Elsevier

Physiology & Behavior

Volume 225, 15 October 2020, 113082
Physiology & Behavior

Gut microbiota composition modulates inflammation and structure of the vagal afferent pathway

https://doi.org/10.1016/j.physbeh.2020.113082Get rights and content

Highlights

  • Both depletion models (antibiotic depletion and germ free) led to receiver rats displaying microbiota profile characteristic of their respective donors.

  • Colonization with a dysbiotic microbiota led to an increase in body weight gain and food intake.

  • Microbiota composition modulates Iba1+ immune cell presence along the gut-brain axis

  • Colonization with a dysbiotic microbiota led to a reduction on vagal innervation to the NTS associated with a loss of CCK-induced satiety.

Abstract

Vagal afferent neurons (VAN), located in the nodose ganglion (NG) innervate the gut and terminate in the nucleus of solitary tract (NTS) in the brainstem. They are the primary sensory neurons integrating gut-derived signals to regulate meal size. Chronic high-fat diet (HFD) consumption impairs vagally mediated satiety, resulting in overfeeding. There is evidence that HFD consumption leads to alterations in both vagal nerve function and structural integrity. HFD also leads to marked gut microbiota dysbiosis; in rodent models, dysbiosis is sufficient to induce weight gain. In this study, we investigated the effect of microbiota dysbiosis on gut-brain vagal innervation independently of diet. To do so, we recolonized microbiota-depleted rats with gastrointestinal (GI) contents isolated from donor animals fed either a HFD (45 or 60% fat) or a low fat diet (LFD, 13% fat). We used two different depletion models while maintaining the animals on LFD: 1) conventionally raised Fischer and Wistar rats that underwent a depletion paradigm using an antibiotic cocktail and 2) germ free (GF) raised Fischer rats. Following recolonization, receiver animals were designated as ConvLF and ConvHF. Fecal samples were collected throughout these studies and analyzed via 16S Illumina sequencing. In both models, bacteria that were identified as characteristic of HFD were successfully transferred to recipient animals. Three weeks post-colonization, ConvHF rats showed significant increases in ionized calcium-binding adapter molecule-1 (Iba1) positive immune cells in the NG compared to ConvLF animals. Additionally, using isolectin B4 (IB4) staining to identify c-fibers, we found that, compared to ConvLF animals, ConvHF rats displayed decreased innervation at the level of the medial NTS; c-fibers at this level are believed to be primarily of vagal origin. This alteration in vagal structure was associated with a loss in satiety induced by the gut peptide cholecystokinin (CCK). Increased presence of immunocompetent Iba1+ cells along the gut-brain axis and alterations in NTS innervation were still evident in ConvHF rats compared to ConvLF animals 12 weeks post-colonization and were associated with increases in food intake and body weight (BW). We conclude from these data that microbiota dysbiosis can alter gut-brain vagal innervation, potentially via recruitment and/or activation of immune cells.

Introduction

Obesity is a major health issue in Western countries. In the U.S., about 40% of the adult population is obese and an additional 32% is overweight [1]. Obesity raises the risk for both metabolic and psychological disorders [2,3], and the economic burden from health care costs related to obesity is estimated at $147 billion per year [4]. Underlying the development of obesity are genetic predispositions and consumption of energy-dense food such as high fat diets (HFDs) [5,6].

In rodent models, HFD feeding leads to an increase in meal size and overall intake [7]. Gut-originating satiety peptides, such as cholecystokinin (CCK), signal via the vagus nerve to trigger meal termination [8,9]. Vagal afferent neurons (VAN) are the primary sensory neurons integrating these gut-derived signals and relaying them to the nucleus of solitary tract (NTS) in the brainstem; postprandial activation of vagal fibers increases neuronal activity in the NTS [10]. HFD consumption in rodent models leads to decreased sensitivity of VAN to satiety peptides, such as CCK [11], [12], [13], [14], and intestinal nutrients [15], [16], [17], [18], as well as reduced postprandial NTS neuronal activation [12,18]. Disrupting the vagal afferent pathway is sufficient to cause hyperphagia and weight gain, identifying aberrant gut-brain signaling as a triggering factor for obesity [19].

