ArticlePDF Available

Systemic LPS administration induces brain inflammation but not dopaminergic neuronal death in the substantia nigra

Authors:

Abstract and Figures

It has been suggested that brain inflammation is important in aggravation of brain damage and/or that inflammation causes neurodegenerative diseases including Parkinson's disease (PD). Recently, systemic inflammation has also emerged as a risk factor for PD. In the present study, we evaluated how systemic inflammation induced by intravenous (iv) lipopolysaccharides (LPS) injection affected brain inflammation and neuronal damage in the rat. Interestingly, almost all brain inflammatory responses, including morphological activation of microglia, neutrophil infiltration, and mRNA/protein expression of inflammatory mediators, appeared within 4-8 h, and subsided within 1-3 days, in the substantia nigra (SN), where dopaminergic neurons are located. More importantly, however, dopaminergic neuronal loss was not detectable for up to 8 d after iv LPS injection. Together, these results indicate that acute induction of systemic inflammation causes brain inflammation, but this is not sufficiently toxic to induce neuronal injury.
Content may be subject to copyright.
A preview of the PDF is not available
... However, the level of inflammation that is induced in sepsis models is not representative of the chronic low-level inflammation that occurs with age and likely contributes to age-related neurological disease. Previous studies have also demonstrated that neutrophils adhere to the vasculature, infiltrate the brain parenchyma, and can return to the blood stream after interaction with microglia following acute systemic LPS exposure in mice and rats [28,29]. However, no studies have investigated chronic peripheral inflammation and subsequent neutrophil activation in the brain and periphery. ...
Article
Full-text available
Lipopolysaccharide-induced (LPS) inflammation is used as model to understand the role of inflammation in brain diseases. However, no studies have assessed the ability of peripheral low-level chronic LPS to induce neutrophil activation in the periphery and brain. Subclinical levels of LPS were injected intraperitoneally into mice to investigate its impacts on neutrophil frequency and activation. Neutrophil activation, as measured by CD11b expression, was higher in LPS-injected mice compared to saline-injected mice after 4 weeks but not 8 weeks of injections. Neutrophil frequency and activation increased in the periphery 4–12 h and 4–8 h after the fourth and final injection, respectively. Increased levels of G-CSF, TNFa, IL-6, and CXCL2 were observed in the plasma along with increased neutrophil elastase, a marker of neutrophil extracellular traps, peaking 4 h following the final injection. Neutrophil activation was increased in the brain of LPS-injected mice when compared to saline-injected mice 4–8 h after the final injection. These results indicate that subclinical levels of peripheral LPS induces neutrophil activation in the periphery and brain. This model of chronic low-level systemic inflammation could be used to understand how neutrophils may act as mediators of the periphery–brain axis of inflammation with age and/or in mouse models of neurodegenerative or neuroinflammatory disease.
... In the peripheral LPS model, we observed the tra cking of neutrophils to the brain, which is a documented effect of peripheral endotoxin administration. The in ux of neutrophils due to peripheral LPS exposure has been con rmed by others using immunohistochemical approaches 45 and ow cytometry techniques 18,46 . In vivo two-photon imaging has revealed physical contact between microglia and neutrophils in the brain of peripheral LPS-treated mice 47 , consistent with the heightened communication between microglia and neutrophils we report here, especially the increased expression of adhesion molecules, such as Icam1 and Msn respectively, on these two cell types. ...
Preprint
Full-text available
Inflammation is a realized detriment to brain health in a growing number of neurological diseases, but querying neuroinflammation in its cellular complexity remains a challenge. This manuscript aims to provide a reliable and accessible strategy for examining the brain’s immune system. We compare the efficacy of cell isolation methods in producing ample and pure immune samples from mouse brains. Then, with the high-input single-cell genomics platform PIPseq, we generate a rich neuroimmune dataset containing microglia and many peripheral immune populations. To demonstrate this strategy’s utility, we interrogate the well-established model of LPS-induced neuroinflammation with single-cell resolution. We demonstrate the activation of crosstalk between microglia and peripheral phagocytes and highlight the unique contributions of microglia and peripheral immune cells to neuroinflammation. Our approach enables the high-depth evaluation of inflammation in longstanding rodent models of neurological disease to reveal novel insight into the contributions of the immune system to brain health.
... It could also affect the dopaminergic neurotransmission in experimental studies on Parkinsonism Duty and Jenner, 2011;Felger and Miller, 2012). Intracranial / intraperitoneal injection of lipopolysaccharide that activates immune cells and enhances inflammation has been found to damage dopaminergic neurons in the nigrostriatal region (He et al., 2013;Jeong et al., 2010). Activation of TLRs and NF-kβ pathway has also been observed in mouse model of PD (Noelker et al., 2013). ...
