Elsevier

Neuroscience

Volume 428, 21 January 2020, Pages 76-89
Neuroscience

Research Article
Enduring and Sex-specific Changes in Hippocampal Gene Expression after a Subchronic Immune Challenge

https://doi.org/10.1016/j.neuroscience.2019.12.019Get rights and content

Highlights

  • Subchronic immune challenge causes persistent changes in hippocampal gene expression in both sexes.

  • Males show dysregulation of immune- and plasticity related genes in hippocampus months after immune challenge.

  • Females show few changes in gene expression, but altered expression of monoaminergic-related pathways.

Abstract

Major illnesses, including heart attack and sepsis, can cause cognitive impairments, depression, and progressive memory decline that persist long after recovery from the original illness. In rodent models of sepsis or subchronic immune challenge, memory deficits also persist for weeks or months, even in the absence of ongoing neuroimmune activation. This raises the question of what mechanisms in the brain mediate such persistent changes in neural function. Here, we used RNA-sequencing as a large-scale, unbiased approach to identify changes in hippocampal gene expression long after a subchronic immune challenge previously established to cause persistent memory impairments in both males and females. We observed enduring dysregulation of gene expression three months after the end of a subchronic immune challenge. Surprisingly, there were striking sex differences in both the magnitude of changes and the specific genes and pathways altered, where males showed persistent changes in both immune- and plasticity-related genes three months after immune challenge, whereas females showed few such changes. In contrast, females showed striking differential gene expression in response to a subsequent immune challenge. Thus, immune activation has enduring and sex-specific consequences for hippocampal gene expression and the transcriptional response to subsequent stimuli. Together with findings of long-lasting memory impairments after immune challenge, these data suggest that illnesses can cause enduring vulnerability to, cognitive decline, affective disorders, and memory impairments via dysregulation of transcriptional processes in the brain.

Introduction

The neuroimmune system, and its regulation by peripheral immune states, plays an important regulatory role for behavioral, affective, and cognitive functions (Raison et al., 2006, Tchessalova et al., 2018). In addition to their central role in behavioral and physiological changes during illness and injury (Dantzer et al., 1998), neuroimmune cells and cytokines are activated as a consequence of physical and psychological stressors (Bekhbat and Neigh, 2017), and are critical for normal neural functions including synaptic plasticity and memory (Avital et al., 2003, Yirmiya and Goshen, 2011, del Rey et al., 2013, Adamsky et al., 2018).

Activation of the neuroimmune system as a consequence of surgery, injury, or illness also has long-lasting consequences for memory, affective behaviors, and cognition. In rodent models of sepsis, persistent changes in memory and affective processes have been observed after cecal ligation and puncture (CLP) (Chavan et al., 2012, Huerta et al., 2016) and after high-dose lipopolysaccharide (LPS) treatment (Weberpals et al., 2009, Anderson et al., 2015). We have recently demonstrated that a lower-dose subchronic systemic immune challenge results in striking memory deficits that emerge and persist at least three months after the end of the immune challenge (Tchessalova and Tronson, 2019). These long-lasting memory deficits are likely mediated by changes in neuroimmune and neuronal function that persist long after immune activation and likely contribute to cognitive decline, affective dysregulation, and increased risk of dementia and Alzheimer’s disease long after recovery from major illnesses including sepsis and heart attack (Semmler et al., 2007, Gharacholou et al., 2011, Marra et al., 2018).

That transient illness or immune challenge can result in long-lasting memory deficits is now well established, but it is less clear what is persistently changed in the brain to mediate changes in cognition and memory that persist long after resolution of the immune response. One possibility is that neuroimmune signaling lingers long after a peripheral immune challenge. Indeed, some sepsis models observe that persistent microglial activation and neuroimmune signaling correlate with memory impairments at least one month after surgery or LPS injection (Weberpals et al., 2009, Fenn et al., 2014, Anderson et al., 2015). In contrast, other studies observe memory deficits in the absence of ongoing neuroimmune activation in both CLP and LPS models, suggesting that persistent neuroimmune activation is not required for late-emerging memory impairments (Ming et al., 2015, Huerta et al., 2016, Singer et al., 2016, Tchessalova and Tronson, 2019). In these studies, structural changes in the brain after transient immune challenge, including dysregulation of dendritic spine turnover or density and neuronal loss have been implicated as potential mechanisms for persistent memory impairments (Semmler et al., 2007, Liu et al., 2008, Kondo et al., 2011, Volpe et al., 2015). Additionally, recent work has demonstrated changes in function of seemingly quiescent microglia months after prior immune challenge (Wendeln et al., 2018). Thus prior immune experience may result in “priming” of microglia (Norden et al., 2015), or “training” of the innate immune system (Netea and van der Meer, 2017), in which immune cells respond in an exaggerated way to subsequent immune, stress, or other experiences.

