammation in the brain embryos treated with magnetite. Our study represents the very first in vivo evidence concerning the effects of magnetite on brain development.
Collapse
Affiliation(s)
- Pietro Cacialli
- Department of Biological, Geological and Environmental Sciences (BIGEA), University of Bologna, 40126 Bologna, Italy
| | - Serena Ricci
- Department of Biological, Geological and Environmental Sciences (BIGEA), University of Bologna, 40126 Bologna, Italy
| | | | - Valeria Franceschini
- Department of Biological, Geological and Environmental Sciences (BIGEA), University of Bologna, 40126 Bologna, Italy
| | - Francisco Ruiz-Zepeda
- Department of Physics and Chemistry of Materials, Institute of Metals and Technology, Lepi pot 11, 1000 Ljubljana, Slovenia
- Department of Materials Chemistry, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Ruggero Vigliaturo
- Department of Earth Sciences, University of Turin, 10124 Turin, Italy
- Interdepartmental Centre for Studies on Asbestos and Other Toxic Particulates “G. Scansetti”, University of Turin, 10124 Turin, Italy
| |
Collapse
|
10
|
Tseng CEJ, Canales C, Marcus RE, Parmar AJ, Hightower BG, Mullett JE, Makary MM, Tassone AU, Saro HK, Townsend PH, Birtwell K, Nowinski L, Thom RP, Palumbo ML, Keary C, Catana C, McDougle CJ, Hooker JM, Zürcher NR. In vivo translocator protein in females with autism spectrum disorder: a pilot study. Neuropsychopharmacology 2024; 49:1193-1201. [PMID: 38615126 PMCID: PMC11109261 DOI: 10.1038/s41386-024-01859-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
Sex-based differences in the prevalence of autism spectrum disorder (ASD) are well-documented, with a male-to-female ratio of approximately 4:1. The clinical presentation of the core symptoms of ASD can also vary between sexes. Previously, positron emission tomography (PET) studies have identified alterations in the in vivo levels of translocator protein (TSPO)-a mitochondrial protein-in primarily or only male adults with ASD, with our group reporting lower TSPO relative to whole brain mean in males with ASD. However, whether in vivo TSPO levels are altered in females with ASD, specifically, is unknown. This is the first pilot study to measure in vivo TSPO in the brain in adult females with ASD using [11C]PBR28 PET-magnetic resonance imaging (MRI). Twelve adult females with ASD and 10 age- and TSPO genotype-matched controls (CON) completed one or two [11C]PBR28 PET-MRI scans. Females with ASD exhibited elevated [11C]PBR28 standardized uptake value ratio (SUVR) in the midcingulate cortex and splenium of the corpus callosum compared to CON. No brain area showed lower [11C]PBR28 SUVR in females with ASD compared to CON. Test-retest over several months showed stable [11C]PBR28 SUVR across time in both groups. Elevated regional [11C]PBR28 SUVR in females with ASD stand in stark contrast to our previous findings of lower regional [11C]PBR28 SUVR in males with ASD. Preliminary evidence of regionally elevated mitochondrial protein TSPO relative to whole brain mean in ASD females may reflect neuroimmuno-metabolic alterations specific to females with ASD.
Collapse
Affiliation(s)
- Chieh-En Jane Tseng
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Camila Canales
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Rachel E Marcus
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Anjali J Parmar
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Baileigh G Hightower
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Jennifer E Mullett
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
| | - Meena M Makary
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Cairo, Egypt
| | - Alison U Tassone
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Hannah K Saro
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Paige Hickey Townsend
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Kirstin Birtwell
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Lisa Nowinski
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Robyn P Thom
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Michelle L Palumbo
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Christopher Keary
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Christopher J McDougle
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA
| | - Nicole R Zürcher
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA.
| |
Collapse
|
11
|
Morris RH, Counsell SJ, McGonnell IM, Thornton C. Exposure to urban particulate matter (UPM) impairs mitochondrial dynamics in BV2 cells, triggering a mitochondrial biogenesis response. J Physiol 2024; 602:2737-2750. [PMID: 38795332 DOI: 10.1113/jp285978] [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: 11/14/2023] [Accepted: 05/06/2024] [Indexed: 05/27/2024] Open
Abstract
World Health Organisation data suggest that up to 99% of the global population are exposed to air pollutants above recommended levels. Impacts to health range from increased risk of stroke and cardiovascular disease to chronic respiratory conditions, and air pollution may contribute to over 7 million premature deaths a year. Additionally, mounting evidence suggests that in utero or early life exposure to particulate matter (PM) in ambient air pollution increases the risk of neurodevelopmental impairment with obvious lifelong consequences. Identifying brain-specific cellular targets of PM is vital for determining its long-term consequences. We previously established that microglial-like BV2 cells were particularly sensitive to urban (U)PM-induced damage including reactive oxygen species production, which was abrogated by a mitochondrially targeted antioxidant. Here we extend those studies to find that UPM treatment causes a rapid impairment of mitochondrial function and increased mitochondrial fragmentation. However, there is a subsequent restoration of mitochondrial and therefore cell health occurring concomitantly with upregulated measures of mitochondrial biogenesis and mitochondrial load. Our data highlight that protecting mitochondrial function may represent a valuable mechanism to offset the effects of UPM exposure in the neonatal brain. KEY POINTS: Air pollution represents a growing risk to long-term health especially in early life, and the CNS is emerging a target for airborne particulate matter (PM). We previously showed that microglial-like BV2 cells were vulnerable to urban (U)PM exposure, which impaired cell survival and promoted reactive oxygen species production. Here we find that, following UPM exposure, BV2 mitochondrial membrane potential is rapidly reduced, concomitant with decreased cellular bioenergetics and increased mitochondrial fission. However, markers of mitochondrial biogenesis and mitochondrial mass are subsequently induced, which may represent a cellular mitigation strategy. As mitochondria are more vulnerable in the developing brain, exposure to air pollution may represent a greater risk to lifelong health in this cohort; conversely, promoting mitochondrial integrity may offset these risks.
Collapse
Affiliation(s)
- Rebecca H Morris
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Serena J Counsell
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Imelda M McGonnell
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Claire Thornton
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| |
Collapse
|
12
|
Ahmed C, Greve HJ, Garza‐Lombo C, Malley JA, Johnson JA, Oblak AL, Block ML. Peripheral HMGB1 is linked to O 3 pathology of disease-associated astrocytes and amyloid. Alzheimers Dement 2024; 20:3551-3566. [PMID: 38624088 PMCID: PMC11095433 DOI: 10.1002/alz.13825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/07/2024] [Accepted: 02/26/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Ozone (O3) is an air pollutant associated with Alzheimer's disease (AD) risk. The lung-brain axis is implicated in O3-associated glial and amyloid pathobiology; however, the role of disease-associated astrocytes (DAAs) in this process remains unknown. METHODS The O3-induced astrocyte phenotype was characterized in 5xFAD mice by spatial transcriptomics and proteomics. Hmgb1fl/fl LysM-Cre+ mice were used to assess the role of peripheral myeloid cell high mobility group box 1 (HMGB1). RESULTS O3 increased astrocyte and plaque numbers, impeded the astrocyte proteomic response to plaque deposition, augmented the DAA transcriptional fingerprint, increased astrocyte-microglia contact, and reduced bronchoalveolar lavage immune cell HMGB1 expression in 5xFAD mice. O3-exposed Hmgb1fl/fl LysM-Cre+ mice exhibited dysregulated DAA mRNA markers. DISCUSSION Astrocytes and peripheral myeloid cells are critical lung-brain axis interactors. HMGB1 loss in peripheral myeloid cells regulates the O3-induced DAA phenotype. These findings demonstrate a mechanism and potential intervention target for air pollution-induced AD pathobiology. HIGHLIGHTS Astrocytes are part of the lung-brain axis, regulating how air pollution affects plaque pathology. Ozone (O3) astrocyte effects are associated with increased plaques and modified by plaque localization. O3 uniquely disrupts the astrocyte transcriptomic and proteomic disease-associated astrocyte (DAA) phenotype in plaque associated astrocytes (PAA). O3 changes the PAA cell contact with microglia and cell-cell communication gene expression. Peripheral myeloid cell high mobility group box 1 regulates O3-induced transcriptomic changes in the DAA phenotype.
Collapse
Affiliation(s)
- Chandrama Ahmed
- Department of Pharmacology and ToxicologyIndiana University School of MedicineThe Stark Neurosciences Research InstituteIndianapolisIndianaUSA
| | - Hendrik J. Greve
- Department of Pharmacology and ToxicologyIndiana University School of MedicineThe Stark Neurosciences Research InstituteIndianapolisIndianaUSA
| | - Carla Garza‐Lombo
- Department of Pharmacology and ToxicologyIndiana University School of MedicineThe Stark Neurosciences Research InstituteIndianapolisIndianaUSA
| | - Jamie A. Malley
- Department of Pharmacology and ToxicologyIndiana University School of MedicineThe Stark Neurosciences Research InstituteIndianapolisIndianaUSA
| | - James A. Johnson
- Department of Pharmacology and ToxicologyIndiana University School of MedicineThe Stark Neurosciences Research InstituteIndianapolisIndianaUSA
| | - Adrian L. Oblak
- Department of Radiology and Imaging SciencesIndiana University School of MedicineThe Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Michelle L. Block
- Department of Pharmacology and ToxicologyIndiana University School of MedicineThe Stark Neurosciences Research InstituteIndianapolisIndianaUSA
| |
Collapse
|
13
|
Lubrano C, Parisi F, Cetin I. Impact of Maternal Environment and Inflammation on Fetal Neurodevelopment. Antioxidants (Basel) 2024; 13:453. [PMID: 38671901 PMCID: PMC11047368 DOI: 10.3390/antiox13040453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
During intrauterine life, external stimuli including maternal nutrition, lifestyle, socioeconomic conditions, anxiety, stress, and air pollution can significantly impact fetal development. The human brain structures begin to form in the early weeks of gestation and continue to grow and mature throughout pregnancy. This review aims to assess, based on the latest research, the impact of environmental factors on fetal and neonatal brain development, showing that oxidative stress and inflammation are implied as a common factor for most of the stressors. Environmental insults can induce a maternal inflammatory state and modify nutrient supply to the fetus, possibly through epigenetic mechanisms, leading to significant consequences for brain morphogenesis and neurological outcomes. These risk factors are often synergic and mutually reinforcing. Fetal growth restriction and preterm birth represent paradigms of intrauterine reduced nutrient supply and inflammation, respectively. These mechanisms can lead to an increase in free radicals and, consequently, oxidative stress, with well-known adverse effects on the offspring's neurodevelopment. Therefore, a healthy intrauterine environment is a critical factor in supporting normal fetal brain development. Hence, healthcare professionals and clinicians should implement effective interventions to prevent and reduce modifiable risk factors associated with an increased inflammatory state and decreased nutrient supply during pregnancy.
