Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 115: 617-630, 2024 01.
Article in English | MEDLINE | ID: mdl-37967662

ABSTRACT

Birth is an inflammatory event for the newborn, characterized by elevations in interleukin (IL)-6, IL-10, and tumor necrosis factor (TNF)-α peripherally and/or centrally, as well as changes in brain microglia. However, the mechanism(s) underlying these responses is unknown. Toll-like receptors (TLRs) play crucial roles in innate immunity and initiate inflammatory cascades upon recognition of endogenous or exogenous antigens. Most TLR signaling depends on the adaptor molecule myeloid differentiation primary response 88 (MyD88). We independently varied MyD88 gene status in mouse dams and their offspring to determine whether the inflammatory response to birth depends on MyD88 signaling and, if so, whether that signaling occurs in the offspring, the mother, or both. We find that the perinatal surges in plasma IL-6 and brain expression of TNF-α depend solely on MyD88 gene status of the offspring, whereas postnatal increases in plasma IL-10 and TNF-α depend on MyD88 in both the pup and dam. Interestingly, MyD88 genotype of the dam primarily drives differences in offspring brain microglial density and has robust effects on developmental neuronal cell death. Milk cytokines were evaluated as a possible source of postnatal maternal influence; although we found high levels of CXCL1/GROα and several other cytokines in ingested post-partum milk, their presence did not require MyD88. Thus, the inflammatory response previously described in the late-term fetus and newborn depends on MyD88 (and, by extension, TLRs), with signaling in both the dam and offspring contributing. Unexpectedly, naturally-occuring neuronal cell death in the newborn is modulated primarily by maternal MyD88 gene status.


Subject(s)
Interleukin-10 , Myeloid Differentiation Factor 88 , Animals , Female , Mice , Pregnancy , Adaptor Proteins, Signal Transducing/metabolism , Cytokines/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Mice, Inbred C57BL , Mothers , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Front Neurosci ; 17: 1130347, 2023.
Article in English | MEDLINE | ID: mdl-37207179

ABSTRACT

At birth, mammals experience a massive colonization by microorganisms. We previously reported that newborn mice gestated and born germ-free (GF) have increased microglial labeling and alterations in developmental neuronal cell death in the hippocampus and hypothalamus, as well as greater forebrain volume and body weight when compared to conventionally colonized (CC) mice. To test whether these effects are solely due to differences in postnatal microbial exposure, or instead may be programmed in utero, we cross-fostered GF newborns immediately after birth to CC dams (GF→CC) and compared them to offspring fostered within the same microbiota status (CC→CC, GF→GF). Because key developmental events (including microglial colonization and neuronal cell death) shape the brain during the first postnatal week, we collected brains on postnatal day (P) 7. To track gut bacterial colonization, colonic content was also collected and subjected to 16S rRNA qPCR and Illumina sequencing. In the brains of GF→GF mice, we replicated most of the effects seen previously in GF mice. Interestingly, the GF brain phenotype persisted in GF→CC offspring for almost all measures. In contrast, total bacterial load did not differ between the CC→CC and GF→CC groups on P7, and bacterial community composition was also very similar, with a few exceptions. Thus, GF→CC offspring had altered brain development during at least the first 7 days after birth despite a largely normal microbiota. This suggests that prenatal influences of gestating in an altered microbial environment programs neonatal brain development.

3.
Brain Behav Immun ; 104: 122-136, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35661680

ABSTRACT

Birth is preceded by inflammation at the fetal/maternal interface. Additionally, the newborn experiences stimuli that under any other circumstance could elicit an immune response. It is unknown, however, whether birth elicits an inflammatory response in the newborn that extends to the brain. Moreover, it is unknown whether birth mode may alter such a response. To study these questions, we first measured corticosterone and pro- and anti-inflammatory cytokines in plasma of mouse offspring at several timepoints spaced closely before and after a vaginal or Cesarean birth. We found highest levels of IL-6 one day before birth and surges in corticosterone and IL-10 just after birth, regardless of birth mode. We next examined the neuroimmune response by measuring cytokine mRNA expression and microglial number and morphology in the paraventricular nucleus of the hypothalamus and hippocampus around the time of birth. We found a marked increase in TNF-α expression in both brain regions a day after birth, and rapid increases in microglial cell number in the first three days postnatal, with subtle differences by birth mode. To test whether the association between birth and cytokine production or expansion of microglia is causal, we manipulated birth timing. Remarkably, advancing birth by a day advanced the increases in all of the markers tested. Thus, birth triggers an immune response in the body and brain of offspring. Our results may provide a mechanism for effects of birth (e.g., acute changes in cell death and neural activation) previously reported in the newborn brain.