If ongoing neuroimmune signaling is not necessary for persistent changes in memory, then how are such long-lasting changes in synaptic function, neuroimmune function, and memory processes maintained long after the immune challenge? One possibility is that these persistent changes in memory are mediated and maintained by enduring alterations of gene expression and associated transcriptional regulatory mechanisms (Tchessalova et al., 2018). Changes in transcriptional regulation as a result of chromatin and DNA modifications (Cholewa-Waclaw et al., 2016) occur after a variety of environmental exposures and life experience, including stress and exposure to addictive drugs, and mediate persistent changes in cellular function and behavior (Gray et al., 2014, Nestler, 2014, Tafet and Nemeroff, 2016).

We have previously demonstrated that a subchronic LPS challenge results in memory impairments that emerge and persist months after the last injection (Tchessalova and Tronson, 2019). Here we examined whether the same protocol also causes changes in hippocampal gene expression that persist long after an immune challenge in males and in females. We used an unbiased approach, RNA-sequencing, to identify both immune- and non-immune-related gene expression changes long after a systemic immune challenge in both males and females. Since changes in gene expression after experiences such as stress (Gray et al., 2014) are often more evident in response to a subsequent stressor, we determined gene expression both months after a subchronic immune challenge, and in response to a subsequent immune challenge. We found that a transient, subchronic LPS protocol (Tchessalova and Tronson, 2019) resulted in persistent and sex-specific dysregulation of gene expression in the hippocampus. Males and females differed in both number and patterns of gene expression, where males show more changes in baseline gene expression and females show greater changes in response to an additional, acute challenge. These data have important implications for identifying the precise mechanisms by which a prior illness or injury can impact on cognitive function and vulnerability to cognitive decline and dementia.

Section snippets

Animals

Twenty four male and female 8–9 week old C57BL/6N mice were purchased from Envigo (Indianapolis, IN, USA). The sample size for these experiments was n = 6 per immune challenge condition (Vehicle, Long-Term, Long-term+Acute, Acute), with 3 males and 3 females per group. All mice were individually housed with ad libitum access to standard mouse chow and water as individual housing in mice prevents fighting-induced stress in males (Meakin et al., 2013) and is ethologically appropriate for males

Subchronic immune challenge induces persistent changes in gene expression

We observed striking changes in gene expression in the hippocampus 12 weeks after subchronic immune challenge in males, with fewer changes observed in females. Of over 20,000 genes detected, there were 230 DEGs in the hippocampus of males and 26 DEGs in the hippocampus of females (Fig. 1). In males, 183 genes were significantly upregulated and 47 significantly downregulated (see https://doi.org/10.7302/7zz3-jh58 for Supp. Table 1 for a full list of DEGs). In females, 7 genes were significantly

Discussion

Here we demonstrated long-lasting consequences of subchronic immune challenge, and striking sex differences, on gene expression in the hippocampus. In males, we observed enduring dysregulation of gene expression months after the end of a two-week immune challenge. In contrast, females showed few persistent changes at baseline, but striking changes in gene expression in response to an additional acute LPS injection three months after the subchronic immune challenge. These different

Acknowledgements

Thanks to University of Michigan Bioinformatics core for their expertise and data analysis of differentially expressed genes and the University of Michigan DNA sequencing core for their processing of samples; to Dr. Shigeki Iwase and Dr. Christina Vallianatos for assistance with prepping of the RNA samples; Dr. Dave Bridges for helpful discussions on RNAseq and analyses; Ms. Brynne Raines, Ms. Melanie Gil, and Ms. Caitlin Posillico for their extensive feedback on this manuscript.

Funding and disclosures

NIH/NIMH R00 MH0934509 to NCT. The authors declare no conflicts of interest.

Data availability

This data is also publicly available on the following databases:

Gene Expression Omnibus (GEO), GEO accession number GSE126678; Sequence Read Archive (SRA), with SRA number SRP186132 and BioProject number PRJNA522922. Supplemental tables are available at Deep Blue Data https://doi.org/10.7302/7zz3-jh58.

References (71)

  • Z. Ming et al.

    Acute systemic LPS-mediated inflammation induces lasting changes in mouse cortical neuromodulation and behavior

    Neurosci Lett

    (2015)
  • R. Mychasiuk et al.

    Chronic stress induces persistent changes in global DNA methylation and gene expression in the medial prefrontal cortex, orbitofrontal cortex, and hippocampus

    Neuroscience

    (2016)
  • M.G. Netea et al.

    Trained immunity: an ancient way of remembering

    Cell Host Microbe

    (2017)
  • D.M. Norden et al.