Collapse
Affiliation(s)
- Chiara Lubrano
- Nutritional Sciences, Doctoral Programme (PhD), Università degli Studi di Milano, 20157 Milan, Italy;
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Parisi
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy;
| | - Irene Cetin
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy;
| |
Collapse
|
14
|
Jetton TL, Galbraith OT, Peshavaria M, Bonney EA, Holmén BA, Fukagawa NK. Sex-specific metabolic adaptations from in utero exposure to particulate matter derived from combustion of petrodiesel and biodiesel fuels. CHEMOSPHERE 2024; 346:140480. [PMID: 37879369 PMCID: PMC10841900 DOI: 10.1016/j.chemosphere.2023.140480] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023]
Abstract
Maternal exposure to particulate matter derived from diesel exhaust has been shown to cause metabolic dysregulation, neurological problems, and increased susceptibility to diabetes in the offspring. Diesel exhaust is a major source of air pollution and the use of biodiesel (BD) and its blends have been progressively increasing throughout the world; however, studies on the health impact of BD vs. petrodiesel combustion-generated exhaust have been controversial in part, due to differences in the chemical and physical nature of the associated particulate matter (PM). To explore the long-term impact of prenatal exposure, pregnant mice were exposed to PM generated by combustion of petrodiesel (B0) and a 20% soy BD blend (B20) by intratracheal instillation during embryonic days 9-17 and allowed to deliver. Offspring were then followed for 52 weeks. We found that mother's exposure to B0 and B20 PM manifested in striking sex-specific phenotypes with respect to metabolic adaptation, maintenance of glucose homeostasis, and medial hypothalamic glial cell makeup in the offspring. The data suggest PM exposure limited to a narrower critical developmental window may be compensated for by the mother and/or the fetus by altered metabolic programming in a marked sex-specific and fuel-derived PM-specific manner, leading to sex-specific risk for diseases related to environmental exposure later in life.
Collapse
Affiliation(s)
- Thomas L Jetton
- From the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, USA.
| | - Oban T Galbraith
- From the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, USA
| | - Mina Peshavaria
- From the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, USA
| | | | - Britt A Holmén
- Larner College of Medicine, Department of Civil & Environmental Engineering, College of Engineering and Mathematical Sciences, USA
| | - Naomi K Fukagawa
- From the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, USA; University of Vermont, Burlington, VT 05405, USA; USDA-ARS, Beltsville Human Nutrition Research Center, Beltsville, MD 20705-2350, USA
| |
Collapse
|
15
|
Tamayo JM, Osman HC, Schwartzer JJ, Pinkerton KE, Ashwood P. Characterizing the neuroimmune environment of offspring in a novel model of maternal allergic asthma and particulate matter exposure. J Neuroinflammation 2023; 20:252. [PMID: 37919762 PMCID: PMC10621097 DOI: 10.1186/s12974-023-02930-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
Inflammation during pregnancy is associated with an increased risk for neurodevelopmental disorders (NDD). Increased gestational inflammation can be a result of an immune condition/disease, exposure to infection, and/or environmental factors. Epidemiology studies suggest that cases of NDD are on the rise. Similarly, rates of asthma are increasing, and the presence of maternal asthma during pregnancy increases the likelihood of a child being later diagnosed with NDD such as autism spectrum disorders (ASD). Particulate matter (PM), via air pollution, is an environmental factor known to worsen the symptoms of asthma, but also, PM has been associated with increased risk of neuropsychiatric disorders. Despite the links between asthma and PM with neuropsychiatric disorders, there is a lack of laboratory models investigating combined prenatal exposure to asthma and PM on offspring neurodevelopment. Thus, we developed a novel mouse model that combines exposure to maternal allergic asthma (MAA) and ultrafine iron-soot (UIS), a common component of PM. In the current study, female BALB/c mice were sensitized for allergic asthma with ovalbumin (OVA) prior to pregnancy. Following mating and beginning on gestational day 2 (GD2), dams were exposed to either aerosolized OVA to induce allergic asthma or phosphate buffered saline (PBS) for 1 h. Following the 1-h exposure, pregnant females were then exposed to UIS with a size distribution of 55 to 169 nm at an average concentration of 176 ± 45 μg/m3) (SD), or clean air for 4 h, over 8 exposure sessions. Offspring brains were collected at postnatal days (P)15 and (P)35. Cortices and hippocampal regions were then isolated and assessed for changes in cytokines using a Luminex bead-based multiplex assay. Analyses identified changes in many cytokines across treatment groups at both timepoints in the cortex, including interleukin-1 beta (IL-1β), and IL-17, which remained elevated from P15 to P35 in all treatment conditions compared to controls. There was a suppressive effect of the combined MAA plus UIS on the anti-inflammatory cytokine IL-10. Potentially shifting the cytokine balance towards more neuroinflammation. In the hippocampus at P15, elevations in cytokines were also identified across the treatment groups, namely IL-7. The combination of MAA and UIS exposure (MAA-UIS) during pregnancy resulted in an increase in microglia density in the hippocampus of offspring, as identified by IBA-1 staining. Together, these data indicate that exposure to MAA, UIS, and MAA-UIS result in changes in the neuroimmune environment of offspring that persist into adulthood.
Collapse
Affiliation(s)
- Juan M Tamayo
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, 2805, 50th Street Sacramento, Davis, CA, 95817, USA
| | - Hadley C Osman
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, 2805, 50th Street Sacramento, Davis, CA, 95817, USA
| | - Jared J Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA, 01075, USA
| | - Kent E Pinkerton
- Center for Health and the Environment, University of California at Davis, Davis, CA, 95616, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, 2805, 50th Street Sacramento, Davis, CA, 95817, USA.
| |
Collapse
|
16
|
Hou Y, Yan W, Li G, Sang N. Transcriptome sequencing analysis reveals a potential role of lncRNA NONMMUT058932.2 and NONMMUT029203.2 in abnormal myelin development of male offspring following prenatal PM 2.5 exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 895:165004. [PMID: 37348736 DOI: 10.1016/j.scitotenv.2023.165004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/27/2023] [Accepted: 06/17/2023] [Indexed: 06/24/2023]
Abstract
Numerous epidemiological studies have shown that PM2.5 exposure in early life can influence brain development and increase the risk of neurodevelopmental disorders in boys, but the underlying molecular mechanisms remain unclear. In the current study, pregnant C57BL/6 J mice were oropharyngeally administered with PM2.5 suspension (3mg/kg/2 days) until the birth of offspring. Based on mRNA expression profiles, two-way analysis of variance (two-way ANOVA) and weighted gene co-expression network analysis (WGCNA) were conducted to explore the most impacted neurodevelopmental processes in male offspring and the most significantly associated gene modules. Gene Ontology (GO) enrichment and Encyclopedia of Genes and Genomes (KEGG) pathway analyses suggested that prenatal PM2.5 exposure significantly altered several biological processes (such as substrate adhesion-dependent cell spreading, myelination, and ensheathment of neurons) and KEGG pathways (such as tight junction and axon guidance). We further found that PM2.5 exposure significantly changed the expression of myelination-related genes in male offspring during postnatal development and impaired myelin ultrastructure on PNDs 14 and 21, as demonstrated by the decreased thickness of myelin sheaths in the optic nerves, and mild loss of myelin in the corpus callosum. Importantly, lncRNA NONMMUT058932.2 and NONMMUT029203.2 played key roles in abnormal myelination by regulating the expression of several myelination-related genes (Fa2h, Mal, Sh3tc2, Trf and Tppp) through the binding to transcription factor Ctcf. Our work provides genomic evidence for prenatal PM2.5 exposure-induced neurodevelopmental disorders in male offspring.
Collapse
Affiliation(s)
- Yanwen Hou
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Wei Yan
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China.
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
17
|
Reddaway J, Richardson PE, Bevan RJ, Stoneman J, Palombo M. Microglial morphometric analysis: so many options, so little consistency. Front Neuroinform 2023; 17:1211188. [PMID: 37637472 PMCID: PMC10448193 DOI: 10.3389/fninf.2023.1211188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/05/2023] [Indexed: 08/29/2023] Open
Abstract
Quantification of microglial activation through morphometric analysis has long been a staple of the neuroimmunologist's toolkit. Microglial morphological phenomics can be conducted through either manual classification or constructing a digital skeleton and extracting morphometric data from it. Multiple open-access and paid software packages are available to generate these skeletons via semi-automated and/or fully automated methods with varying degrees of accuracy. Despite advancements in methods to generate morphometrics (quantitative measures of cellular morphology), there has been limited development of tools to analyze the datasets they generate, in particular those containing parameters from tens of thousands of cells analyzed by fully automated pipelines. In this review, we compare and critique the approaches using cluster analysis and machine learning driven predictive algorithms that have been developed to tackle these large datasets, and propose improvements for these methods. In particular, we highlight the need for a commitment to open science from groups developing these classifiers. Furthermore, we call attention to a need for communication between those with a strong software engineering/computer science background and neuroimmunologists to produce effective analytical tools with simplified operability if we are to see their wide-spread adoption by the glia biology community.
Collapse
Affiliation(s)
- Jack Reddaway
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Innovation Institute (NMHII), Cardiff University, Cardiff, United Kingdom
| | | | - Ryan J. Bevan
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Jessica Stoneman
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Marco Palombo
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- School of Computer Science and Informatics, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
18
|
Tamayo JM, Osman HC, Schwartzer JJ, Pinkerton K, Ashwood P. Characterizing the Neuroimmune Environment of Offspring in a Novel Model of Maternal Allergic Asthma and Particulate Matter Exposure. RESEARCH SQUARE 2023:rs.3.rs-3140415. [PMID: 37503062 PMCID: PMC10371118 DOI: 10.21203/rs.3.rs-3140415/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental disorders characterized by the presence of decreased social interactions and an increase in stereotyped and repetitive behaviors. Epidemiology studies suggest that cases of ASD are on the rise. Similarly, rates of asthma are increasing, and the presence of maternal asthma during pregnancy increases the likelihood of a child being later diagnosed with ASD. Particulate matter (PM), via air pollution, is an environmental factor known to worsen the symptoms of asthma, but also, PM has been associated with increased risk of neuropsychiatric disorders including ASD. Despite the links between asthma and PM with neuropsychiatric disorders, there is a lack of laboratory models investigating combined prenatal exposure to asthma and PM on offspring neurodevelopment. Thus, we developed a novel mouse model that combines exposure to maternal allergic asthma (MAA) and ultrafine iron-soot (UIS), a common component of PM. In the current study, female BALB/c mice were primed for allergic asthma with ovalbumin (OVA) prior to pregnancy. Following mating and beginning on gestational day 2 (GD2), dams were exposed to either aerosolized OVA or phosphate buffered saline (PBS) for 1 hour. Following the 1-hour exposure, pregnant females were then exposed to UIS or clean air for 4 hours. Offspring brains were collected at postnatal days (P)15 and (P)35. Cortices and hippocampal regions were then isolated and assessed for changes in cytokines using a Luminex bead-based multiplex assay. Analyses identified changes in many cytokines across treatment groups at both timepoints in the cortex, including interleukin-1 beta (IL-1β), IL-2, IL-13, and IL-17, which remained elevated from P15 to P35 in all treatment conditions compared to controls. In the hippocampus at P15, elevations in cytokines were also identified across the treatment groups, namely interferon gamma (IFNγ) and IL-7. The combination of MAA and UIS exposure (MAA-UIS) during pregnancy resulted in an increase in microglia density in the hippocampus of offspring, as identified by IBA-1 staining. Together, these data indicate that exposure to MAA, UIS, and MAA-UIS result in changes in the neuroimmune environment of offspring that persist into adulthood.