4.
Neuroendocrinology ; 112(7): 636-648, 2022.
Article in English | MEDLINE | ID: mdl-34547753

ABSTRACT

INTRODUCTION: Neurons expressing estrogen receptor (ER) ɑ in the arcuate (ARC) and ventromedial (VMH) nuclei of the hypothalamus sex-specifically control energy homeostasis, sexual behavior, and bone density. Females have more ERɑ neurons in the VMH and ARC than males, and the sex difference in the VMH is eliminated by neonatal treatment with testosterone or a DNA methylation inhibitor. OBJECTIVE: Here, we tested the roles of testosterone and DNA methylation/demethylation in development of ERɑ in the ARC. METHODS: ERɑ was examined at birth and weaning in mice that received vehicle or testosterone subcutaneously, and vehicle or DNA methyltransferase inhibitor intracerebroventricularly, as neonates. To examine effects of DNA demethylation on the ERɑ cell number in the ARC, mice were treated neonatally with small interfering RNAs against ten-eleven translocase enzymes. The methylation status of the ERɑ gene (Esr1) was determined in the ARC and VMH using pyrosequencing of bisulfite-converted DNA. RESULTS: A sex difference in ERɑ in the ARC, favoring females, developed between birth and weaning and was due to programming effects of testosterone. Neonatal inhibition of DNA methylation decreased ERɑ in the ARC of females, and an inhibition of demethylation increased ERɑ in the ARC of males. The promoter region of Esr1 exhibited a small sex difference in percent of total methylation in the ARC (females > males) that was opposite to that in the VMH (males > females). CONCLUSION: DNA methylation and demethylation regulate ERɑ cell number in the ARC, and methylation correlates with activation of Esr1 in this region.


Subject(s)
Arcuate Nucleus of Hypothalamus , Estrogen Receptor alpha , Animals , Arcuate Nucleus of Hypothalamus/metabolism , DNA Methylation , Demethylation , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Male , Mice , Sex Characteristics , Testosterone/pharmacology
5.
Horm Behav ; 136: 105080, 2021 11.
Article in English | MEDLINE | ID: mdl-34749276

ABSTRACT

Birth is an extraordinary event for placental mammals and occurs at a time when key developmental processes are shaping the brain. Remarkably, little is known about the contributions of birth to brain development and whether birth mode (vaginal vs. Cesarean) alters neurodevelopmental trajectories. We previously reported that Cesarean birth reduces vasopressin (VP) neuron number in the hypothalamic paraventricular nucleus (PVN) of mice at weaning. In this study, we investigated whether this effect extends to adulthood and whether birth mode affects oxytocin (OT) neurons, which are another prominent population in the PVN. We found that Cesarean-born adults had fewer VP neurons in the PVN, specifically in magnocellular regions. Interestingly, these regions also had more dying cells following a Cesarean birth, suggesting that cell death may be the underlying mechanism. The PVN of Cesarean-born adults also had smaller VP neuron somas and reduced VP efferent projections. Additionally, Cesarean-born mice showed fewer and smaller OT neurons in the PVN, but these effects were less robust than for VP neurons. We also examined VP and OT neuron number in the supraoptic and suprachiasmatic nuclei but found no effect of birth mode in these regions. Thus, Cesarean birth causes long-term effects on the VP and, to a lesser extent, OT systems in the PVN, suggesting that this region is particularly sensitive to the effects of birth mode. Our findings may help explain the social deficits reported for Cesarean-born mice, and are also of clinical significance given the widespread practice of Cesarean births across the world.