Article
The risk to develop neurobehavioural abnormalities in humans on exposure to lambda-cyhalothrin (LCT) - a type II synthetic pyrethroid has enhanced significantly due to its extensive uses in agriculture, households, veterinary practices and public health programs. Earlier we found that the brain dopaminergic system is vulnerable to LCT and affects motor functions in rats. In continuation to this, the present study is focused to unravel the role of neuroinflammationin LCT-induced neurotoxicity in substantia nigra and corpus striatum in rats.Increase in the mRNA expression of proinflammatory cytokines (TNF- α, IL-1β, IL-6) and iNOS whereas a decrease in anti-inflammatory cytokine (IL-10)was distinct both in substantia nigra and corpus striatum of rats treated with LCT (0.5, 1.0, 3.0mg/kg body weight, p.o, for 45 days) as compared to control rats. Further, LCT-treated rats exhibited increasedlevels of glial fibrillary acidic protein (GFAP)and ionized calcium-binding adapter molecule 1 (Iba-1), the glial marker proteins both in substantia nigra and corpus striatum as compared to controls. Exposure of rats to LCT also caused alteration in the levelsof heat shock protein 60 (HSP60) and mRNA expression of toll-like receptors(TLR2 and TLR4) in the substantia nigra and corpus striatum.An increase in the phosphorylation of key proteins involved in NF-kβ (P65, Iκβ, IKKα, IKKβ)signalingand STAT1 and STAT3 and alteration in the protein levels of JAK1 and JAK2 were prominent in LCT-treated rats.Histological studies revealed damage of dopaminergic neurons and reactive gliosis as evidenced by the presence of darkly stained pyknotic neurons anddecrease in Nisslsubstance and an increase in infiltration of immune cells both in substantia nigra and corpus striatum of LCT-treated rats. The results exhibit that LCT may damage dopaminergic neurons in the substantia nigra and corpus striatum by inducing inflammation as a result of stimulation of neuroglial cells involving activation of NF-κβ and JAK/STAT signaling.
... Our results showed that gene responses that are important parts of these pathways, which are crucial for the response to LPS, were dampened by the presence of the novel female in two or more brain regions. In rat and mice, systemic LPS administration leads to neuroinflammation, including expression of cytokines, activated microglia (the brain's resident macrophages -the main inflammatory cells in the brain) and neutrophil infiltration (57)(58)(59). ...
Article
Sick animals display drastic changes in their behavioral patterns, including decreased activity, decreased food and water intake, and decreased interest in social interactions. These behaviors, collectively called "sickness behaviors", can be socially modulated. For example, when provided with mating opportunities, males of several species show reduced sickness behaviors. While the behavior is known to change, how the social environment affects neural molecular responses to sickness is not known. Here, we used a species, the zebra finch, Taeniopygia guttata, where males have been shown to decrease sickness behaviors when presented with novel females. Using this paradigm, we obtained samples from three brain regions (the hypothalamus, the bed nucleus of the stria terminalis, and the nucleus taeniae) from lipopolysaccharide (LPS) or control treated males housed under four different social environments. Manipulation of the social environment rapidly changed the strength and co-expression patterns of the neural molecular responses to the immune challenge in all brain regions tested, therefore suggesting that the social environment plays a significant role in determining the neural responses to an infection. In particular, brains of males paired with a novel female showed muted immune responses to LPS, as well as altered synaptic signaling. Neural metabolic activity in response to the LPS challenge was also affected by the social environment. Our results provide new insights into the effects of the social environment on brain responses to an infection, thereby improving our understanding of how the social environment can affect health.
... Mechanisms underlying LPS-induced cognitive impairments, such as inflammatory cascade induction, are likely manifold and are actively being explored. Indeed, LPS exposure is known to cause generalized inflammation in brain areas critically involved in learning and memory (Jeong et al., 2010;Liu et al., 2018). Increased protein and gene expression levels of inflammatory cytokines, mainly IL-1β and IL-6, TLR2 and 4 mRNA, and also GFAP and Iba-1 (activated microglia) have been consistently reported in cognitive brain regions (Kranjac et al., 2012;Liu et al., 2018;Noh et al., 2014;Zhao et al., 2019) While some studies note persistent inflammation when LPS is administered repeatedly over a short timespan, such as elevated levels of IL-1β and TNF-α two weeks post-LPS withdrawal in repeatedly exposed mice (2.5 mg/kg/day for 7 days) , inflammatory consequences in an intermittent inflammation model, like the one we used here, are not yet well studied. ...