Collapse
|
19
|
Costa AN, Ferguson BJ, Hawkins E, Coman A, Schauer J, Ramirez-Celis A, Hecht PM, Bruce D, Tilley M, Talebizadeh Z, Van de Water J, Beversdorf DQ. The Relationship between Maternal Antibodies to Fetal Brain and Prenatal Stress Exposure in Autism Spectrum Disorder. Metabolites 2023; 13:663. [PMID: 37233704 PMCID: PMC10224143 DOI: 10.3390/metabo13050663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Environmental and genetic factors contribute to the etiology of autism spectrum disorder (ASD), but their interaction is less well understood. Mothers that are genetically more stress-susceptible have been found to be at increased risk of having a child with ASD after exposure to stress during pregnancy. Additionally, the presence of maternal antibodies for the fetal brain is associated with a diagnosis of ASD in children. However, the relationship between prenatal stress exposure and maternal antibodies in the mothers of children diagnosed with ASD has not yet been addressed. This exploratory study examined the association of maternal antibody response with prenatal stress and a diagnosis of ASD in children. Blood samples from 53 mothers with at least one child diagnosed with ASD were examined by ELISA. Maternal antibody presence, perceived stress levels during pregnancy (high or low), and maternal 5-HTTLPR polymorphisms were examined for their interrelationship in ASD. While high incidences of prenatal stress and maternal antibodies were found in the sample, they were not associated with each other (p = 0.709, Cramér's V = 0.051). Furthermore, the results revealed no significant association between maternal antibody presence and the interaction between 5-HTTLPR genotype and stress (p = 0.729, Cramér's V = 0.157). Prenatal stress was not found to be associated with the presence of maternal antibodies in the context of ASD, at least in this initial exploratory sample. Despite the known relationship between stress and changes in immune function, these results suggest that prenatal stress and immune dysregulation are independently associated with a diagnosis of ASD in this study population, rather than acting through a convergent mechanism. However, this would need to be confirmed in a larger sample.
Collapse
Affiliation(s)
- Amy N Costa
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Bradley J Ferguson
- Department of Health Psychology, University of Missouri, Columbia, MO 65211, USA
- Thompson Center for Autism and Neurodevelopment, University of Missouri, Columbia, MO 65211, USA
- Interdiscipinary Neuroscience Program, University of Missouri, Columbia, MO 65211, USA
| | - Emily Hawkins
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA
- Thompson Center for Autism and Neurodevelopment, University of Missouri, Columbia, MO 65211, USA
- Department of Biological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Adriana Coman
- Department of Biochemistry, Grinnell College, Grinnell, IA 50112, USA
| | - Joseph Schauer
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Immunology, University of California, Davis, CA 95161, USA
| | - Alex Ramirez-Celis
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Immunology, University of California, Davis, CA 95161, USA
| | - Patrick M Hecht
- Interdiscipinary Neuroscience Program, University of Missouri, Columbia, MO 65211, USA
| | - Danielle Bruce
- Department of Biology, Central Methodist University, Fayette, MO 65248, USA
| | - Michael Tilley
- Department of Biology, Central Methodist University, Fayette, MO 65248, USA
| | - Zohreh Talebizadeh
- The American College of Medical Genetics and Genomics, Bethesda, MD 20814, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Immunology, University of California, Davis, CA 95161, USA
| | - David Q Beversdorf
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA
- Thompson Center for Autism and Neurodevelopment, University of Missouri, Columbia, MO 65211, USA
- Interdiscipinary Neuroscience Program, University of Missouri, Columbia, MO 65211, USA
- Departments of Radiology and Neurology, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
20
|
Sun X, Liu C, Ji H, Li W, Miao M, Yuan W, Yuan Z, Liang H, Kan H. Prenatal exposure to ambient PM 2.5 and its chemical constituents and child intelligence quotient at 6 years of age. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 255:114813. [PMID: 36948012 DOI: 10.1016/j.ecoenv.2023.114813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 06/18/2023]
Abstract
There are limited studies on the associations between prenatal exposure to constituents of fine particulate matter (PM2.5) and children's intelligence quotient (IQ). Our study aimed to explore the associations between prenatal PM2.5 and its six constituents and the IQ levels of 6-year-old children. We included 512 mother-child pairs. We used a satellite-based modelling framework to estimate prenatal PM2.5 and its six constituents (ammonium, sulfate, nitrate, organic carbon, soil dust, and black carbon). We assessed the children's IQ using the short form of the Wechsler Intelligence Scale. Perceptual Reasoning Index (PRI), Verbal Comprehension Index (VCI), and Full Scale IQ (FSIQ) scores were computed. The multiple informant model (MIM) was applied to explore the trimester specific effects of PM2.5 and its six constituents' exposure on children's PRI, VCI, and FSIQ. To examine whether the duration of breastfeeding and physical activity (PA) could modify the effects of PM2.5 on children's IQ, we stratified the analyses according to the duration of breastfeeding (≤6 and >6 months) and time of outdoor activities after school (≤2 and >2 h/week). The first trimester PM2.5 and its five constituents' exposures were inversely associated with FSIQ [β = -1.34, 95 % confidence interval [CI] (-2.71, 0.04) for PM2.5] and PRI [β = -2.18, 95 %CI (-3.80, -0.57) for PM2.5] in children. The associations were magnified among boys and those with less outdoor activities or shorter breastfeeding duration. Our results indicate that prenatal PM2.5 and several of its main constituents' exposure may disrupt cognitive development in children aged 6 years. More PA and longer breastfeeding duration may alleviate the detrimental effects of prenatal PM2.5 exposure on children's cognitive function.
Collapse
Affiliation(s)
- Xiaowei Sun
- NHC Key Lab. Of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Cong Liu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
| | - Honglei Ji
- NHC Key Lab. Of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Weihua Li
- NHC Key Lab. Of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Maohua Miao
- NHC Key Lab. Of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Wei Yuan
- NHC Key Lab. Of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Hong Liang
- NHC Key Lab. Of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China.
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education, NHC Key Lab of Health Technology Assessment, IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Bos B, Barratt B, Batalle D, Gale-Grant O, Hughes EJ, Beevers S, Cordero-Grande L, Price AN, Hutter J, Hajnal JV, Kelly FJ, David Edwards A, Counsell SJ. Prenatal exposure to air pollution is associated with structural changes in the neonatal brain. ENVIRONMENT INTERNATIONAL 2023; 174:107921. [PMID: 37058974 PMCID: PMC10410199 DOI: 10.1016/j.envint.2023.107921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Prenatal exposure to air pollution is associated with adverse neurologic consequences in childhood. However, the relationship between in utero exposure to air pollution and neonatal brain development is unclear. METHODS We modelled maternal exposure to nitrogen dioxide (NO2) and particulate matter (PM2.5 and PM10) at postcode level between date of conception to date of birth and studied the effect of prenatal air pollution exposure on neonatal brain morphology in 469 (207 male) healthy neonates, with gestational age of ≥36 weeks. Infants underwent MR neuroimaging at 3 Tesla at 41.29 (36.71-45.14) weeks post-menstrual age (PMA) as part of the developing human connectome project (dHCP). Single pollutant linear regression and canonical correlation analysis (CCA) were performed to assess the relationship between air pollution and brain morphology, adjusting for confounders and correcting for false discovery rate. RESULTS Higher exposure to PM10 and lower exposure to NO2 was strongly canonically correlated to a larger relative ventricular volume, and moderately associated with larger relative size of the cerebellum. Modest associations were detected with higher exposure to PM10 and lower exposure to NO2 and smaller relative cortical grey matter and amygdala and hippocampus, and larger relaive brainstem and extracerebral CSF volume. No associations were found with white matter or deep grey nuclei volume. CONCLUSIONS Our findings show that prenatal exposure to air pollution is associated with altered brain morphometry in the neonatal period, albeit with opposing results for NO2 and PM10. This finding provides further evidence that reducing levels of maternal exposure to particulate matter during pregnancy should be a public health priority and highlights the importance of understanding the impacts of air pollution on this critical development window.
Collapse
Affiliation(s)
- Brendan Bos
- MRC Centre for Environment and Health, Imperial College London, UK
| | - Ben Barratt
- MRC Centre for Environment and Health, Imperial College London, UK
| | - Dafnis Batalle
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Department of Forensic and Neurodevelopmental Science, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Oliver Gale-Grant
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Department of Forensic and Neurodevelopmental Science, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Emer J Hughes
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Sean Beevers
- MRC Centre for Environment and Health, Imperial College London, UK
| | - Lucilio Cordero-Grande
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK; Biomedical Image Technologies, ETSI Telecomunicación, Universidad Politécnica de Madrid and CIBER-BBN, Madrid, Spain
| | - Anthony N Price
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Jana Hutter
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Joseph V Hajnal
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Frank J Kelly
- MRC Centre for Environment and Health, Imperial College London, UK
| | - A David Edwards
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Serena J Counsell
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK.
| |
Collapse
|
22
|
Wong FK, Favuzzi E. The brain's polymath: Emerging roles of microglia throughout brain development. Curr Opin Neurobiol 2023; 79:102700. [PMID: 36848726 DOI: 10.1016/j.conb.2023.102700] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 01/29/2023] [Accepted: 02/05/2023] [Indexed: 02/27/2023]
Abstract
Microglia, the resident brain immune cells, have garnered a reputation as major effectors of circuit wiring due to their ability to prune synapses. Other roles of microglia in regulating neuronal circuit development have so far received comparatively less attention. Here, we review the latest studies that have contributed to our increased understanding of how microglia regulate brain wiring beyond their role in synapse pruning. We summarize recent findings showing that microglia regulate neuronal numbers and influence neuronal connectivity through a bidirectional communication between microglia and neurons, processes regulated by neuronal activity and the remodeling of the extracellular matrix. Finally, we speculate on the potential contribution of microglia to the development of functional networks and propose an integrative view of microglia as active elements of neural circuits.
Collapse
Affiliation(s)
- Fong Kuan Wong
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, UK. https://twitter.com/brainotopia
| | - Emilia Favuzzi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06519, USA; Wu Tsai Institute, Yale University, New Haven, CT, 06519, USA.
| |
Collapse
|
23
|
Greve HJ, Dunbar AL, Lombo CG, Ahmed C, Thang M, Messenger EJ, Mumaw CL, Johnson JA, Kodavanti UP, Oblak AL, Block ML. The bidirectional lung brain-axis of amyloid-β pathology: ozone dysregulates the peri-plaque microenvironment. Brain 2023; 146:991-1005. [PMID: 35348636 PMCID: PMC10169526 DOI: 10.1093/brain/awac113] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/07/2022] [Accepted: 02/27/2022] [Indexed: 11/14/2022] Open
Abstract
The mechanisms underlying how urban air pollution affects Alzheimer's disease (AD) are largely unknown. Ozone (O3) is a reactive gas component of air pollution linked to increased AD risk, but is confined to the respiratory tract after inhalation, implicating the peripheral immune response to air pollution in AD neuropathology. Here, we demonstrate that O3 exposure impaired the ability of microglia, the brain's parenchymal immune cells, to associate with and form a protective barrier around Aβ plaques, leading to augmented dystrophic neurites and increased Aβ plaque load. Spatial proteomic profiling analysis of peri-plaque proteins revealed a microenvironment-specific signature of dysregulated disease-associated microglia protein expression and increased pathogenic molecule levels with O3 exposure. Unexpectedly, 5xFAD mice exhibited an augmented pulmonary cell and humoral immune response to O3, supporting that ongoing neuropathology may regulate the peripheral O3 response. Circulating HMGB1 was one factor upregulated in only 5xFAD mice, and peripheral HMGB1 was separately shown to regulate brain Trem2 mRNA expression. These findings demonstrate a bidirectional lung-brain axis regulating the central and peripheral AD immune response and highlight this interaction as a potential novel therapeutic target in AD.