Subject(s)
Oxytocin , Paraventricular Hypothalamic Nucleus , Animals , Female , Mammals/metabolism , Mice , Neurons/metabolism , Oxytocin/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Placenta/metabolism , Pregnancy , Vasopressins/metabolism
6.
Front Cell Neurosci ; 15: 682505, 2021.
Article in English | MEDLINE | ID: mdl-34168540

ABSTRACT

The microbiota plays important roles in host metabolism and immunity, and its disruption affects adult brain physiology and behavior. Although such findings have been attributed to altered neurodevelopment, few studies have actually examined microbiota effects on the developing brain. This review focuses on developmental effects of the earliest exposure to microbes. At birth, the mammalian fetus enters a world teeming with microbes which colonize all body sites in contact with the environment. Bacteria reach the gut within a few hours of birth and cause a measurable response in the intestinal epithelium. In adults, the gut microbiota signals to the brain via the vagus nerve, bacterial metabolites, hormones, and immune signaling, and work in perinatal rodents is beginning to elucidate which of these signaling pathways herald the very first encounter with gut microbes in the neonate. Neural effects of the microbiota during the first few days of life include changes in neuronal cell death, microglia, and brain cytokine levels. In addition to these effects of direct exposure of the newborn to microbes, accumulating evidence points to a role for the maternal microbiota in affecting brain development via bacterial molecules and metabolites while the offspring is still in utero. Hence, perturbations to microbial exposure perinatally, such as through C-section delivery or antibiotic treatment, alter microbiota colonization and may have long-term neural consequences. The perinatal period is critical for brain development and a close look at microbiota effects during this time promises to reveal the earliest, most primary effects of the microbiota on neurodevelopment.

7.
Sci Rep ; 11(1): 2335, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504846

ABSTRACT

Long-standing clinical findings report a dramatic surge of vasopressin in umbilical cord blood of the human neonate, but the neural underpinnings and function(s) of this phenomenon remain obscure. We studied neural activation in perinatal mice and rats, and found that birth triggers activation of the suprachiasmatic, supraoptic, and paraventricular nuclei of the hypothalamus. This was seen whether mice were born vaginally or via Cesarean section (C-section), and when birth timing was experimentally manipulated. Neuronal phenotyping showed that the activated neurons were predominantly vasopressinergic, and vasopressin mRNA increased fivefold in the hypothalamus during the 2-3 days before birth. Copeptin, a surrogate marker of vasopressin, was elevated 30-to 50-fold in plasma of perinatal mice, with higher levels after a vaginal than a C-section birth. We also found an acute decrease in plasma osmolality after a vaginal, but not C-section birth, suggesting that the difference in vasopressin release between birth modes is functionally meaningful. When vasopressin was administered centrally to newborns, we found an ~ 50% reduction in neuronal cell death in specific brain areas. Collectively, our results identify a conserved neuroendocrine response to birth that is sensitive to birth mode, and influences peripheral physiology and neurodevelopment.


Subject(s)
Hypothalamus/metabolism , Neurosecretory Systems/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Vasopressins/metabolism , Animals , Biomarkers/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Osmoregulation/genetics , Osmoregulation/physiology , Vasopressins/genetics
8.
eNeuro ; 7(1)2020.
Article in English | MEDLINE | ID: mdl-32015098

ABSTRACT

Developmental cell death eliminates half of the neurons initially generated in the mammalian brain, and occurs perinatally in many species. It is possible that the timing of neuronal cell death is developmentally programmed, and only coincidentally associated with birth. Alternatively, birth may play a role in shaping cell death. To test these competing hypotheses, we experimentally advanced or delayed birth by 1 d in mice (within the normal range of gestation for the species) and examined effects on the temporal pattern and magnitude (amount) of neuronal cell death, using immunohistochemical detection of activated caspase-3 as a cell death marker. In order to detect effects of subtle changes in birth timing, we focused on brain areas that exhibit sharp postnatal peaks in cell death. We find that advancing birth advances peak cell death, supporting the hypothesis that birth triggers cell death. However, a delay of birth does not delay cell death. Thus, birth can advance cell death, but if postponed, a developmental program governs. Advancing or delaying birth also caused region-specific changes in the overall magnitude of cell death. Our findings shed light on the long-standing question of what controls the timing and magnitude of developmental neuronal cell death, and position birth as an orchestrator of brain development. Because humans across the world now routinely alter birth timing, these findings may have implications for current obstetric practices.