Article
Full-text available
Age-related cognitive decline, a common component of the brain aging process, is associated with significant impairment in daily functioning and quality of life among geriatric adults. While the complexity of mechanisms underlying cognitive aging are still being elucidated, microbial exposure and the multifactorial inflammatory cascades associated with systemic infections are emerging as potential drivers of neurological senescence. The negative cognitive and neurobiological consequences of a single pathogen-associated inflammatory experience, such as that modeled through treatment with lipopolysaccharide (LPS), are well documented. Yet, the brain aging impacts of repeated, intermittent inflammatory challenges are less well studied. To extend the emerging literature assessing the impact of infection burden on cognitive function among normally aging mice, here, we repeatedly exposed adult mice to intermittent LPS challenges during the aging period. Male 10-month-old C57BL6 mice were systemically administered escalating doses of LPS once every two weeks for 2.5 months. We evaluated cognitive consequences using the non-spatial step-through inhibitory avoidance task, and both spatial working and reference memory versions of the Morris water maze. We also probed several potential mechanisms, including cortical and hippocampal cytokine/chemokine gene expression, as well as hippocampal neuronal function via extracellular field potential recordings. Though there was limited evidence for an ongoing inflammatory state in cortex and hippocampus, we observed impaired learning and memory and a disruption of hippocampal long-term potentiation. These data suggest that a history of intermittent exposure to LPS-induced inflammation is associated with subtle but significantly impaired cognition among normally aging mice. The broader impact of these findings may have important implications for standard of care involving infections in aging individuals or populations at-risk for dementia.
... In the current study, the largest induction of inflammatory cytokines occurred only 2 h after LPS administration; therefore, this reaction occurred within hours and was mainly caused by CNS residual microglia. On the other hand, peripheral inflammatory cells including monocyte-derived macrophage and neutrophils are reported to infiltrate into the CNS 24 h after LPS treatment [26,27]. It is possible that peripheral cells are involved in the inflammatory changes that occurred afterward in the spinal cord. ...
Article
Full-text available
Septic patients commonly present with central nervous system (CNS) disorders including impaired consciousness and delirium. Today, the main mechanism regulating sepsis-induced cerebral disorders is believed to be neuroinflammation. However, it is unknown how another component of the CNS, the spinal cord, is influenced during sepsis. In the present study, we intraperitoneally injected mice with lipopolysaccharide (LPS) to investigate molecular and immunohistochemical changes in the spinal cord of a sepsis model. After LPS administration in the spinal cord, pro-inflammatory cytokines including interleukin (IL)-1β, IL-6, and tumor necrosis factor alpha mRNA were rapidly and drastically induced. Twenty-four-hour after the LPS injection, severe neuronal ischemic damage spread into gray matter, especially around the anterior horns, and the anterior column had global edematous changes. Immunostaining analyses showed that spinal microglia were significantly activated and increased, but astrocytes did not show significant change. The current results indicate that sepsis induces acute neuroinflammation, including microglial activation and pro-inflammatory cytokine upregulation in the spinal cord, causing drastic neuronal ischemia and white matter edema in the spinal cord.
... To validate the model, we first used an established rat paradigm of inflammation based on intracerebral lipopolysaccharide (LPS) administration (16). In this paradigm, neuronal viability and morphology are preserved, while inducing microglial activation within a few hours, and a delayed astrocytic response that is detectable only 24 hours after injection (17). Therefore, glial responses can be transiently dissociated from neuronal degeneration, and the signature of reactive microglia investigated independently of any astrogliosis. ...
Article
Full-text available
While glia are increasingly implicated in the pathophysiology of psychiatric and neurodegenerative disorders, available methods for imaging these cells in vivo involve either invasive procedures or positron emission tomography radiotracers, which afford low resolution and specificity. Here, we present a noninvasive diffusion-weighted magnetic resonance imaging (MRI) method to image changes in glia morphology. Using rat models of neuroinflammation, degeneration, and demyelination, we demonstrate that diffusion-weighted MRI carries a fingerprint of microglia and astrocyte activation and that specific signatures from each population can be quantified noninvasively. The method is sensitive to changes in glia morphology and proliferation, providing a quantitative account of neuroinflammation, regardless of the existence of a concomitant neuronal loss or demyelinating injury. We prove the translational value of the approach showing significant associations between MRI and histological microglia markers in humans. This framework holds the potential to transform basic and clinical research by clarifying the role of inflammation in health and disease.