Collapse
Affiliation(s)
- Hendrik J Greve
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - August L Dunbar
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Carla Garza Lombo
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chandrama Ahmed
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Morrent Thang
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Evan J Messenger
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christen L Mumaw
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James A Johnson
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Urmila P Kodavanti
- Cardiopulmonary and Immunotoxicology Branch, Public Health and Integrated Toxicology Division, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Adrian L Oblak
- Department of Radiology and Imaging Sciences, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michelle L Block
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA
| |
Collapse
|
24
|
Yang Y, Yang T, Zhou J, Cao Z, Liao Z, Zhao Y, Su X, He J, Hua J. Prenatal exposure to concentrated ambient PM 2.5 results in spatial memory defects regulated by DNA methylation in male mice offspring. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:35142-35152. [PMID: 36526934 PMCID: PMC10017658 DOI: 10.1007/s11356-022-24663-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Ambient fine particulate matter (PM2.5) exposures during pregnancy could lead to adverse birth outcomes, including neurobehavioral development defects. However, limited studies explored the effects and potential epigenetic mechanisms of maternal PM2.5 exposure on offspring spatial memory defects. This study aims to explore the effects and underlying epigenetic mechanisms of maternal concentrated ambient PM2.5 exposure in male mice offspring with spatial memory defects. Pregnant female C57BL/6 mice were exposed daily to concentrated ambient PM2.5 (CAP) or filtered air (FA) throughout gestation, with the concentration of particulates (102.99 ± 78.74 μg/m3) and (2.78 ± 1.19 μg/m3), respectively. Adult male mice offspring were subsequently assessed for spatial learning and memory ability using Morris Water Maze tests and locomotor activities in open field tests. The hippocampus of the male mice offspring was harvested to test mRNA expression and DNA methylation. Results from the probe test of Morris Water Maze showed that the mice offspring in the CAP group had shorter swimming distance travelled in the target quadrant, shorter duration in the target quadrant, and less number of entries into the target quadrant (p < 0.05), suggesting spatial memory impairments. The acquisition trials of Morris Water Maze did not show a significant difference in learning ability between the groups. The mRNA level of interleukin 6 (IL-6) in the CAP group hippocampus (10.80 ± 7.03) increased significantly compared to the FA group (1.08 ± 0.43). Interestingly, the methylation levels of the CpG sites in the IL-6 promoter region declined significantly in the CAP group, (5.66 ± 0.83)% vs. (4.79 ± 0.48)%. Prenatal exposure to concentrated ambient PM2.5 induced long-lasting spatial memory defects in male mice offspring. The underlying biological mechanism might be mediated by an inflammatory reaction which is regulated by DNA methylation.
Collapse
Affiliation(s)
- Yingying Yang
- Department of Women and Children's Health Care, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Tingting Yang
- Department of Social Medicine, School of Public Health, Fudan University, Shanghai, China
| | - Ji Zhou
- Shanghai Key Laboratory of Meteorology and Health, Shanghai Meteorological Bureau, Shanghai, China
- Shanghai Typhoon Institute, CMA, Shanghai, China
- Department of Atmospheric and Oceanic Sciences, & Institute of Atmospheric Sciences, Fudan University, Shanghai, China
| | - Zhijuan Cao
- Department of Women and Children's Health Care, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Yan Zhao
- Department of Women and Children's Health Care, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiujuan Su
- Department of Women and Children's Health Care, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jia He
- School of Medicine, Tongji University, Shanghai, China
| | - Jing Hua
- Department of Women and Children's Health Care, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
25
|
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: 13] [Impact Index Per Article: 6.5] [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
|
26
|
Peterson BS, Bansal R, Sawardekar S, Nati C, Elgabalawy ER, Hoepner LA, Garcia W, Hao X, Margolis A, Perera F, Rauh V. Prenatal exposure to air pollution is associated with altered brain structure, function, and metabolism in childhood. J Child Psychol Psychiatry 2022; 63:1316-1331. [PMID: 35165899 DOI: 10.1111/jcpp.13578] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Prenatal exposure to air pollution disrupts cognitive, emotional, and behavioral development. The brain disturbances associated with prenatal air pollution are largely unknown. METHODS In this prospective cohort study, we estimated prenatal exposures to fine particulate matter (PM2.5 ) and polycyclic aromatic hydrocarbons (PAH), and then assessed their associations with measures of brain anatomy, tissue microstructure, neurometabolites, and blood flow in 332 youth, 6-14 years old. We then assessed how those brain disturbances were associated with measures of intelligence, ADHD and anxiety symptoms, and socialization. RESULTS Both exposures were associated with thinning of dorsal parietal cortices and thickening of postero-inferior and mesial wall cortices. They were associated with smaller white matter volumes, reduced organization in white matter of the internal capsule and frontal lobe, higher metabolite concentrations in frontal cortex, reduced cortical blood flow, and greater microstructural organization in subcortical gray matter nuclei. Associations were stronger for PM2.5 in boys and PAH in girls. Youth with low exposure accounted for most significant associations of ADHD, anxiety, socialization, and intelligence measures with cortical thickness and white matter volumes, whereas it appears that high exposures generally disrupted these neurotypical brain-behavior associations, likely because strong exposure-related effects increased the variances of these brain measures. CONCLUSIONS The commonality of effects across exposures suggests PM2.5 and PAH disrupt brain development through one or more common molecular pathways, such as inflammation or oxidative stress. Progressively higher exposures were associated with greater disruptions in local volumes, tissue organization, metabolite concentrations, and blood flow throughout cortical and subcortical brain regions and the white matter pathways interconnecting them. Together these affected regions comprise cortico-striato-thalamo-cortical circuits, which support the regulation of thought, emotion, and behavior.
Collapse
Affiliation(s)
- Bradley S Peterson
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Department of Psychiatry, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Ravi Bansal
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Siddhant Sawardekar
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Carlo Nati
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Eman R Elgabalawy
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Lori A Hoepner
- Department of Environmental and Occupational Health Sciences, SUNY Downstate School of Public Health, Brooklyn, NY, USA
| | - Wanda Garcia
- Heilbrunn Department of Population and Family Health, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Xuejun Hao
- Department of Psychiatry, Columbia Presbyterian Medical Center & New York State Psychiatric Institute, New York, NY, USA
| | - Amy Margolis
- Department of Psychiatry, Columbia Presbyterian Medical Center & New York State Psychiatric Institute, New York, NY, USA
| | - Frederica Perera
- Columbia Center for Children's Environmental Health, New York, NY, USA.,Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Virginia Rauh
- Heilbrunn Department of Population and Family Health, Mailman School of Public Health, Columbia University, New York, NY, USA.,Columbia Center for Children's Environmental Health, New York, NY, USA
| |
Collapse
|
27
|
Block CL, Eroglu O, Mague SD, Smith CJ, Ceasrine AM, Sriworarat C, Blount C, Beben KA, Malacon KE, Ndubuizu N, Talbot A, Gallagher NM, Chan Jo Y, Nyangacha T, Carlson DE, Dzirasa K, Eroglu C, Bilbo SD. Prenatal environmental stressors impair postnatal microglia function and adult behavior in males. Cell Rep 2022; 40:111161. [PMID: 35926455 PMCID: PMC9438555 DOI: 10.1016/j.celrep.2022.111161] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 05/18/2022] [Accepted: 07/13/2022] [Indexed: 12/14/2022] Open
Abstract
Gestational exposure to environmental toxins and socioeconomic stressors is epidemiologically linked to neurodevelopmental disorders with strong male bias, such as autism. We model these prenatal risk factors in mice by co-exposing pregnant dams to an environmental pollutant and limited-resource stress, which robustly activates the maternal immune system. Only male offspring display long-lasting behavioral abnormalities and alterations in the activity of brain networks encoding social interactions. Cellularly, prenatal stressors diminish microglial function within the anterior cingulate cortex, a central node of the social coding network, in males during early postnatal development. Precise inhibition of microglial phagocytosis within the anterior cingulate cortex (ACC) of wild-type (WT) mice during the same critical period mimics the impact of prenatal stressors on a male-specific behavior, indicating that environmental stressors alter neural circuit formation in males via impairing microglia function during development. Block et al. show that combined exposure to air pollution and maternal stress during pregnancy activates the maternal immune system and induces male-specific impairments in social behavior and circuit connectivity in offspring. Cellularly, prenatal stressors diminish microglia phagocytic function, and inhibition of microglia phagocytosis phenocopies behavioral deficits from prenatal stressors.
Collapse
Affiliation(s)
- Carina L Block
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA
| | - Oznur Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Stephen D Mague
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Caroline J Smith
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA
| | - Alexis M Ceasrine
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA
| | | | - Cameron Blount
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kathleen A Beben
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA
| | - Karen E Malacon
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA
| | - Nkemdilim Ndubuizu
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Austin Talbot
- Department of Statistical Science, Duke University, Durham, NC 27710, USA
| | - Neil M Gallagher
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Young Chan Jo
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA
| | - Timothy Nyangacha
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - David E Carlson
- Department of Civil and Environmental Engineering, Duke University, Durham, NC 27710, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - Kafui Dzirasa
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences, Durham, NC 27710, USA; Howard Hughes Medical Institute, Duke University, Durham, NC 27710, USA.
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences, Durham, NC 27710, USA; Howard Hughes Medical Institute, Duke University, Durham, NC 27710, USA.
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences, Durham, NC 27710, USA; Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
28
|
Koshko L, Scofield S, Mor G, Sadagurski M. Prenatal Pollutant Exposures and Hypothalamic Development: Early Life Disruption of Metabolic Programming. Front Endocrinol (Lausanne) 2022; 13:938094. [PMID: 35909533 PMCID: PMC9327615 DOI: 10.3389/fendo.2022.938094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Environmental contaminants in ambient air pollution pose a serious risk to long-term metabolic health. Strong evidence shows that prenatal exposure to pollutants can significantly increase the risk of Type II Diabetes (T2DM) in children and all ethnicities, even without the prevalence of obesity. The central nervous system (CNS) is critical in regulating whole-body metabolism. Within the CNS, the hypothalamus lies at the intersection of the neuroendocrine and autonomic systems and is primarily responsible for the regulation of energy homeostasis and satiety signals. The hypothalamus is particularly sensitive to insults during early neurodevelopmental periods and may be susceptible to alterations in the formation of neural metabolic circuitry. Although the precise molecular mechanism is not yet defined, alterations in hypothalamic developmental circuits may represent a leading cause of impaired metabolic programming. In this review, we present the current knowledge on the links between prenatal pollutant exposure and the hypothalamic programming of metabolism.