Subject(s)
Brain , Parturition , Animals , Apoptosis , Cell Death , Female , Mice , Neurons , Pregnancy
9.
Endocrinology ; 161(1)2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31742329

ABSTRACT

Many neural sex differences are differences in the number of neurons of a particular phenotype. For example, male rodents have more calbindin-expressing neurons in the medial preoptic area (mPOA) and bed nucleus of the stria terminalis (BNST), and females have more neurons expressing estrogen receptor alpha (ERα) and kisspeptin in the ventromedial nucleus of the hypothalamus (VMH) and the anteroventral periventricular nucleus (AVPV), respectively. These sex differences depend on neonatal exposure to testosterone, but the underlying molecular mechanisms are unknown. DNA methylation is important for cell phenotype differentiation throughout the developing organism. We hypothesized that testosterone causes sex differences in neurochemical phenotype via changes in DNA methylation, and tested this by inhibiting DNA methylation neonatally in male and female mice, and in females given a masculinizing dose of testosterone. Neonatal testosterone treatment masculinized calbindin, ERα and kisspeptin cell number of females at weaning. Inhibiting DNA methylation with zebularine increased calbindin cell number only in control females, thus eliminating sex differences in calbindin in the mPOA and BNST. Zebularine also reduced the sex difference in ERα cell number in the VMH, in this case by increasing ERα neuron number in males and testosterone-treated females. In contrast, the neonatal inhibition of DNA methylation had no effect on kisspeptin cell number. We conclude that testosterone normally increases the number of calbindin cells and reduces ERα cells in males through orchestrated changes in DNA methylation, contributing to, or causing, the sex differences in both cell types.


Subject(s)
Brain/drug effects , DNA Methylation/drug effects , Sex Differentiation/drug effects , Testosterone/pharmacology , Animals , Animals, Newborn , Brain/cytology , Brain/metabolism , Brain Chemistry/drug effects , Calbindins/metabolism , Cytidine/administration & dosage , Cytidine/analogs & derivatives , Cytidine/pharmacology , Estrogen Receptor alpha/metabolism , Female , Kisspeptins/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Sex Differentiation/physiology , Sex Factors , Testosterone/administration & dosage
10.
Dev Neurobiol ; 79(8): 769-779, 2019 08.
Article in English | MEDLINE | ID: mdl-31207134

ABSTRACT

Developmental neuronal cell death has been characterized as a cell autonomous "suicide" program, but recent findings suggest that microglia play an active role in determining the survival of developing neurons. Results have been contradictory, however, with some studies concluding that microglia promote cell death, while others report that microglia are neuroprotective. Here, we depleted microglia throughout the newborn mouse brain using intracerebroventricular injections of clodronate liposomes, and examined effects on naturally occurring cell death across multiple brain areas. Microglial density varied significantly by brain region, and clodronate liposome treatment at birth reduced the number of microglia in all regions examined. The effect of microglia reduction on cell death, however, varied by region: the number of dying cells was reduced in the medial septum and medial amygdala in clodronate treated animals, but was increased in the oriens layer of the hippocampus, and unchanged in several other brain regions. In most brain regions, the average size of microglia was greater in microglia-depleted than in control animals, suggesting that the remaining microglia compensate to some extent for a reduction in microglial number. The hippocampal oriens was exceptional in this regard, in that microglial size was reduced following treatment with clodronate. Microglia produce cytokines which mediate many of their effects, and we found higher expression of inflammatory cytokines in the hippocampus than in the septum, independent of clodronate treatment. Thus, microglial depletion has opposite effects on cell death in different brain regions of the newborn brain, which may be related to regional heterogeneity in microglia.