Preprint
Full-text available
Inflammation is a realized detriment to brain health in a growing number of neurological diseases, but querying neuroinflammation in its cellular complexity remains a challenge. This manuscript aims to provide a reliable and accessible strategy for examining the brain's immune system. We compare the efficacy of cell isolation methods in producing ample and pure immune samples from mouse brains. Then, with the high-input single-cell genomics platform PIPseq, we generate a rich neuroimmune dataset containing microglia and many peripheral immune populations. To demonstrate this strategy's utility, we interrogate the well-established model of LPS-induced neuroinflammation with single-cell resolution. We demonstrate the activation of crosstalk between microglia and peripheral phagocytes and highlight the unique contributions of microglia and peripheral immune cells to neuroinflammation. Our approach enables the high-depth evaluation of inflammation in longstanding rodent models of neurological disease to reveal novel insight into the contributions of the immune system to brain health.
Article
Neuroinflammation leads to a persistent oxidative stress in the brain, and is closely related to the pathology of various neurological disorders. Hypochlorous acid (HClO) is a reactive oxygen species (ROS) that, at high levels, can cause brain tissue damage and neurogenic apoptosis. Herein, we designed and synthesized a silicon-rhodamine (SiR)-based formohydrazide (FH)-containing fluorescent probe, denoted as SiR-FH, for sensing HClO. This probe showed good selectivity, rapid response and high sensitivity. SiR-FH was successfully used to detect endogenous and exogenous HClO in living cells. Moreover, SiR-FH realized real-time monitoring of change in HClO flux in the brains of mice with LPS-induced neuroinflammation. The probe provides a practical tool for the monitoring of oxidative stress related to neuroinflammation.
Article
Full-text available
Lipopolysaccharide (LPS) proved to be an important tool, not only in the induction of neuroinflammatory models, but also in demonstrating the behavioral and cognitive consequences of endotoxemia. Curcumin, in its native form, has proven to be a worthy candidate for further development as it protects the dopaminergic neurons against LPS-induced neurotoxicity. However, it remains hindered by its poor bioavailability. In this study we aim to explore the possible molecular mechanism of LPS-induced neurotoxicity and the possible protective effects of orally supplemented nanocurcumin. Thirty-six adult male Wistar rats weighing 170–175 g were divided into six groups and treated with single I.P. (intra-peritoneal) dose of LPS (sigma and extracted; separately) (5 mg/kg BW) plus daily oral nanocurcumin (15 mg/kg BW). The rats were followed for 7 days after the LPS injection and nanocurcumin supplementations daily via oral gavage. After scarification, the levels of neurotransmitters, antioxidants, and amyloidogenesis markers were assessed in brain tissues. Nanocurcumin showed adequate antioxidant and neuroprotective effects, rescuing the rats which had been injected intraperitoneally with LPS endotoxin.
Article
Full-text available
Alzheimer’s disease is the most common subtype of dementia. This disease is diagnosed in approximately two thirds of all cases of dementia (1). According to current diagnostic criteria, a diagnosis of Alzheimer’s disease is considered probable when alternative causes of dementia have been excluded (2, 3). A clinical diagnosis of dementia is often made according to the Diagnostic and Statistical Manual of Mental Disorders, Third Edition, Revised (DSM-III-R) criteria for dementia, with a subdiagnosis of Alzheimer’s disease made according to the National Institute of Neurological and Communicative Disorders and Stroke–Alzheimer’s Disease and Related Disorders Association (NINCDS-ADRDA) criteria (2). The pathogenesis of Alzheimer’s disease is largely unknown. In short, the most frequently used explanation is that unknown genetic or environmental factors initiate a cascade of neuropathologic events that feature accumulation of β-amyloid and neurofibrillary tangles. This process is clinically characterized by a long latent phase, then a prodromal stage with a gradual and progressive decline in long-term episodic memory and impairment of other cognitive domains of mental functioning (4). Eventually, the person crosses a threshold of cognitive loss, after which the full syndrome is evident (5).
Article
It is well known that systemic infections cause flare-ups of disease in individuals with asthma and rheumatoid arthritis, and that relapses in multiple sclerosis can often be associated with upper respiratory-tract infections. Here we review evidence to support our hypothesis that in chronic neurodegenerative diseases such as Alzheimer's disease, with an ongoing innate immune response in the brain, systemic infections and inflammation can cause acute exacerbations of symptoms and drive the progression of neurodegeneration.