Collapse
Affiliation(s)
- Lisa Koshko
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Sydney Scofield
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology School of Medicine, Wayne State University, Detroit, MI, United States
| | - Marianna Sadagurski
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| |
Collapse
|
29
|
Indirect mediators of systemic health outcomes following nanoparticle inhalation exposure. Pharmacol Ther 2022; 235:108120. [PMID: 35085604 PMCID: PMC9189040 DOI: 10.1016/j.pharmthera.2022.108120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023]
Abstract
The growing field of nanoscience has shed light on the wide diversity of natural and anthropogenic sources of nano-scale particulates, raising concern as to their impacts on human health. Inhalation is the most robust route of entry, with nanoparticles (NPs) evading mucociliary clearance and depositing deep into the alveolar region. Yet, impacts from inhaled NPs are evident far outside the lung, particularly on the cardiovascular system and highly vascularized organs like the brain. Peripheral effects are partly explained by the translocation of some NPs from the lung into the circulation; however, other NPs largely confined to the lung are still accompanied by systemic outcomes. Omic research has only just begun to inform on the complex myriad of molecules released from the lung to the blood as byproducts of pulmonary pathology. These indirect mediators are diverse in their molecular make-up and activity in the periphery. The present review examines systemic outcomes attributed to pulmonary NP exposure and what is known about indirect pathological mediators released from the lung into the circulation. Further focus was directed to outcomes in the brain, a highly vascularized region susceptible to acute and longer-term outcomes. Findings here support the need for big-data toxicological studies to understand what drives these health outcomes and better predict, circumvent, and treat the potential health impacts arising from NP exposure scenarios.
Collapse
|
30
|
Marinello WP, Gillera SEA, Fanning MJ, Malinsky LB, Rhodes CL, Horman BM, Patisaul HB. Effects of developmental exposure to FireMaster® 550 (FM 550) on microglia density, reactivity and morphology in a prosocial animal model. Neurotoxicology 2022; 91:140-154. [PMID: 35526706 DOI: 10.1016/j.neuro.2022.04.015] [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: 02/01/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 11/20/2022]
Abstract
Microglia are known to shape brain sex differences critical for social and reproductive behaviors. Chemical exposures can disrupt brain sexual differentiation but there is limited data regarding how they may impact microglia distribution and function. We focused on the prevalent flame retardant mixture Firemaster 550 (FM 550) which is used in foam-based furniture and infant products including strollers and nursing pillows because it disrupts sexually dimorphic behaviors. We hypothesized early life FM 550 exposure would disrupt microglial distribution and reactivity in brain regions known to be highly sexually dimorphic or associated with social disorders in humans. We used prairie voles (Microtus ochrogaster) because they display spontaneous prosocial behaviors not seen in rats or mice and are thus a powerful model for studying chemical exposure-related impacts on social behaviors and their underlying neural systems. We have previously demonstrated that perinatal FM 550 exposure sex-specifically impacts socioemotional behaviors in prairie voles. We first established that, unlike in rats, the postnatal colonization of the prairie vole brain is not sexually dimorphic. Vole dams were then exposed to FM 550 (0, 500, 1000, 2000 µg/day) via subcutaneous injections through gestation, and pups were directly exposed beginning the day after birth until weaning. Adult offspring's brains were assessed for number and type (ramified, intermediate, ameboid) of microglia in the medial prefrontal cortex (mPFC), cerebellum (lobules VI-VII) and amygdala. Effects were sex- and dose-specific in the regions of interests. Overall, FM 550 exposure resulted in reduced numbers of microglia in most regions examined, with the 1000 µg FM 550 exposed males particularly affected. To further quantify differences in microglia morphology in the 1000 µg FM 550 group, Sholl and skeleton analysis were carried out on individual microglia. Microglia from control females had a more ramified phenotype compared to control males while 1000 µg FM 550-exposed males had decreased branching and ramification compared to same-sex controls. Future studies will examine the impact on the exposure to FM 550 on microglia during development given the critical role of these cells in shaping neural circuits.
Collapse
Affiliation(s)
- William P Marinello
- Department of Biological Sciences, NC State University, Raleigh, NC 27695, USA
| | | | - Marley J Fanning
- Department of Biological Sciences, NC State University, Raleigh, NC 27695, USA
| | - Lacey B Malinsky
- Department of Biological Sciences, NC State University, Raleigh, NC 27695, USA
| | - Cassie L Rhodes
- Department of Biological Sciences, NC State University, Raleigh, NC 27695, USA
| | - Brian M Horman
- Department of Biological Sciences, NC State University, Raleigh, NC 27695, USA
| | - Heather B Patisaul
- Department of Biological Sciences, NC State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, NC State University, Raleigh, NC 27695, USA.
| |
Collapse
|
31
|
Cardenas-Iniguez C, Burnor E, Herting MM. Neurotoxicants, the Developing Brain, and Mental Health. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 2:223-232. [PMID: 35911498 PMCID: PMC9337627 DOI: 10.1016/j.bpsgos.2022.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 12/31/2022] Open
Abstract
While life in urban environments may confer a number of benefits, it may also result in a variety of exposures, with toxic consequences for neurodevelopment and neuropsychological health. Neurotoxicants are any of a large number of chemicals or substances that interfere with normal function and/or compromise adaptation in the central and/or peripheral nervous system. Evidence suggests that neurotoxicant effects have a greater effect when occurring in utero and during early childhood. Recent findings exploring neural-level mechanisms provide a crucial opportunity to explore the ways in which environmental conditions may get "under the skin" to impact a number of psychological behaviors and cognitive processes, ultimately allowing for greater synergy between macro- and microlevel efforts to improve mental health in the presence of neurotoxicant exposures. In this review, we provide an overview of 3 types of neurotoxicants related to the built environment and relevant to brain development during childhood and adolescence: lead exposure, outdoor particulate matter pollution, and endocrine-disrupting chemicals. We also discuss mechanisms through which these neurotoxicants affect central nervous system function, including recent evidence from neuroimaging literature. Furthermore, we discuss neurotoxicants and mental health during development in the context of social determinants and how differences in the spatial distribution of neurotoxicant exposures result in health disparities that disproportionately affect low-income and minority populations. Multifaceted approaches incorporating social systems and their effect on neurotoxicant exposures and downstream mental health will be key to reduce societal costs and improve quality of life for children, adolescents, and adults.
Collapse
Affiliation(s)
- Carlos Cardenas-Iniguez
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Elisabeth Burnor
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Megan M. Herting
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, California
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, California
| |
Collapse
|
32
|
Guilloteau E, Coll P, Lu Z, Djouina M, Cazaunau M, Waxin C, Bergé A, Caboche S, Gratien A, Al Marj E, Hot D, Dubuquoy L, Launay D, Vignal C, Lanone S, Body-Malapel M. Murine in utero exposure to simulated complex urban air pollution disturbs offspring gut maturation and microbiota during intestinal suckling-to-weaning transition in a sex-dependent manner. Part Fibre Toxicol 2022; 19:41. [PMID: 35706036 PMCID: PMC9199156 DOI: 10.1186/s12989-022-00481-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/25/2022] [Indexed: 11/23/2022] Open
Abstract
Background Emerging data indicate that prenatal exposure to air pollution may lead to higher susceptibility to several non-communicable diseases. Limited research has been conducted due to difficulties in modelling realistic air pollution exposure. In this study, pregnant mice were exposed from gestational day 10–17 to an atmosphere representative of a 2017 pollution event in Beijing, China. Intestinal homeostasis and microbiota were assessed in both male and female offspring during the suckling-to-weaning transition. Results Sex-specific differences were observed in progeny of gestationally-exposed mice. In utero exposed males exhibited decreased villus and crypt length, vacuolation abnormalities, and lower levels of tight junction protein ZO-1 in ileum. They showed an upregulation of absorptive cell markers and a downregulation of neonatal markers in colon. Cecum of in utero exposed male mice also presented a deeply unbalanced inflammatory pattern. By contrast, in utero exposed female mice displayed less severe intestinal alterations, but included dysregulated expression of Lgr5 in colon, Tjp1 in cecum, and Epcam, Car2 and Sis in ileum. Moreover, exposed female mice showed dysbiosis characterized by a decreased weighted UniFrac β-diversity index, a higher abundance of Bacteroidales and Coriobacteriales orders, and a reduced Firmicutes/Bacteroidetes ratio. Conclusion Prenatal realistic modelling of an urban air pollution event induced sex-specific precocious alterations of structural and immune intestinal development in mice. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-022-00481-y.
Collapse
Affiliation(s)
- Eva Guilloteau
- Univ. Lille, INSERM, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Patrice Coll
- Université Paris Cité and Univ Paris Est Créteil, CNRS, LISA, 75013, Paris, France
| | - Zhuyi Lu
- Univ. Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
| | - Madjid Djouina
- Univ. Lille, INSERM, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Mathieu Cazaunau
- Univ. Paris Est Créteil and Université Paris Cité, CNRS, LISA, 94010, Créteil, France
| | - Christophe Waxin
- Univ. Lille, INSERM, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Antonin Bergé
- Université Paris Cité and Univ Paris Est Créteil, CNRS, LISA, 75013, Paris, France
| | - Ségolène Caboche
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, UMR2014-US41-PLBS-Plateformes Lilloises de Biologie & Santé, 59000, Lille, France
| | - Aline Gratien
- Université Paris Cité and Univ Paris Est Créteil, CNRS, LISA, 75013, Paris, France
| | - Elie Al Marj
- Université Paris Cité and Univ Paris Est Créteil, CNRS, LISA, 75013, Paris, France
| | - David Hot
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, UMR2014-US41-PLBS-Plateformes Lilloises de Biologie & Santé, 59000, Lille, France
| | - Laurent Dubuquoy
- Univ. Lille, INSERM, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - David Launay
- Univ. Lille, INSERM, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Cécile Vignal
- Univ. Lille, INSERM, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Sophie Lanone
- Univ. Paris Est Créteil, INSERM, IMRB, 94010, Créteil, France
| | - Mathilde Body-Malapel
- Univ. Lille, INSERM, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France.
| |
Collapse
|
33
|
Olude MA, Mouihate A, Mustapha OA, Farina C, Quintana FJ, Olopade JO. Astrocytes and Microglia in Stress-Induced Neuroinflammation: The African Perspective. Front Immunol 2022; 13:795089. [PMID: 35707531 PMCID: PMC9190229 DOI: 10.3389/fimmu.2022.795089] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Africa is laden with a youthful population, vast mineral resources and rich fauna. However, decades of unfortunate historical, sociocultural and leadership challenges make the continent a hotspot for poverty, indoor and outdoor pollutants with attendant stress factors such as violence, malnutrition, infectious outbreaks and psychological perturbations. The burden of these stressors initiate neuroinflammatory responses but the pattern and mechanisms of glial activation in these scenarios are yet to be properly elucidated. Africa is therefore most vulnerable to neurological stressors when placed against a backdrop of demographics that favor explosive childbearing, a vast population of unemployed youths making up a projected 42% of global youth population by 2030, repressive sociocultural policies towards women, poor access to healthcare, malnutrition, rapid urbanization, climate change and pollution. Early life stress, whether physical or psychological, induces neuroinflammatory response in developing nervous system and consequently leads to the emergence of mental health problems during adulthood. Brain inflammatory response is driven largely by inflammatory mediators released by glial cells; namely astrocytes and microglia. These inflammatory mediators alter the developmental trajectory of fetal and neonatal brain and results in long-lasting maladaptive behaviors and cognitive deficits. This review seeks to highlight the patterns and mechanisms of stressors such as poverty, developmental stress, environmental pollutions as well as malnutrition stress on astrocytes and microglia in neuroinflammation within the African context.