Subject(s)
Brain/cytology , Microglia/cytology , Animals , Animals, Newborn , Brain/drug effects , Cell Death/drug effects , Clodronic Acid/pharmacology , Mice, Inbred C57BL , Microglia/metabolism , Neurogenesis/drug effects , Neurons/drug effects
11.
Exp Neurol ; 311: 305-312, 2019 01.
Article in English | MEDLINE | ID: mdl-30031021

ABSTRACT

Current understanding of the pathogenesis of the familial form of amyotrophic lateral sclerosis has been aided by the study of transgenic mice that over-express mutated forms of the human CuZn-superoxide dismutase (SOD1) gene. While mutant SOD1 in motor neurons determines disease onset, other non-cell autonomous factors are critical for disease progression, and altered energy metabolism has been implicated as a contributing factor. Since most energy expended by laboratory mice is utilized to defend body temperature (Tb), we analyzed thermoregulation in transgenic mice carrying the G93A mutation of the human SOD1 gene, using implantable temperature data loggers to continuously record Tb for up to 85 days. At room (22 °C) ambient temperature, G93A mice exhibited a diminished amplitude of the daily Tb rhythm compared to C57BL/6J controls, secondary to decreased Tb values during the dark (behaviorally active) phase of the light-dark cycle. The defect arose at 85-99 days of age, around the age of symptom onset (as assessed by grip strength), well before observable weakness and weight loss, and could not be accounted for by decreased levels of locomotor activity or food consumption. Housing under thermoneutral (29 °C) ambient temperature partially rescued the defect, but age-dependently (only in animals >100 days of age), suggesting that the deficit in older mice was due in part to inadequate thermogenesis by "peripheral" thermogenic organs as the disease progressed. In younger mice, we found that cold-induced thermogenesis and energy expenditure were intact, hinting that an initial "central" defect might localize to the subparaventricular zone, involving neural output pathways from the circadian clock in the hypothalamic suprachiasmatic nucleus to forebrain thermoregulatory circuitry.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Body Temperature Regulation/physiology , Circadian Rhythm/physiology , Disease Models, Animal , Amyotrophic Lateral Sclerosis/enzymology , Animals , Humans , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics
12.
Proc Natl Acad Sci U S A ; 115(46): 11826-11831, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30322936

ABSTRACT

Labor and a vaginal delivery trigger changes in peripheral organs that prepare the mammalian fetus to survive ex utero. Surprisingly little attention has been given to whether birth also influences the brain, and to how alterations in birth mode affect neonatal brain development. These are important questions, given the high rates of cesarean section (C-section) delivery worldwide, many of which are elective. We examined the effect of birth mode on neuronal cell death, a widespread developmental process that occurs primarily during the first postnatal week in mice. Timed-pregnant dams were randomly assigned to C-section deliveries that were yoked to vaginal births to carefully match gestation length and circadian time of parturition. Compared with rates of cell death just before birth, vaginally-born offspring had an abrupt, transient decrease in cell death in many brain regions, suggesting that a vaginal delivery is neuroprotective. In contrast, cell death was either unchanged or increased in C-section-born mice. Effects of delivery mode on cell death were greatest for the paraventricular nucleus of the hypothalamus (PVN), which is central to the stress response and brain-immune interactions. The greater cell death in the PVN of C-section-delivered newborns was associated with a reduction in the number of PVN neurons expressing vasopressin at weaning. C-section-delivered mice also showed altered vocalizations in a maternal separation test and greater body mass at weaning. Our results suggest that vaginal birth acutely impacts brain development, and that alterations in birth mode may have lasting consequences.


Subject(s)
Brain/embryology , Cesarean Section/adverse effects , Parturition/physiology , Animals , Cell Death/physiology , Delivery, Obstetric/veterinary , Female , Gestational Age , Labor, Obstetric/physiology , Mice , Mice, Inbred C57BL , Paraventricular Hypothalamic Nucleus/physiology , Pregnancy
13.
Biol Sex Differ ; 9(1): 7, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29351816