The mechanisms by which HFD affects vagal signaling are not fully understood, however, there is evidence that inflammation may play a role in diet-driven vagal alterations. We have previously shown that leptin enhances CCK-induced satiety [20] and HFD leads to vagal leptin resistance and reduced CCK sensitivity, potentially via a suppressor of cytokine signaling 3 (SOCS3) dependent pathway [16]. SOCS3 is a negative regulator of cytokine signaling and its expression is upregulated during chronic inflammation [21,22]. Interestingly, HFD consumption leads to a rapid increase in ionized calcium binding adaptor molecule 1 (Iba1) positive cells in the nodose ganglia (NG) [23,24], where VAN cell bodies are located, and in the NTS [25]. Iba1 is a pan-marker for monocytes and microglia. Microglia are the resident parenchymal myeloid cells of the central nervous system (CNS) [26]. Microglia activation and microglia-induced inflammation are observed in several brain pathologies, and there is evidence that chronically activated microglia exert direct deleterious effects on neurons [27], [28], [29]. HFD consumption leads to microglia-mediated inflammation in the CNS [30], [31], [32] that is associated with neuronal death [32].

Increases in Iba1+ cells along the gut-brain axis are accompanied by alterations in vagal structure. HFD consumption leads to a decrease in isolectin B4 (IB4) staining at the level of the medial NTS. IB4 staining has been used as a proxy for vagal projections as it binds to unmyelinated c-fibers, which at the level of the NTS, are predominantly of vagal origin [33]. Additionally, caspase deletion of vagal c-fibers abolishes IB4 labeling in the medial NTS [34]. The timing of NTS vagal withdrawal coincides with onset of weight gain and hyperphagia in HFD fed rats [20]. Interestingly, in HFD fed rats, a decrease in Iba1+ cells presence along the gut-brain axis is associated with normalization of NTS IB4 staining and reduced food intake [25,35]. These data suggest that HFD-associated inflammation may induce structural remodeling of the vagal afferent pathway, potentially altering vagal signaling and regulation of intake.

One potent source of inflammation under direct dietary influence is the gut microbiota. The gut contains more than 1014 microorganisms, the majority of them bacteria, which can impact host physiology and behavior, including energy homeostasis [36]. There is accumulating evidence showing that HFD consumption leads to rapid unfavorable changes in microbiota composition (dysbiosis) [37,38]. HFD-driven dysbiosis is characterized by a decrease in bacterial diversity, a higher ratio between the dominant phyla, Firmicutes and Bacteroides [39], and an increase in the inflammatory potential of the microbiota [40]. Colonization of GF or microbiota-depleted animals with microbiota from obese donors leads to excessive weight gain [41], highlighting a triggering role for the microbiota in obesity. The gut microbiota may affect the vagal afferent pathway. Vagal nerve terminals located in the gastrointestinal (GI) lamina propria are ideally positioned to respond to changes in microbiota composition and/or the release of bacterial byproducts [42], and VAN express receptors for bacterial byproducts [43], [44], [45]. There is evidence that the GI bacterial population communicates with the CNS via a vagal pathway as the ability of the probiotic Lactobacillus rhamonus to protect against stress-induced anxiety is abolished by vagotomy [46]. Neurons and microglia alike can sense bacterial products. In the CNS, microglia alter synaptic function and axonal growth in response to pro-inflammatory bacterial compounds [47], [48], [49]. We have previously found that chronic administration of an obesity-associated pro-inflammatory bacterial factor, lipopolysaccharide (LPS), induces vagal leptin resistance, impairs CCK-induced satiety, and promotes intake and excessive weight gain in rats [45]. Conversely, preventing HFD-associated dysbiosis by antibiotic treatment [25] or prebiotic supplementation [35] prevents diet-associated changes in IB4 staining of the NTS [25]. Prebiotic administration in HFD fed rats reduces intake and preserves CCK-induced satiety [35].

