Elsevier

Neurobiology of Aging

Volume 49, January 2017, Pages 60-68
Neurobiology of Aging

Regular article
Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly

https://doi.org/10.1016/j.neurobiolaging.2016.08.019Get rights and content

Abstract

The pathway leading from amyloid-β deposition to cognitive impairment is believed to be a cornerstone of the pathogenesis of Alzheimer's disease (AD). However, what drives amyloid buildup in sporadic nongenetic cases of AD is still unknown. AD brains feature an inflammatory reaction around amyloid plaques, and a specific subset of the gut microbiota (GMB) may promote brain inflammation. We investigated the possible role of the GMB in AD pathogenesis by studying the association of brain amyloidosis with (1) GMB taxa with pro- and anti-inflammatory activity; and (2) peripheral inflammation in cognitively impaired patients. We measured the stool abundance of selected bacterial GMB taxa (Escherichia/Shigella, Pseudomonas aeruginosa, Eubacterium rectale, Eubacterium hallii, Faecalibacterium prausnitzii, and Bacteroides fragilis) and the blood expression levels of cytokines (pro-inflammatory cytokines: CXCL2, CXCL10, interleukin [IL]-1β, IL-6, IL-18, IL-8, inflammasome complex (NLRP3), tumor necrosis factor-alpha [TNF-α]; anti-inflammatory cytokines: IL-4, IL-10, IL-13) in cognitively impaired patients with (n = 40, Amy+) and with no brain amyloidosis (n = 33, Amy−) and also in a group of controls (n = 10, no brain amyloidosis and no cognitive impairment). Amy+ patients showed higher levels of pro-inflammatory cytokines (IL-6, CXCL2, NLRP3, and IL-1β) compared with both controls and with Amy− patients. A reduction of the anti-inflammatory cytokine IL-10 was observed in Amy+ versus Amy−. Amy+ showed lower abundance of E. rectale and higher abundance of Escherichia/Shigella compared with both healthy controls (fold change, FC = −9.6, p < 0.001 and FC = +12.8, p < 0.001, respectively) and to Amy− (FC = −7.7, p < 0.001 and FC = +7.4, p = 0.003). A positive correlation was observed between pro-inflammatory cytokines IL-1β, NLRP3, and CXCL2 with abundance of the inflammatory bacteria taxon Escherichia/Shigella (rho = 0.60, p < 0.001; rho = 0.57, p < 0.001; and rho = 0.30, p = 0.007, respectively) and a negative correlation with the anti-inflammatory E. rectale (rho = −0.48, p < 0.001; rho = −0.25, p = 0.024; rho = −0.49, p < 0.001). Our data indicate that an increase in the abundance of a pro-inflammatory GMB taxon, Escherichia/Shigella, and a reduction in the abundance of an anti-inflammatory taxon, E. rectale, are possibly associated with a peripheral inflammatory state in patients with cognitive impairment and brain amyloidosis. A possible causal relation between GMB-related inflammation and amyloidosis deserves further investigation.

Introduction

Neurodegenerative disorders, including Alzheimer's disease (AD), are characterized by the accumulation of neurotoxic proteins in the brain. In AD, these are amyloid-β (Aβ) and hyperphosphorylated tau, representing the major components of extracellular senile plaques and intracellular neurofibrillary tangles, respectively. The common feature of these proteins is the loss of their physiologic activity and the gain of toxic properties, promoting neurodegeneration. Aβ is widely believed to be the key in AD pathophysiology (Jack et al., 2013). In nongenetic cases of AD, the pathophysiological mechanisms of Aβ deposition and the ensuing neurodegeneration and cognitive symptoms remain to be elucidated, but neuroinflammation seems to play a key role (Heppner et al., 2015). Indeed, in addition to plaques and tangles, AD patients feature central inflammation, mediated by activated microglia, reactive astrocytes, and complement activation, that have been especially observed in the vicinity of amyloid plaques and even in the early stages of AD (Clark and Vissel, 2015, Heneka et al., 2015, Heppner et al., 2015, Latta et al., 2015, Stoeck et al., 2014). Enhanced inflammation occurs also in body fluids of AD patients, such as cerebrospinal fluid and blood (Kauwe et al., 2014, Monson et al., 2014, Nascimento et al., 2014, Vom Berg et al., 2012).

The increased interest in the complex network of inflammatory mediators and the immune system has allowed to identify a growing number of pro-inflammatory molecules involved in central nervous system disorders, such as interleukin (IL)-6, tumor necrosis factor-alpha (TNF-α), and the inflammasome complex (NLRP3). These have been found associated with cognitive impairment and AD pathology (Chen et al., 2015, Doecke et al., 2012, Leung et al., 2013, Ray et al., 2007, Soares et al., 2012b, Tan et al., 2013). However, the pathophysiological cascade linking inflammation with Aβ deposition is still unknown (Heppner et al., 2015). Some recent observations indicate that a specific subset of the gut microbiota (GMB) can drive neuroinflammation in rodents (Palm et al., 2015, Petra et al., 2015 Erny et al., 2015) and affect brain function and behavior in rodents and humans (Bercik et al., 2011, Diaz Heijtz et al., 2011, Li et al., 2009).