Collapse
Affiliation(s)
- Matthew Ayokunle Olude
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
- *Correspondence: Matthew Ayokunle Olude,
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Kuwait City, Kuwait
| | - Oluwaseun Ahmed Mustapha
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - Cinthia Farina
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) San Raffaele Scientific Institute, Institute of Experimental Neurology (INSPE) and Division of Neuroscience, Milan, Italy
| | - Francisco Javier Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - James Olukayode Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
34
|
Yu X, Rahman MM, Wang Z, Carter SA, Schwartz J, Chen Z, Eckel SP, Hackman D, Chen JC, Xiang AH, McConnell R. Evidence of susceptibility to autism risks associated with early life ambient air pollution: A systematic review. ENVIRONMENTAL RESEARCH 2022; 208:112590. [PMID: 34929192 PMCID: PMC11409923 DOI: 10.1016/j.envres.2021.112590] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Many studies have found associations between early life air pollution exposure and subsequent onset of autism spectrum disorder (ASD). However, characteristics that affect susceptibility remain unclear. OBJECTIVE This systematic review examined epidemiologic studies on the modifying roles of social, child, genetic and maternal characteristics in associations between prenatal and early postnatal air pollution exposure and ASD. METHODS A systematic literature search in PubMed and Embase was conducted. Studies that examined modifiers of the association between air pollution and ASD were included. RESULTS A total of 19 publications examined modifiers of the associations between early life air pollution exposures and ASD. In general, estimates of effects on risk of ASD in boys were larger than in girls (based on 11 studies). Results from studies of effects of family education (2 studies) and neighborhood deprivation (2 studies) on air pollution-ASD associations were inconsistent. Limited data (1 study) suggest pregnant women with insufficient folic acid intake might be more susceptible to ambient particulate matter less than 2.5 μm (PM2.5) and 10 μm (PM10) in aerodynamic diameter, and to nitrogen dioxide (NO2). Children of mothers with gestational diabetes had increased risk of ozone-associated ASD (1 study). Two genetic studies reported that copy number variations may amplify the effect of ozone, and MET rs1858830 CC genotype may augment effects of PM and near-roadway pollutants on ASD. CONCLUSIONS Child's sex, maternal nutrition or diabetes, socioeconomic factors, and child risk genotypes were reported to modify the effect of early-life air pollutants on ASD risk in the epidemiologic literature. However, the sparsity of studies on comparable modifying hypotheses precludes conclusive findings. Further research is needed to identify susceptible populations and potential targets for preventive intervention.
Collapse
Affiliation(s)
- Xin Yu
- Spatial Science Institute, University of Southern California, Los Angeles, CA, USA
| | - Md Mostafijur Rahman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zhongying Wang
- Spatial Science Institute, University of Southern California, Los Angeles, CA, USA
| | - Sarah A Carter
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zhanghua Chen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sandrah P Eckel
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel Hackman
- USC Suzanne Dworak-Peck School of Social Work, University of Southern California, Los Angeles, CA, USA
| | - Jiu-Chiuan Chen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anny H Xiang
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Rob McConnell
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Miller JG, Dennis EL, Heft-Neal S, Jo B, Gotlib IH. Fine Particulate Air Pollution, Early Life Stress, and Their Interactive Effects on Adolescent Structural Brain Development: A Longitudinal Tensor-Based Morphometry Study. Cereb Cortex 2022; 32:2156-2169. [PMID: 34607342 PMCID: PMC9113318 DOI: 10.1093/cercor/bhab346] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/18/2021] [Accepted: 08/22/2021] [Indexed: 11/13/2022] Open
Abstract
Air pollution is a major environmental threat to public health; we know little, however, about its effects on adolescent brain development. Exposure to air pollution co-occurs, and may interact, with social factors that also affect brain development, such as early life stress (ELS). Here, we show that severity of ELS and fine particulate air pollution (PM2.5) are associated with volumetric changes in distinct brain regions, but also uncover regions in which ELS moderates the effects of PM2.5. We interviewed adolescents about ELS events, used satellite-derived estimates of ambient PM2.5 concentrations, and conducted longitudinal tensor-based morphometry to assess regional changes in brain volume over an approximately 2-year period (N = 115, ages 9-13 years at Time 1). For adolescents who had experienced less severe ELS, PM2.5 was associated with volumetric changes across several gray and white matter regions. Fewer effects of PM2.5 were observed for adolescents who had experienced more severe ELS, although occasionally they were in the opposite direction. This pattern of results suggests that for many brain regions, moderate to severe ELS largely constrains the effects of PM2.5 on structural development. Further theory and research is needed on the joint effects of ELS and air pollution on the brain.
Collapse
Affiliation(s)
- Jonas G Miller
- Department of Psychology, Stanford University, Stanford, CA 94305, USA
| | - Emily L Dennis
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Sam Heft-Neal
- Center for Food Security and the Environment, Stanford University, Stanford, CA 94305, USA
| | - Booil Jo
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Ian H Gotlib
- Department of Psychology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
36
|
Westacott LJ, Wilkinson LS. Complement Dependent Synaptic Reorganisation During Critical Periods of Brain Development and Risk for Psychiatric Disorder. Front Neurosci 2022; 16:840266. [PMID: 35600620 PMCID: PMC9120629 DOI: 10.3389/fnins.2022.840266] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
We now know that the immune system plays a major role in the complex processes underlying brain development throughout the lifespan, carrying out a number of important homeostatic functions under physiological conditions in the absence of pathological inflammation or infection. In particular, complement-mediated synaptic pruning during critical periods of early life may play a key role in shaping brain development and subsequent risk for psychopathology, including neurodevelopmental disorders such as schizophrenia and autism spectrum disorders. However, these disorders vary greatly in their onset, disease course, and prevalence amongst sexes suggesting complex interactions between the immune system, sex and the unique developmental trajectories of circuitries underlying different brain functions which are yet to be fully understood. Perturbations of homeostatic neuroimmune interactions during different critical periods in which regional circuits mature may have a plethora of long-term consequences for psychiatric phenotypes, but at present there is a gap in our understanding of how these mechanisms may impact on the structural and functional changes occurring in the brain at different developmental stages. In this article we will consider the latest developments in the field of complement mediated synaptic pruning where our understanding is beginning to move beyond the visual system where this process was first described, to brain areas and developmental periods of potential relevance to psychiatric disorders.
Collapse
Affiliation(s)
- Laura J. Westacott
- Neuroscience and Mental Health Innovation Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Lawrence S. Wilkinson
- Neuroscience and Mental Health Innovation Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
37
|
Morris RH, Chabrier G, Counsell SJ, McGonnell IM, Thornton C. Differential effects of Urban Particulate Matter on BV2 microglial-like and C17.2 neural stem/precursor cells. Dev Neurosci 2022; 44:309-319. [PMID: 35500557 DOI: 10.1159/000524829] [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: 10/28/2021] [Accepted: 03/09/2022] [Indexed: 11/19/2022] Open
Abstract
Air pollution affects the majority of the world's population and has been linked to over 7 million premature deaths per year. Exposure to particulate matter (PM) contained within air pollution is associated with cardiovascular, respiratory and neurological ill health. There is increasing evidence that exposure to air pollution in utero and in early childhood is associated with altered brain development. However, the underlying mechanisms for impaired brain development are not clear. While oxidative stress and neuroinflammation are documented consequences of PM exposure, cell-specific mechanisms that may be triggered in response to air pollution exposure are less well defined. Here we assess the effect of urban (U)PM exposure on two different cell types, microglial-like BV2 cells and neural stem / precursor-like C17.2 cells. We found that, contrary to expectations, immature C17.2 cells were more resistant to PM-mediated oxidative stress and cell death than BV2 cells. PM exposure resulted in decreased mitochondrial health and increased mitochondrial ROS in BV2 cells which could be prevented by mitoTEMPO antioxidant treatment. Our data suggest that not only is mitochondrial dysfunction a key trigger in PM-mediated cytotoxicity, but that such deleterious effects may also depend on cell type and maturity.
Collapse
Affiliation(s)
- Rebecca H Morris
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, United Kingdom
| | - Gwladys Chabrier
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Serena J Counsell
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, United Kingdom
| | - Imelda M McGonnell
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Claire Thornton
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, United Kingdom
| |
Collapse
|
38
|
Elgueta D, Murgas P, Riquelme E, Yang G, Cancino GI. Consequences of Viral Infection and Cytokine Production During Pregnancy on Brain Development in Offspring. Front Immunol 2022; 13:816619. [PMID: 35464419 PMCID: PMC9021386 DOI: 10.3389/fimmu.2022.816619] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Infections during pregnancy can seriously damage fetal neurodevelopment by aberrantly activating the maternal immune system, directly impacting fetal neural cells. Increasing evidence suggests that these adverse impacts involve alterations in neural stem cell biology with long-term consequences for offspring, including neurodevelopmental disorders such as autism spectrum disorder, schizophrenia, and cognitive impairment. Here we review how maternal infection with viruses such as Influenza A, Cytomegalovirus, and Zika during pregnancy can affect the brain development of offspring by promoting the release of maternal pro-inflammatory cytokines, triggering neuroinflammation of the fetal brain, and/or directly infecting fetal neural cells. In addition, we review insights into how these infections impact human brain development from studies with animal models and brain organoids. Finally, we discuss how maternal infection with SARS-CoV-2 may have consequences for neurodevelopment of the offspring.
Collapse
Affiliation(s)
- Daniela Elgueta
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Paola Murgas
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Tecnología Médica, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Erick Riquelme
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Tecnología Médica, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Guang Yang
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, Calgary, AB, Canada
| | - Gonzalo I Cancino
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| |
Collapse
|
39
|
Wang F, Cai YJ, Ma X, Wang N, Wu ZB, Sun Y, Xu YX, Yang H, Liu TT, Xia Q, Yu Z, Zhu DF. Synaptic loss in a mouse model of euthyroid Hashimoto's thyroiditis: possible involvement of the microglia. BMC Neurosci 2022; 23:25. [PMID: 35468730 PMCID: PMC9036731 DOI: 10.1186/s12868-022-00710-2] [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: 08/13/2021] [Accepted: 04/05/2022] [Indexed: 11/24/2022] Open
Abstract
Background Hashimoto’s thyroiditis (HT) is an autoimmune illness that renders individuals vulnerable to neuropsychopathology even in the euthyroid state, the mechanisms involved remain unclear. We hypothesized that activated microglia might disrupt synapses, resulting in cognitive disturbance in the context of euthyroid HT, and designed the present study to test this hypothesis. Methods Experimental HT model was induced by immunizing NOD mice with thyroglobulin and adjuvant twice. Morris Water Maze was measured to determine mice spatial learning and memory. The synaptic parameters such as the synaptic density, synaptic ultrastructure and synaptic-markers (SYN and PSD95) as well as the interactions of microglia with synapses were also determined. Results HT mice had poorer performance in Morris Water Maze than controls. Concurrently, HT resulted in a significant reduction in synapse density and ultrastructure damage, along with decreased synaptic puncta visualized by immunostaining with synaptophysin and PSD-95. In parallel, frontal activated microglia in euthyroid HT mice showed increased engulfment of PSD95 and EM revealed that the synaptic structures were visible within the microglia. These functional alterations in microglia corresponded to structural increases in their attachment to neuronal perikarya and a reduction in presynaptic terminals covering the neurons. Conclusion Our results provide initial evidence that HT can induce synaptic loss in the euthyroid state with deficits might be attributable to activated microglia, which may underlie the deleterious effects of HT on spatial learning and memory. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-022-00710-2.