ABSTRACT

BACKGROUND: Gut dysbiosis is observed in several neuropsychiatric disorders exhibiting increases in anxiety behavior, and recent work suggests links between gut inflammation and such disorders. One source of this inflammation may be lipopolysaccharide (LPS), a toxic component of gram-negative bacteria. Here, we (1) determine whether oral gavage of LPS, as a model of gut-derived endotoxemia, affects anxiety-like and/or repetitive behaviors; (2) test whether these changes depend on TLR4 signaling; and (3) test the extent to which gut-derived endotoxin and TLR4 antagonism affects males and females differently. METHODS: In experiment 1, male wild-type (WT) and Tlr4-/- mice were tested for locomotor, anxiety-like, and repetitive behaviors in an automated open field test apparatus, 2 h after oral gavage of LPS or saline. In experiment 2, male and female WT mice received an oral gavage of LPS and an injection of one or two TLR4 antagonists that target different TLR4 signaling pathways ((+)-naloxone and LPS derived from R. sphaeroides (LPS-RS)). Univariate and multivariate analyses were used to identify effects of treatment, sex, and genotype and their interaction. RESULTS: In experiment 1, oral gavage of LPS increased anxiety-like behavior in male WT mice but not in Tlr4-/- mice. In experiment 2, oral gavage of LPS increased anxiety-like and decreased repetitive behaviors in WT mice of both sexes. Neither antagonist directly blocked the effects of orally administered LPS. However, treatment with (+)-naloxone, which blocks the TRIF pathway of TLR4, had opposing behavioral effects in males and females (independent of LPS treatment). We also identified sex differences in the expression of interleukin-6, a pro-inflammatory cytokine, in the gut both in basal conditions and in response to LPS. CONCLUSION: In spite of the ubiquitous nature of LPS in the gut lumen, this is the first study to demonstrate that intestinally derived LPS can initiate behavioral aspects of the sickness response. While an increased enteric load of LPS increases anxiety-like behavior in both sexes, it likely does so via sex-specific mechanisms. Similarly, TLR4 signaling may promote baseline expression of repetitive behavior differently in males and females. This study lays the groundwork for future interrogations into connections between gut-derived endotoxin and behavioral pathology in males and females.


Subject(s)
Anxiety/chemically induced , Behavior, Animal/drug effects , Dysbiosis , Lipopolysaccharides/pharmacology , Animals , Female , Gastrointestinal Microbiome , Male , Mice, Inbred C57BL , Mice, Knockout , Naloxone/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics
14.
Sci Rep ; 8(1): 1202, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352256

ABSTRACT

Sociality has beneficial effects on fitness, and timing the activities of animals may be critical. Social cues could influence daily rhythmic activities via direct effects on the circadian clock or on processes that bypass it (masking), but these possibilities remain incompletely addressed. We investigated the effects of social cues on the circadian body temperature (Tb) rhythms in pairs of co-housed and isolated grass rats, Arvicanthis niloticus (a social species), in constant darkness (DD). Cohabitation did not induce synchronization of circadian Tb rhythms. However, socio-sexual history did affect circadian properties: accelerating the clock in sexually experienced males and females in DD and advancing rhythm phase in the females in a light-dark cycle. To address whether synchronization occurs at an ultradian scale, we analyzed Tb and activity rhythms in pairs of co-housed sisters or couples in DD. Regardless of pair type, co-housing doubled the percentage of time individuals were simultaneously active without increasing individual activity levels, suggesting that activity bouts were synchronized by redistribution over 24 h. Together, our laboratory findings show that social cues affect individual "time allocation" budgets via mechanisms at multiple levels of biological organization. We speculate that in natural settings these effects could be adaptive, especially for group-living animals.


Subject(s)
Behavior, Animal , Rodentia , Social Behavior , Animals , Circadian Rhythm , Female , Male , Photoperiod , Time Management
15.
Brain Behav Immun ; 67: 218-229, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28890156

ABSTRACT

The mammalian fetus develops in a largely sterile environment, and direct exposure to a complex microbiota does not occur until birth. We took advantage of this to examine the effect of the microbiota on brain development during the first few days of life. The expression of anti- and pro-inflammatory cytokines, developmental cell death, and microglial colonization in the brain were compared between newborn conventionally colonized mice and mice born in sterile, germ-free (GF) conditions. Expression of the pro-inflammatory cytokines interleukin 1ß and tumor necrosis factor α was markedly suppressed in GF newborns. GF mice also had altered cell death, with some regions exhibiting higher rates (paraventricular nucleus of the hypothalamus and the CA1 oriens layer of the hippocampus) and other regions exhibiting no change or lower rates (arcuate nucleus of the hypothalamus) of cell death. Microglial labeling was elevated in GF mice, due to an increase in both microglial cell size and number. The changes in cytokine expression, cell death and microglial labeling were evident on the day of birth, but were absent on embryonic day 18.5, approximately one-half day prior to expected delivery. Taken together, our results suggest that direct exposure to the microbiota at birth influences key neurodevelopmental events and does so within hours. These findings may help to explain some of the behavioral and neurochemical alterations previously seen in adult GF mice.