While there is some evidence that dysbiosis is necessary for HFD-driven alteration in vagal structure and function, in this study we aimed to determine if dysbiosis was sufficient to induce vagal inflammation and alter vagal innervation to the NTS. To isolate the microbiota-driven effects from diet and establish a causal relationship between an obesity-associated dysbiotic microbiota and vagal remodeling, we used two different models of microbiota manipulation: 1) conventionally raised, immuno-competent, male Fischer and Wistar rats with microbiota depleted using antibiotic treatment and 2) immuno-deficient and microbiota-deficient GF male Fischer rats. Microbiota-depleted or GF rats were recolonized or conventionalized, respectively, with microbiota from low fat diet (LFD) fed (ConvLF group) or HFD fed donor rats (ConvHF group). We found that bacteria characteristic of each dietary treatment were successfully transferred to receiver animals. Additionally, colonization with a dysbiotic microbiota led to a significant increase in Iba1+ cells in both the NG and NTS and was associated with a decrease in NTS IB4 staining compared to colonization with a normal microbiota. These changes were associated with a loss in exogenous CCK-induced satiety. Finally, ConvHF animals displayed significant increases in food intake and BW compared to ConvLF rats.

Section snippets

Experiment 1

Eight-week-old Fischer rats (187 ± 5 g, Envigo, Indianapolis, IN) were single-housed in shoebox cages in a temperature-controlled animal facility with a 12/12 h light-dark cycle. Following three days of acclimation to the new facility, animals were divided into HFD fed donors (HFD, n=4), LFD fed donors (LFD, n=4) and an antibiotic-treated group (n=11). The antibiotic-treated group was further split into three groups: antibiotic controls (CtrlA, n=3), recolonized with HFD-type microbiota

Antibiotic depletion model (experiment 1)

Antibiotic treatment reduced GI bacterial load by 86% (Fig. 2). Fecal bacterial DNA concentration was significantly decreased following antibiotic administration (252.8 ± 25.38 vs. 35.92 ± 5.02 ng/µl, T(10)=2.228, p<0.0001. The same antibiotic depletion protocol appears to be slightly more effective in smaller animals as a 90–95% decrease in fecal bacterial DNA has previously been reported in mice [38]. While antibiotic administration was successful at reducing bacterial presence in the GI

Conclusion

Microbiota composition changes rapidly with HFD [79,80], and a pro-inflammatory-type microbiota is necessary and sufficient to cause weight gain [41,88,89]. We have previously demonstrated that pro-inflammatory bacterial signals administered chronically can alter vagal signaling and promote overeating [45]. In this study, using microbiota-depleted rats, we showed that microbiota composition modulates Iba1+ cell activation along the gut-brain axis and that a HFD-type microbiota promote NTS

Sources of financial support

This project was funded by the National Institutes of Health (NIH) grant no. R01DC01390, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) grant no. 1021RR581526.

Declaration of Competing Interest

The authors declare no potential conflict of interest.

Acknowledgements

The authors would like to acknowledge Dr. Jesse Thomas for his help with the microbiome data analysis. C.D.L.S and G.D.L designed the research; J.S.K., R.K., S.H.L, C.C., K.W.R, and D.M.W. conducted the research; J.S.K. and C.D.L.S analyzed data; J.S.K and C.D.L.S. wrote the manuscript; and C.D.L.S. had primary responsibilities for the final manuscript.

References (89)

  • T.M.Z. Waise

    One-day high-fat diet induces inflammation in the nodose ganglion and hypothalamus of mice

    Biochem. Biophys. Res. Commun.

    (2015)
  • S.M. Hermes et al.

    Localization of TRPV1 and P2X3 in unmyelinated and myelinated vagal afferents in the rat

    J. Chem. Neuroanat.

    (2016)
  • C.B. de La Serre et al.