Collapse
Affiliation(s)
- Fen Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yao-Jun Cai
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiao Ma
- Department of Respiratoration, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, 241000, China
| | - Nan Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zhang-Bi Wu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yan Sun
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yong-Xia Xu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Hao Yang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Tian-Tian Liu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Qin Xia
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zhen Yu
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - De-Fa Zhu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
40
|
Shin J, Park H, Kim HS, Kim EJ, Kim KN, Hong YC, Ha M, Kim Y, Ha E. Pre- and postnatal exposure to multiple ambient air pollutants and child behavioral problems at five years of age. ENVIRONMENTAL RESEARCH 2022; 206:112526. [PMID: 34921822 DOI: 10.1016/j.envres.2021.112526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/30/2021] [Accepted: 12/04/2021] [Indexed: 06/14/2023]
Abstract
Ambient air pollution is emerging as a risk factor for adverse neurological symptoms and early childhood diseases. This study aimed to evaluate the association between pre- and postnatal exposure to air pollutants and childhood behavior by using MOCEH prospective birth cohort data. In total, 353 mother-child pairs at birth, who completed child behavioral assessments using the Korean version of the Child Behavior Checklist at five years of age, were included in the study. Multivariate linear regression (MLR) for single pollutant and Bayesian kernel machine regression (BKMR) for multiple pollutants were conducted. MLR analysis showed that air pollutant exposures during the first trimester were significantly associated with the internalizing problems score after adjusting for covariates. The estimates were 0.19 (0.05-0.32) per 1 μg/m3 increase in PM2.5, 0.13 (0.04-0.22) per 1 μg/m3 increase in PM10, and 0.20 (0.02-0.37) per 1 ppb increase in NO2. The BKMR model analysis revealed that the overall effects of multiple air pollutants during the first trimester of pregnancy and 0-6 months of the infantile period were significantly associated with behavioral problems. Boys showed a stronger associations than girls. Taken together, these results showed that the first trimester of pregnancy and 0-6 months of the infantile period were important for air pollutant exposure because exposure at these periods was associated with behavioral problems in 5-year-old children. Future efforts are required to control air pollution levels and reduce the health burden of vulnerable populations, including pregnant women and children.
Collapse
Affiliation(s)
- Jiyoung Shin
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Environmental Medicine, Graduate Program in System Health Science and Engineering, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Hyesook Park
- Department of Preventive Medicine, Graduate Program in System Health Science and Engineering, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Hae Soon Kim
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eui-Jung Kim
- Department of Psychiatry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Kyoung-Nam Kim
- Department of Preventive Medicine and Public Health, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yun-Chul Hong
- Department of Human Systems Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Mina Ha
- Department of Preventive Medicine, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Yangho Kim
- Department of Occupational and Environmental Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Eunhee Ha
- Department of Environmental Medicine, Graduate Program in System Health Science and Engineering, College of Medicine, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
41
|
Dash S, Syed YA, Khan MR. Understanding the Role of the Gut Microbiome in Brain Development and Its Association With Neurodevelopmental Psychiatric Disorders. Front Cell Dev Biol 2022; 10:880544. [PMID: 35493075 PMCID: PMC9048050 DOI: 10.3389/fcell.2022.880544] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome has a tremendous influence on human physiology, including the nervous system. During fetal development, the initial colonization of the microbiome coincides with the development of the nervous system in a timely, coordinated manner. Emerging studies suggest an active involvement of the microbiome and its metabolic by-products in regulating early brain development. However, any disruption during this early developmental process can negatively impact brain functionality, leading to a range of neurodevelopment and neuropsychiatric disorders (NPD). In this review, we summarize recent evidence as to how the gut microbiome can influence the process of early human brain development and its association with major neurodevelopmental psychiatric disorders such as autism spectrum disorders, attention-deficit hyperactivity disorder, and schizophrenia. Further, we discuss how gut microbiome alterations can also play a role in inducing drug resistance in the affected individuals. We propose a model that establishes a direct link of microbiome dysbiosis with the exacerbated inflammatory state, leading to functional brain deficits associated with NPD. Based on the existing research, we discuss a framework whereby early diet intervention can boost mental wellness in the affected subjects and call for further research for a better understanding of mechanisms that govern the gut-brain axis may lead to novel approaches to the study of the pathophysiology and treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Somarani Dash
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Yasir Ahmed Syed
- School of Biosciences and Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building, Cardiff, United Kingdom
| | - Mojibur R. Khan
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, India
- *Correspondence: Mojibur R. Khan,
| |
Collapse
|
42
|
Lowery R, Latchney S, Peer R, Lamantia C, Lordy K, Opanashuk L, McCall M, Majewska A. Gestational and lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin primes cortical microglia to tissue injury. Brain Behav Immun 2022; 101:288-303. [PMID: 35065196 PMCID: PMC9007156 DOI: 10.1016/j.bbi.2022.01.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/22/2021] [Accepted: 01/16/2022] [Indexed: 11/16/2022] Open
Abstract
Recent studies have shown that the aryl hydrocarbon receptor (AhR) is expressed in the brain's native immune cells, known as microglia. However, while the impact of exposure to AhR ligands is well studied in the peripheral immune system, the impact of such exposure on immune function in the brain is less well defined. Microglia serve dual roles in providing synaptic and immunological support for neighboring neurons and in mediating responses to environmental stimuli, including exposure to environmental chemicals. Because of their dual roles in regulating physiological and pathological processes, cortical microglia are well positioned to translate toxic stimuli into defects in cortical function via aberrant synaptic and immunological functioning, mediated either through direct microglial AhR activation or in response to AhR activation in neighboring cells. Here, we use gene expression studies, histology, and two-photon in vivo imaging to investigate how developmental exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a high-affinity and persistent AhR agonist, modulates microglial characteristics and function in the intact brain. Whole cortical RT-qPCR analysis and RNA-sequencing of isolated microglia revealed that gestational and lactational TCDD exposure produced subtle, but durable, changes in microglia transcripts. Histological examination and two-photon in vivo imaging revealed that while microglia density, distribution, morphology, and motility were unaffected by TCDD exposure, exposure resulted in microglia that responded more robustly to focal tissue injury. However, this effect was rectified with depletion and repopulation of microglia. These results suggest that gestational and lactational exposure to AhR ligands can result in long-term priming of microglia to produce heightened responses towards tissue injury which can be restored to normal function through microglial repopulation.
Collapse
Affiliation(s)
- R.L. Lowery
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - S.E. Latchney
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - R.P. Peer
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - C.E. Lamantia
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - K.A. Lordy
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | | | - M. McCall
- Department of Biostatistics and Computational Biology, University of Rochester, NY 14642,Department of Biomedical Genetics, University of Rochester, NY 14642
| | - A.K Majewska
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642,Corresponding Author: Ania K. Majewska, University of Rochester, School of Medicine and Dentistry, Department of Neuroscience, Center for Visual Science, 601 Elmwood Avenue, Box 603, Rochester, New York 14642, , Phone: (585) 276-2254
| |
Collapse
|
43
|
Gzielo K, Nikiforuk A. Astroglia in Autism Spectrum Disorder. Int J Mol Sci 2021; 22:11544. [PMID: 34768975 PMCID: PMC8583956 DOI: 10.3390/ijms222111544] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/13/2021] [Accepted: 10/21/2021] [Indexed: 01/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is an umbrella term encompassing several neurodevelopmental disorders such as Asperger syndrome or autism. It is characterised by the occurrence of distinct deficits in social behaviour and communication and repetitive patterns of behaviour. The symptoms may be of different intensity and may vary in types. Risk factors for ASD include disturbed brain homeostasis, genetic predispositions, or inflammation during the prenatal period caused by viruses or bacteria. The number of diagnosed cases is growing, but the main cause and mechanism leading to ASD is still uncertain. Recent findings from animal models and human cases highlight the contribution of glia to the ASD pathophysiology. It is known that glia cells are not only "gluing" neurons together but are key players participating in different processes crucial for proper brain functioning, including neurogenesis, synaptogenesis, inflammation, myelination, proper glutamate processing and many others. Despite the prerequisites for the involvement of glia in the processes related to the onset of autism, there are far too little data regarding the engagement of these cells in the development of ASD.
Collapse
Affiliation(s)
- Kinga Gzielo
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Behavioral Neuroscience and Drug Development, 12 Smętna Street, 31-343 Kraków, Poland;
| | | |
Collapse
|
44
|
Patisaul HB. REPRODUCTIVE TOXICOLOGY: Endocrine disruption and reproductive disorders: impacts on sexually dimorphic neuroendocrine pathways. Reproduction 2021; 162:F111-F130. [PMID: 33929341 PMCID: PMC8484365 DOI: 10.1530/rep-20-0596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/30/2021] [Indexed: 11/08/2022]
Abstract
We are all living with hundreds of anthropogenic chemicals in our bodies every day, a situation that threatens the reproductive health of present and future generations. This review focuses on endocrine-disrupting compounds (EDCs), both naturally occurring and man-made, and summarizes how they interfere with the neuroendocrine system to adversely impact pregnancy outcomes, semen quality, age at puberty, and other aspects of human reproductive health. While obvious malformations of the genitals and other reproductive organs are a clear sign of adverse reproductive health outcomes and injury to brain sexual differentiation, the hypothalamic-pituitary-gonadal (HPG) axis can be much more difficult to discern, particularly in humans. It is well-established that, over the course of development, gonadal hormones shape the vertebrate brain such that sex-specific reproductive physiology and behaviors emerge. Decades of work in neuroendocrinology have elucidated many of the discrete and often very short developmental windows across pre- and postnatal development in which this occurs. This has allowed toxicologists to probe how EDC exposures in these critical windows can permanently alter the structure and function of the HPG axis. This review includes a discussion of key EDC principles including how latency between exposure and the emergence of consequential health effects can be long, along with a summary of the most common and less well-understood EDC modes of action. Extensive examples of how EDCs are impacting human reproductive health, and evidence that they have the potential for multi-generational physiological and behavioral effects are also provided.