Article
The clinical diagnosis of Parkinson disease (PD) is based on the identification of some combination of the cardinal motor signs of bradykinesia, rigidity, tremor, and postural instability, but few attempts have been made to develop explicit diagnostic criteria. We propose a clinical diagnostic classification based on a comprehensive review of the literature regarding the sensitivity and specificity of the characteristic clinical features of PD. Three levels of diagnostic confidence are differentiated: Definite, Probable, and Possible. The diagnoses of Possible and Probable PD are based on clinical criteria alone. Neuropathologic confirmation is required for the diagnosis of Definite PD in patients with the clinical diagnosis of Possible or Probable PD. Criteria for histopathologic confirmation of PD are also presented.
Article
Interleukin-1β (IL-1β), interleukin-2 (IL-2), and interleukin-6 (IL-6) were measured in the cerebrospinal fluid (CSF) and plasma of 12 control subjects, 11 sporadic Alzheimer's disease (AD) and 22 de novo Parkinson's disease (PD) patients using high sensitivity enzyme-linked immunosorbent assays (ELISA). IL-1β and IL-6 contents were significantly elevated in the CSF of de novo PD and AD patients in comparison to the control group. In contrast, the plasma levels were not significantly affected. IL-2 contents in the CSF and plasma samples were unchanged in the three groups compared. Because the two cytokines IL-1β and IL-6 are known to play a key role in the interaction between the nervous and immune system, e.g. in the so-called acute phase response, our results support the involvement of immunological events in the complex process of neurodegeneration in AD and PD.
Article
Introduction - Several lines of evidence suggest that neuroimmune mechanisms may also be involved in neurodegeneration in Parkinson's disease (PD). The potential role of cytokines such as interleukin 6 (IL-6), in the interaction between neurons and immune system has been emphasized by recent findings. IL-6 induces acute phase protein synthesis, differentiation of neuronal cells and improves catecholaminergic and cholinergic cell survival in the brain. Subjects and methods - We determined levels of IL-6 in cerebrospinal fluid (CSF) of untreated parkinsonian patients and age- and sex-matched controls. Intensity of disease was evaluated by the Unified Parkinson's Disease Rating scale. Results - Significantly elevated levels of IL-6 were found in the CSF of parkinsonian patients. Moreover a significant inverse correlation between severity of PD and IL-6 CSF levels appeared. Discussion - Elevated IL-6 levels in the CSF of untreated parkinsonian patients may reflect the original condition in the course of disease. We speculate that an endogenous upregulation of IL-6 synthesis occurs in order to regenerate lesioned neurons probably at an early phase of the degenerative process in PD.
Article
Interleukin-1 (IL-1) is an important mediator of immunoinflammatory responses in the brain. In the present study, we examined whether prostaglandin E2 (PGE2) production after IL-1β stimulation is dependent upon activation of protein kinases in astroglial cells. Astrocyte cultures stimulated with IL-1β or the phorbol ester, PMA significantly increased PGE2 secretion. The stimulatory action of IL-1β on PGE2 production was totally abolished by NS-398, a specific inhibitor of cyclo-oxygenase-2 activity, as well as by the protein synthesis inhibitor cycloheximide, and the glucocorticoid dexamethasone. Furthermore, IL-1β induced the expression of COX-2 mRNA. This occurred early at 2 h, with a maximum at 4 h and declined at 12 h. IL-1 β treatment also induced the expression of COX-2 protein as determined by immunoblot analysis. In that case the expression of the protein remained high at least up to 12 h. Treatment of cells with protein kinase C inhibitors (H-7, bisindolylmaleimide and calphostin C) inhibited IL-1β stimulation of PGE2. In addition, PKC-depleted astrocyte cultures by overnight treatment with PMA no longer responded to PMA or IL-1. The ablation of the effects of PMA and IL-1β on PGE2 production, likely results from down-regulation of phorbol ester sensitive-PKC isoenzymes. Immunoblot analysis demonstrated the translocation of the conventional isoform cPKC-α from cytosol to membrane following treatment with IL-1β. In addition, IL-1β treatment led to activation of extracellular signal-regulated kinase (ERK1/2) and p38 subgroups of MAP kinases in astroglial cells. Interestingly, the inhibition of ERK kinase with PD 98059, as well as the inhibition of p38 MAPK with SB 203580, prevented IL-1β-induced PGE2 release. ERK1/2 activation by IL-1β was sensitive to inhibition by the PKC inhibitor bisindolylmaleimide suggesting that ERK phosphorylation is a downstream signal of PKC activation. These results suggest key roles for PKC as well as for ERK1/2 and p38 MAP kinase cascades in the biosynthesis of PGE2, likely by regulating the induction of cyclo-oxygenase-2, in IL-1β-stimulated astroglial cells. British Journal of Pharmacology (2000) 131, 152–159; doi:10.1038/sj.bjp.0703557