    Chronic exposure to Low dose bacterial lipopolysaccharide inhibits leptin signaling in vagal afferent neurons

    Physiol. Behav.

    (2015)
  • A.J. Bruce-Keller

    Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity

    Biol. Psychiatry

    (2015)
  • S.J. Melhorn

    Acute exposure to a high-fat diet alters meal patterns and body composition

    Physiol. Behav.

    (2010)
  • M. Hatori

    Time restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high fat diet

    Cell Metab.

    (2012)
  • A. Kohsaka

    High-fat diet disrupts behavioral and molecular circadian rhythms in Mice

    Cell Metab.

    (2007)
  • M.K. Olsen

    Time-restricted feeding on weekdays restricts weight gain: A study using rat models of high-fat diet-induced obesity

    Physiol. Behav.

    (2017)
  • G. Singhal

    Effects of aging on the motor, cognitive and affective behaviors, neuroimmune responses and hippocampal gene expression

    Behav. Brain Res.

    (2020)
  • D. Ito

    Microglia-specific localisation of a novel calcium binding protein, Iba1

    Brain Res. Mol. Brain Res.

    (1998)
  • M. Hanani

    Satellite glial cells in sensory ganglia: from form to function

    Brain Res. Brain Res. Rev.

    (2005)
  • K.E. Miller et al.

    Glutamine-, gutamine synthetase-, glutamate dehydrogenase- and pyruvate carboxylase-immunoreactivites in the rat dorsal root ganglion and peripheral nerve

    Brain Res.

    (2002)
  • Z.R. Gallaher

    Changes in microglial activation within the hindbrain, nodose ganglia, and the spinal cord following subdiaphragmatic vagotomy

    Neurosci. Lett.

    (2012)
  • D. Fritze

    TNFα causes thrombin-dependent vagal neuron apoptosis in inflammatory bowel disease

    J. Gastrointest. Surg.

    (2014)
  • OECD, Health at a glance 2017: OECD indicators....
  • A.J. McCullough

    Epidemiology of the metabolic syndrome in the USA

    J. Dig. Dis.

    (2011)
  • E.A. Finkelstein

    Annual medical spending attributable to obesity: payer-and service-specific estimates

    Health Aff. (Millwood)

    (2009)
  • S. Hu

    Dietary fat, but not protein or carbohydrate, regulates energy intake and causes adiposity in Mice

    Cell Metab.

    (2018)
  • S.C. Woods

    Gastrointestinal satiety signals I. An overview of gastrointestinal signals that influence food intake

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2004)
  • Moran, T., Neural and Hormonal Controls of Food Intake and Satiety in Physiology of the Gastrointestinal Tract, Fourth...
  • M. Covasa et al.

    Adaptation to high-fat diet reduces inhibition of gastric emptying by CCK and intestinal oleate

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2000)
  • G. de Lartigue

    Leptin resistance in vagal afferent neurons inhibits cholecystokinin signaling and satiation in diet induced obese rats

    PLoS ONE

    (2012)
  • M. Covasa et al.

    Diminished satiation in rats exposed to elevated levels of endogenous or exogenous cholecystokinin

    Am. J. Physiol. Regul. Integr. Comp. Physiol.

    (2001)
  • F.A. Duca

    Decreased intestinal nutrient response in diet-induced obese rats: role of gut peptides and nutrient receptors

    Int. J. Obes. (Lond)

    (2013)
  • F.A. Duca

    Increased oral detection, but decreased intestinal signaling for fats in mice lacking gut microbiota

    PLoS ONE

    (2012)
  • G. de Lartigue

    Diet-induced obesity leads to the development of leptin resistance in vagal afferent neurons

    Am. J. Physiol. Endocrinol. Metab.

    (2011)
  • H. Qin

    Molecular mechanism of lipopolysaccharide-induced SOCS-3 gene expression in macrophages and microglia

    J. Immunol.

    (2007)
  • A.C. Vaughn

    Energy-dense diet triggers changes in gut microbiota, reorganization of gut-brain vagal communication and increases body fat accumulation

    Acta Neurobiol. Exp. (Wars)

    (2017)
  • A. Aguzzi et al.