    Microglial priming and enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative disease

    Neuropharmacology

    (2015)
  • C. Raison et al.

    Cytokines sing the blues: inflammation and the pathogenesis of depression

    Trends Immunol

    (2006)
  • M. Randesi et al.

    Sex differences after chronic stress in the expression of opioid- and neuroplasticity-related genes in the rat hippocampus

    Neurobiol Stress

    (2018)
  • S. Rosi

    Neuroinflammation and the plasticity-related immediate-early gene Arc

    Brain Behav Immun

    (2011)
  • A. Semmler et al.

    Long-term cognitive impairment, neuronal loss and reduced cortical cholinergic innervation after recovery from sepsis in a rodent model

    Exp Neurol

    (2007)
  • J. Serrats et al.

    Pro-inflammatory immune-to-brain signaling is involved in neuroendocrine responses to acute emotional stress

    Brain Behav Immun

    (2017)
  • I. Song et al.

    Crosstalk between glia, extracellular matrix and neurons

    Brain Res Bull

    (2018)
  • E.A. Stone et al.

    Brain alpha 1-adrenergic neurotransmission is necessary for behavioral activation to environmental change in mice

    Neuroscience

    (1999)
  • D. Tchessalova et al.

    Memory deficits in males and females long after subchronic immune challenge

    Neurobiol Learn Mem

    (2019)
  • D.M. Walker

    Cocaine self-administration alters transcriptome-wide responses in the brain’s reward circuitry

    Biol Psychiatry

    (2018)
  • C. Wang et al.

    Ventral hippocampus modulates anxiety-like behavior in male but not female C57BL/6 J mice

    Neuroscience

    (2019)
  • R. Yirmiya et al.

    Immune modulation of learning, memory, neural plasticity and neurogenesis

    Brain Behav Immun

    (2011)
  • A. Avital et al.

    Impaired interleukin-1 signaling is associated with deficits in hippocampal memory processes and neural plasticity

    Hippocampus

    (2003)
  • R.D. Badgaiyan et al.

    Dopamine release during human emotional processing

    Neuroimage

    (2010)
  • J.B. Becker et al.

    Sex differences in animal models: Focus on addiction

    Pharmacol Rev

    (2016)
  • M. Bekhbat et al.

    Sex differences in the neuro-immune consequences of stress: focus on depression and anxiety

    Brain Behav Immun

    (2017)
  • S.S. Chavan et al.

    HMGB1 mediates cognitive impairment in sepsis survivors

    Mol Med

    (2012)
  • J. Cholewa-Waclaw et al.

    The role of epigenetic mechanisms in the regulation of gene expression in the nervous system

    J Neurosci

    (2016)
  • F.T. Crews et al.

    Neuroimmune function and the consequences of alcohol exposure

    Alcohol Res

    (2015)
  • R. Dantzer et al.

    Cytokines and sickness behavior

    Ann N Y Acad Sci

    (1998)
  • D.A. Finn et al.

    Binge ethanol drinking produces sexually divergent and distinct changes in nucleus accumbens signaling cascades and pathways in adult C57BL/6J mice

    Front Genet

    (2018)
  • M.G. Frank et al.

    Stress-induced glucocorticoids as a neuroendocrine alarm signal of danger

    Brain Behav Immun

    (2017)
  • Cited by (10)

    • Early-life stress affects peripheral, blood-brain barrier, and brain responses to immune challenge in juvenile and adult rats

      2023, Brain, Behavior, and Immunity
      Citation Excerpt :

      On the other hand, transcriptome analysis of isolated microglia from the HP of adult mice revealed the opposite trend, i.e., under basal conditions, male microglia had a more inflammatory phenotype, whereas female microglia showed a neuroprotective phenotype (Villa et al., 2018). Other studies on mice demonstrated that adult males also had greater and enduring responses to immune challenge in the HP than females (Tchessalova and Tronson, 2020; Posillico et al., 2021). Our results concerning sex differences in the healthy and LPS-challenged immune systems of control rats did not fall into any of the patterns described above.

    • Adolescent social instability stress leads to immediate and lasting sex-specific changes in the neuroendocrine-immune-gut axis in rats

      2020, Hormones and Behavior
      Citation Excerpt :

      The RT-qPCR was conducted separately for females and males, and thus we cannot address sex differences directly. Nevertheless, the effects of SS and of LPS-treatment suggested sex-specific patterns of gene expression, consistent with other reports indicating sex differences in hippocampal gene expression in response to LPS (e.g., Tchessalova and Tronson, 2020). In brief, across the measures, gene expression in the hippocampus was altered 48 h after treatment with LPS, with bigger effects of LPS evident in CTL than in SS female rats, whereas the opposite was observed for male rats.

    View all citing articles on Scopus
    View full text