Collapse
Affiliation(s)
- Heather B Patisaul
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
45
|
Lowery RL, Cealie MY, Lamantia CE, Mendes MS, Drew PD, Majewska AK. Microglia and astrocytes show limited, acute alterations in morphology and protein expression following a single developmental alcohol exposure. J Neurosci Res 2021; 99:2008-2025. [PMID: 33606320 PMCID: PMC8349862 DOI: 10.1002/jnr.24808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) are the most common cause of nonheritable, preventable mental disability and are characterized by cognitive, behavioral, and physical impairments. FASD occurs in almost 5% of births in the United States, but despite this prevalence there is no known cure, largely because the biological mechanisms that translate alcohol exposure to neuropathology are not well understood. While the effects of early ethanol exposure on neuronal survival and circuitry have received more attention, glia, the cells most closely tied to initiating and propagating inflammatory events, could be an important target for alcohol in the developing brain. Inflammation is known to alter developmental trajectories, but it has recently been shown that even small changes in both astrocytes and microglia in the absence of full-blown inflammatory signaling can alter brain function long-term. Here, we studied the acute response of astrocytes and microglia to a single exposure to ethanol in development across sexes in a mouse model of human third trimester exposure, in order to understand how these cells may transition from their normal developmental path to a different program that leads to FASD neuropathology. We found that although a single ethanol exposure delivered subcutaneously on postnatal day 4 did not cause large changes in microglial morphology or the expression of AldH1L1 and GFAP in the cortex and hippocampus, subtle effects were observed. These findings suggest that even a single, early ethanol exposure can induce mild acute alterations in glia that could contribute to developmental deficits.
Collapse
Affiliation(s)
- Rebecca L Lowery
- Department of Neuroscience, School of Medicine and Dentistry, Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - MaKenna Y Cealie
- Department of Neuroscience, School of Medicine and Dentistry, Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Cassandra E Lamantia
- Department of Neuroscience, School of Medicine and Dentistry, Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Monique S Mendes
- Department of Neuroscience, School of Medicine and Dentistry, Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Paul D Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ania K Majewska
- Department of Neuroscience, School of Medicine and Dentistry, Center for Visual Science, University of Rochester, Rochester, NY, USA
| |
Collapse
|
46
|
Song Y, Southam K, Bennett E, Johnston F, Foa L, Wheeler AJ, Zosky GR. Adverse effects of prenatal exposure to residential dust on post-natal brain development. ENVIRONMENTAL RESEARCH 2021; 198:110489. [PMID: 33220241 DOI: 10.1016/j.envres.2020.110489] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Previous studies have shown an association between prenatal exposure to particulate matter (PM) and adverse brain development. However, it is unclear whether gestational exposure to community-sampled residential PM has an impact on the developing brain. OBJECTIVES We aimed to test whether in utero exposure to PM from residential roof spaces (ceiling voids) alters critical foetal neurodevelopmental processes. METHODS Pregnant C57BL/6 mice were intranasally exposed to 100 μg of roof space particles (~5 mg kg-1) in 50 μl of saline, or saline alone under light methoxyflurane anaesthesia, throughout mid-to-late gestation. At 2 weeks post-natal age, pups were sacrificed and assessed for body and brain growth. The brain tissue was collected and examined for a range of neurodevelopmental markers for synaptogenesis, synaptic plasticity, gliogenic events and myelination by immunohistochemistry. RESULTS Gestational exposure to roof space PM reduced post-natal body and brain weights. There was no significant effect of roof space PM exposure on synaptogenesis, synaptic plasticity or astrocyte density. However, PM exposure caused increased myelin load in the white matter and elevated microglial density which was dependent on the PM sample. These effects were found to be consistent between male and female mice. CONCLUSIONS Our data suggest that exposure to residential roof space PM during pregnancy impairs somatic growth and causes neuropathological changes in the developing brain.
Collapse
Affiliation(s)
- Yong Song
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, 7000, Australia.
| | - Katherine Southam
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, 7000, Australia.
| | - Ellen Bennett
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, 7000, Australia.
| | - Fay Johnston
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, 7000, Australia.
| | - Lisa Foa
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, 7000, Australia.
| | - Amanda J Wheeler
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, 7000, Australia; Mary Mackillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, 3350, Australia.
| | - Graeme R Zosky
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, 7000, Australia; Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, 7000, Australia.
| |
Collapse
|
47
|
Patisaul HB. Endocrine disrupting chemicals (EDCs) and the neuroendocrine system: Beyond estrogen, androgen, and thyroid. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 92:101-150. [PMID: 34452685 DOI: 10.1016/bs.apha.2021.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hundreds of anthropogenic chemicals occupy our bodies, a situation that threatens the health of present and future generations. This chapter focuses on endocrine disrupting compounds (EDCs), both naturally occurring and man-made, that affect the neuroendocrine system to adversely impact health, with an emphasis on reproductive and metabolic pathways. The neuroendocrine system is highly sexually dimorphic and essential for maintaining homeostasis and appropriately responding to the environment. Comprising both neural and endocrine components, the neuroendocrine system is hormone sensitive throughout life and touches every organ system in the body. The integrative nature of the neuroendocrine system means that EDCs can have multi-system effects. Additionally, because gonadal hormones are essential for the sex-specific organization of numerous neuroendocrine pathways, endocrine disruption of this programming can lead to permanent deficits. Included in this review is a brief history of the neuroendocrine disruption field and a thorough discussion of the most common and less well understood neuroendocrine disruption modes of action. Also provided are extensive examples of how EDCs are likely contributing to neuroendocrine disorders such as obesity, and evidence that they have the potential for multi-generational effects.
Collapse
Affiliation(s)
- Heather B Patisaul
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States.
| |
Collapse
|
48
|
Stoessel MB, Majewska AK. Little cells of the little brain: microglia in cerebellar development and function. Trends Neurosci 2021; 44:564-578. [PMID: 33933255 PMCID: PMC8222145 DOI: 10.1016/j.tins.2021.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 02/23/2021] [Accepted: 04/05/2021] [Indexed: 12/31/2022]
Abstract
Microglia are long-lived resident macrophages of the brain with diverse roles that span development, adulthood, and aging. Once thought to be a relatively homogeneous population, there is a growing recognition that microglia are highly specialized to suit their specific brain region. Cerebellar microglia represent an example of such specialization, exhibiting a dynamical, transcriptional, and immunological profile that differs from that of other microglial populations. Here we review the evidence that cerebellar microglia shape the cerebellar environment and are in turn shaped by it. We examine the roles microglia play in cerebellar function, development, and aging. The emerging findings on cerebellar microglia may also provide insights into disease processes involving cerebellar dysfunction.
Collapse
Affiliation(s)
- Mark B Stoessel
- Department of Neuroscience, University of Rochester, Rochester, NY 14642, USA; Neuroscience Graduate Program, University of Rochester, Rochester, NY 14642, USA
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
49
|
Zengeler KE, Lukens JR. Innate immunity at the crossroads of healthy brain maturation and neurodevelopmental disorders. Nat Rev Immunol 2021; 21:454-468. [PMID: 33479477 PMCID: PMC9213174 DOI: 10.1038/s41577-020-00487-7] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/29/2022]
Abstract
The immune and nervous systems have unique developmental trajectories that individually build intricate networks of cells with highly specialized functions. These two systems have extensive mechanistic overlap and frequently coordinate to accomplish the proper growth and maturation of an organism. Brain resident innate immune cells - microglia - have the capacity to sculpt neural circuitry and coordinate copious and diverse neurodevelopmental processes. Moreover, many immune cells and immune-related signalling molecules are found in the developing nervous system and contribute to healthy neurodevelopment. In particular, many components of the innate immune system, including Toll-like receptors, cytokines, inflammasomes and phagocytic signals, are critical contributors to healthy brain development. Accordingly, dysfunction in innate immune signalling pathways has been functionally linked to many neurodevelopmental disorders, including autism and schizophrenia. This review discusses the essential roles of microglia and innate immune signalling in the assembly and maintenance of a properly functioning nervous system.
Collapse
Affiliation(s)
- Kristine E Zengeler
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), Charlottesville, VA, USA.
- Neuroscience Graduate Program, Charlottesville, VA, USA.
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - John R Lukens
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), Charlottesville, VA, USA.
- Neuroscience Graduate Program, Charlottesville, VA, USA.
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
50
|
Bernal-Meléndez E, Callebert J, Bouillaud P, Persuy MA, Olivier B, Badonnel K, Chavatte-Palmer P, Baly C, Schroeder H. Dopaminergic and serotonergic changes in rabbit fetal brain upon repeated gestational exposure to diesel engine exhaust. Arch Toxicol 2021; 95:3085-3099. [PMID: 34189592 DOI: 10.1007/s00204-021-03110-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/17/2021] [Indexed: 11/29/2022]
Abstract
Limited studies in humans and in animal models have investigated the neurotoxic risks related to a gestational exposure to diesel exhaust particles (DEP) on the embryonic brain, especially those regarding monoaminergic systems linked to neurocognitive disorders. We previously showed that exposure to DEP alters monoaminergic neurotransmission in fetal olfactory bulbs and modifies tissue morphology along with behavioral consequences at birth in a rabbit model. Given the anatomical and functional connections between olfactory and central brain structures, we further characterized their impacts in brain regions associated with monoaminergic neurotransmission. At gestational day 28 (GD28), fetal rabbit brains were collected from dams exposed by nose-only to either a clean air or filtered DEP for 2 h/day, 5 days/week, from GD3 to GD27. HPLC dosage and histochemical analyses of the main monoaminergic systems, i.e., dopamine (DA), noradrenaline (NA), and serotonin (5-HT) and their metabolites were conducted in microdissected fetal brain regions. DEP exposure increased the level of DA and decreased the dopaminergic metabolites ratios in the prefrontal cortex (PFC), together with sex-specific alterations in the hippocampus (Hp). In addition, HVA level was increased in the temporal cortex (TCx). Serotonin and 5-HIAA levels were decreased in the fetal Hp. However, DEP exposure did not significantly modify NA levels, tyrosine hydroxylase, tryptophan hydroxylase or AChE enzymatic activity in fetal brain. Exposure to DEP during fetal life results in dopaminergic and serotonergic changes in critical brain regions that might lead to detrimental potential short-term neural disturbances as precursors of long-term neurocognitive consequences.
Collapse
Affiliation(s)
- Estefania Bernal-Meléndez
- NeuroBiologie de l'Olfaction, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,CALBINOTOX, EA7488, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Jacques Callebert
- Service de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, Paris, France
| | | | - Marie-Annick Persuy
- NeuroBiologie de l'Olfaction, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,Université Paris-Saclay, UVSQ, INRAE, INRAE, BREED UR1198, Bat. 230, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Benoit Olivier
- CALBINOTOX, EA7488, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Karine Badonnel
- NeuroBiologie de l'Olfaction, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,Université Paris-Saclay, UVSQ, INRAE, INRAE, BREED UR1198, Bat. 230, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, INRAE, BREED UR1198, Bat. 230, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Christine Baly
- NeuroBiologie de l'Olfaction, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France. .,Université Paris-Saclay, UVSQ, INRAE, INRAE, BREED UR1198, Bat. 230, Domaine de Vilvert, 78350, Jouy-en-Josas, France.
| | - Henri Schroeder
- CALBINOTOX, EA7488, Université de Lorraine, Vandœuvre-lès-Nancy, France.
| |
Collapse
|