    Microglia: scapegoat, saboteur, or something else?

    Science

    (2013)
  • V.H. Perry et al.

    Microglia in neurodegenerative disease

    Nat. Rev. Neurol.

    (2010)
  • H. Kettenmann

    Physiology of microglia

    Physiol. Rev.

    (2011)
  • K. Kingwell

    Neurodegenerative disease: Microglia in early disease stages

    Nat. Rev. Neurol.

    (2012)
  • C.T. De Souza

    Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus

    Endocrinology

    (2005)
  • D. Cai et al.

    Hypothalamic inflammation: a double-edged sword to nutritional diseases

    Ann. N. Y. Acad. Sci.

    (2011)
  • Cited by (27)

    • Emotional-Single Prolonged Stress: A promising model to illustrate the gut-brain interaction

      2023, Physiology and Behavior
      Citation Excerpt :

      The gut/brain axis is emerging as a fundamental channel for the transfer of neural signals that inform the brain about the metabolic and physiological state of the body [8]. Besides that, evidence suggests that changes in the commensal gut microbiota have been shown to influence the stress response [9], since dysbiosis can induce impairment of the intestinal barrier and thus promoting an increased translocation of pathogenic bacteria from the intestinal mucosa to the systemic circulation [10,11], resulting in systemic inflammation, characterized by the activation of innate immunity and abnormal bowel function [10,12]. These mechanisms can lead to damage to the CNS function, favoring the development of psychiatric diseases, such as depression and anxiety, highlighting the occurrence of brain-intestine interaction [13].

    • The microbiota-gut-brain axis in pathogenesis of depression: A narrative review

      2023, Physiology and Behavior
      Citation Excerpt :

      Subsequent studies further elucidated the underlying mechanism. Kim et al. have found that pro-inflammatory bacterial signals is sufficient to induce vagal neuronal apoptosis and alter synaptic function [93]. Lactobacillus was reported to regulate the depressive-like behavior and central GABA system via vagus nerves [73].

    • Do common antibiotic treatments influence emotional processing?

      2022, Physiology and Behavior
      Citation Excerpt :

      Resting heart rate is estimated to be 80% attributable to the parasympathetic nervous system (which consists primarily of the vagus nerve) and 20% to the sympathetic nervous system [69]. Since the vagus nerve acts to lower heart rate, one possible explanation is a reduction in vagal activity in the antibiotic group since disruption of the gut microbiota may result in reduced stimulation of the vagus nerve [70] and animal studies have shown that a dysbiotic microbiota can lead to a decrease in vagal innervation [71]. While we did not find a difference in heart rate variability, this does not necessarily mean there were no differences in vagal activity [66–68].

    • Loss of vagal integrity disrupts immune components of the microbiota-gut-brain axis and inhibits the effect of Lactobacillus rhamnosus on behavior and the corticosterone stress response

      2021, Neuropharmacology
      Citation Excerpt :

      Indeed, this IPAN to vagus sensory synapse has been identified as playing a critical role in the rapid activation of vagal afferent fibers following introduction of JB-1 to the gut lumen (Perez-Burgos et al., 2013) that in turn leads to activation of neurons in widespread brain regions (Bharwani et al., 2020). In light of the role for vagal afferents in transmitting sensory information from the gut to the brain, observations that vagotomy prevents the effect of JB-1 (Bravo et al., 2011), and other microbes (Bercik et al., 2011; Malick et al., 2015) on behavior are generally interpreted as evidence that the vagus nerve acts as a “direct line” between the intestinal bacteria and neural circuitry influencing anxiety and depression (Kim et al., 2020). However, the immune system also plays a critical role in mediating the antidepressant and anxiolytic-like effects of JB-1.

    View all citing articles on Scopus
    View full text