Subject(s)
Brain/growth & development , Cell Death , Encephalitis/microbiology , Microbiota , Microglia/physiology , Neurons/physiology , Animals , Brain/microbiology , Encephalitis/metabolism , Female , Inflammation Mediators/metabolism , Male , Mice , Microglia/microbiology , Neurons/microbiology , Pregnancy
16.
Endocrinology ; 158(6): 1838-1848, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28398586

ABSTRACT

Many of the best-studied neural sex differences relate to differences in cell number and are due to the hormonal control of developmental cell death. However, several prominent neural sex differences persist even if cell death is eliminated. We hypothesized that these may reflect cell phenotype "decisions" that depend on epigenetic mechanisms, such as DNA methylation. To test this, we treated newborn mice with the DNA methyltransferase (DNMT) inhibitor zebularine, or vehicle, and examined two sexually dimorphic markers at weaning. As expected, control males had more cells immunoreactive for calbindin-D28k (CALB) in the medial preoptic area (mPOA) and fewer cells immunoreactive for estrogen receptor α (ERα) in the ventrolateral portion of the ventromedial nucleus of the hypothalamus (VMHvl) and the mPOA than did females. Neonatal DNMT inhibition markedly increased CALB cell number in both sexes and ERα cell density in males; as a result, the sex differences in ERα in the VMHvl and mPOA were completely eliminated in zebularine-treated animals. Zebularine treatment did not affect developmental cell death or the total density of Nissl-stained cells at weaning. Thus, a neonatal disruption of DNA methylation apparently has long-term effects on the proportion of cells expressing CALB and ERα, and some of these effects are sex specific. We also found that sex differences in CALB in the mPOA and ERα in the VMHvl persist in mice with a neuron-specific depletion of either Dnmt1 or Dnmt3b, indicating that neither DNMT alone is likely to be required for the sexually dimorphic expression of these markers.


Subject(s)
Brain/drug effects , Cytidine/analogs & derivatives , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Neurons/drug effects , Sex Characteristics , Animals , Animals, Newborn , Brain/cytology , Brain/growth & development , Cytidine/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1 , Down-Regulation/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/physiology , Phenotype , Sex Determination Processes/drug effects , Sex Determination Processes/genetics , Time Factors
17.
Front Neuroendocrinol ; 40: 67-86, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26790970

ABSTRACT

Neuroscientists are likely to discover new sex differences in the coming years, spurred by the National Institutes of Health initiative to include both sexes in preclinical studies. This review summarizes the current state of knowledge of the cellular and molecular mechanisms underlying sex differences in the mammalian nervous system, based primarily on work in rodents. Cellular mechanisms examined include neurogenesis, migration, the differentiation of neurochemical and morphological cell phenotype, and cell death. At the molecular level we discuss evolving roles for epigenetics, sex chromosome complement, the immune system, and newly identified cell signaling pathways. We review recent findings on the role of the environment, as well as genome-wide studies with some surprising results, causing us to re-think often-used models of sexual differentiation. We end by pointing to future directions, including an increased awareness of the important contributions of tissues outside of the nervous system to sexual differentiation of the brain.


Subject(s)
Brain/physiology , Mammals/immunology , Neurogenesis/immunology , Neurogenesis/physiology , Sex Differentiation/physiology , Signal Transduction/immunology , Animals , Humans , Mammals/physiology , Sex Chromosomes/physiology , Sex Differentiation/immunology
18.
Prog Brain Res ; 199: 267-280, 2012.
Article in English | MEDLINE | ID: mdl-22877671

ABSTRACT

Circadian rhythms can be entrained to periodic cues in the environment including the solar day, food resources, and temperature. Work on a variety of organisms has suggested that social interactions within and between species may also influence circadian rhythmicity, but conceptual and technical difficulties relating to animal models, housing environments, rhythm assays, and experimental design have complicated mechanistic investigations in the laboratory. We review these issues and introduce the gregarious Nile grass rat, Arvicanthis niloticus, as a suitable model for research on this problem. Understanding social influences on temporal organization at this supra-organismal, community level is of considerable translational value, as its implications range from conservation biology to human health.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Interpersonal Relations , Animals , Behavior, Animal , Environment , Humans , Motor Activity , Rats
19.
Physiol Behav ; 103(3-4): 384-92, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21402088

ABSTRACT

In the diurnal grass rat nocturnal voluntary wakefulness induces Fos expression in specific cellular populations of arousal and reward areas of the brain. Here, we evaluated whether involuntary wakefulness would result in similar patterns of Fos expression. We assessed this question using male grass rats that were sleep deprived for 6h by gentle stimulation (SD group), starting 2h before lights off (12:12 LD cycle). Then, we examined expression of Fos in cholinergic cells of the basal forebrain (BF), as well as in dopaminergic cells of the reward system, and compared these results to those obtained from an undisturbed control group. Different from previous results with grass rats that were voluntary awake, the BF of SD animals only showed a significant increase in Fos expression in non-cholinergic neurons of the medial septum (MS). These observations differ from reports for nocturnal rodents that are sleep deprived. Thus, our results show that voluntary and induced wakefulness have different effects on neural systems involved in wakefulness and reward, and that the effects of sleep deprivation are different across species. We also investigated whether other arousal promoting regions and circadian and stress related areas responded to sleep deprivation by changing the level of Fos expression. Among these areas, only the lateral hypothalamus (LH) and the ventro lateral preoptic area showed significant effects of sleep deprivation that dissipated after a 2h period of sleep recovery, as it was also the case for the non-cholinergic MS. In addition, we found that Fos expression in the LH was robustly associated with Fos expression in other arousal and reward areas of the brain. This is consistent with the view that the arousal system of the LH modulates neural activity of other arousal regions of the brain, as described for nocturnal rodents.


Subject(s)
Arousal/physiology , Brain/metabolism , Oncogene Proteins v-fos/metabolism , Reward , Wakefulness/physiology , Animals , Brain/anatomy & histology , Cell Count/methods , Choline O-Acetyltransferase/metabolism , Male , Rats
20.
Brain Res ; 1151: 91-101, 2007 Jun 02.
Article in English | MEDLINE | ID: mdl-17397808

ABSTRACT

In nocturnal species cholinergic agonists alter circadian rhythm phase when injected intraventricularly or directly into the suprachiasmatic nucleus (SCN), but the phase shifts obtained differ depending upon the site being injected. Cholinergic projections reach the SCN of nocturnal laboratory rats, however, nothing is known about these projections in diurnal rodents. The first objective of this study was to evaluate the hypothesis that cholinergic projections to the SCN are only present in nocturnal species. The second objective was to evaluate the hypothesis that the lower part of the subparaventricular zone (LSPV) is a candidate for being a site that mediates the phase shifts observed when cholinergic agonists are injected intraventricularly. These hypotheses were tested in the diurnal unstriped Nile grass rat (Arvicanthis niloticus) and the nocturnal laboratory rat. Additionally, we evaluated if the light-dark (LD) cycle had an effect on the expression of the vesicular acetylcholine transporter (VAChT) in the SCN, LSPV, and in two control areas. Animals were kept in a 12:12 LD cycle and perfused at six time points. VAChT immunoreactivity was observed in the SCN, LSPV, and in the control areas of both species. The SCN and LSPV showed a differential distribution and density of cholinergic projections between the two species, but similar temporal patterns of VAChT expression were found across species. These results provide evidence for a differential cholinergic stimulation of the SCN between grass rats and laboratory rats that may reflect a rewiring of neural projections brought about by the adoption of a diurnal activity profile.


Subject(s)
Cholinergic Fibers/physiology , Circadian Rhythm , Gene Expression Regulation/physiology , Paraventricular Hypothalamic Nucleus/physiology , Rodentia/anatomy & histology , Suprachiasmatic Nucleus/physiology , Analysis of Variance , Animals , Male , Neural Pathways , Rats , Rats, Sprague-Dawley , Species Specificity , Vesicular Acetylcholine Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...