Alterations of GMB composition have been observed in multiple sclerosis (MS) and Parkinson's disease (PD), conditions also featuring neuroinflammation and protein misfolding. Indeed, the removal of GMB in animal models of multiple sclerosis prevents the development of relapsing-remitting demyelination (Berer et al., 2011) and oral ingestion of probiotics attenuates neuroinflammation (Luo et al., 2014, Toumi et al., 2014). In PD, the evidence is even stronger. The deposition of alpha (α)-synuclein, the underlying molecular pathology, has been found both in the digestive tract and enteric nervous system, already in the early phases of the disease (Del Tredici et al., 2010, Goedert et al., 2013, Lebouvier et al., 2010). Moreover, the gut mucosa of Parkinsonian patients shows increased permeability, signs of inflammation and invasion of coliform bacteria (Forsyth et al., 2011), and hosts a peculiar GMB composition, characterized by decreased abundance of Prevotellaceae and an increase in Enterobacteriaceae, which are also related to the severity of illness (Scheperjans et al., 2015). Importantly, enhanced inflammation, as a consequence of alterations in GMB composition, has been implicated in the initiation of α-synuclein misfolding (Olanow et al., 2014).

To our knowledge, no evidence of GMB alterations has been reported in AD patients yet; however, it has been recently suggested that bacterial endotoxins may play a key role in the inflammatory and pathologic processes associated with amyloidosis and AD (Asti and Gioglio, 2014, Vom Berg et al., 2012), as bacterial components, such as endotoxins, have been found within the typical senile plaque lesions of the AD brain (Asti and Gioglio, 2014, Schwartz, 2013).

The aim of this study was to test, in elder patients with cognitive impairment, the association between brain amyloidosis and (1) candidate GMB taxa with known inflammatory activity (pro-inflammatory: Escherichia/Shigella and Pseudomonas aeruginosa; anti-inflammatory: Eubacterium rectale, Eubacterium hallii, Faecalibacterium prausnitzii, and Bacteroides fragilis) (Bruzzese et al., 2014, Cantarel et al., 2015, De la Fuente et al., 2014, Friedland, 2015); and (2) peripheral inflammation markers implicated in the pathogenesis of AD (pro-inflammatory cytokines: CXCL2, CXCL10, IL-1β, IL-6, IL-18, IL-8, NLRP3, TNF-α; anti-inflammatory cytokines: IL-4, IL-10, IL-13) (Chen et al., 2015, Doecke et al., 2012, Leung et al., 2013, Ray et al., 2007, Soares et al., 2012b, Tan et al., 2013).

Section snippets

Study design and patients description

The patients have been recruited from a larger study in 18 memory clinics in Eastern Lombardy, Italy, aiming to assess the added value of amyloid imaging in the clinical work-up of patients with cognitive complaints (the Incremental Diagnostic Value of Florbetapir Amyloid Imaging [INDIA-FBP] study) (http://www.centroalzheimer.org/sito/ip_lilly.php). Patients coming to observation with cognitive impairment and AD as a possible etiology were offered, on top and at the end of their routine

Clinical sample description

The 3 groups were similar for age, gender, and BMI. Amy+ patients had lower cognitive performances than both Amy− and HC (Table 1). Both neurodegeneration in the medial temporal lobe and microvascular white matter changes were similar in Amy+ and Amy−. Indeed, Amy− and Amy+ patients did not show neither significant difference on medial temporal atrophy (Scheltens scale: mean ± standard deviation 1.8 ± 1.1 and 2.0 ± 0.8, p = 0.670) nor white matter changes (age-related white matter change scale:

Discussion

In the present study, we have investigated the association of brain amyloidosis with candidate GMB taxa, known to have inflammatory properties, and peripheral blood inflammation biomarkers. We found that subjects with cognitive impairment and brain amyloidosis had lower abundance of the anti-inflammatory E. rectale and higher abundance of the pro-inflammatory Escherichia/Shigella in their stools when they were compared to both a group of control subjects and also to a group of subjects with

Disclosure statement

Giovanni Frisoni has served in advisory boards for Roche, Lilly, BMS, Bayer, Lundbeck, Elan, Astra Zeneca, Pfizer, Taurx, Wyeth, GE, and Baxter. He received research grants from Wyeth International, Lilly International, Lundbeck Italia, GE International, Avid/Lilly, Roche, Piramal, and the Alzheimer's Association. In the last 2 years, he received speaker honoraria from Lundbeck, Piramal, GE, and Avid/Lilly; Marina Boccardi received a research grant from Piramal; Alessandro Padovani received

Acknowledgements

The INDIA-FBP study was funded, thanks to a grant by AVID-Radiopharmaceuticals. Dr Cattaneo received funding from ERANET Neuron (INFLAME-D) and from Ricerca Corrente (Ministry of Health). The authors thank all the patients and their families for their participation to the study.

References (63)

  • A.I. Petra et al.

    Gut-microbiota-brain axis and its effect on neuropsychiatric disorders with suspected immune dysregulation

    Clin. Ther.

    (2015)
  • S.E. Pryde et al.

    The microbiology of butyrate formation in the human colon

    FEMS Microbiol Lett

    (2002)
  • P. Scheltens et al.

    A semiquantative rating scale for the assessment of signal hyperintensities on magnetic resonance imaging

    J. Neurol. Sci.

    (1993)
  • A. Asti et al.

    Can a bacterial endotoxin be a key factor in the kinetics of amyloid fibril formation?

    J. Alzheimers Dis.

    (2014)
  • P. Bercik et al.

    The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice

    Gastroenterology

    (2011)
  • K. Berer et al.

    Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination

    Nature

    (2011)
  • E. Bruzzese et al.

    Disrupted intestinal microbiota and intestinal inflammation in children with cystic fibrosis and its restoration with Lactobacillus GG: a randomised clinical trial

    PLoS One

    (2014)
  • B.L. Cantarel et al.

    Gut microbiota in multiple sclerosis: possible influence of immunomodulators

    J. Investig. Med.

    (2015)
  • A. Cattaneo et al.

    Candidate genes expression profile associated with antidepressants response in the GENDEP study: differentiating between baseline ‘predictors’ and longitudinal ‘targets’

    Neuropsychopharmacology

    (2013)
  • E. Chorell et al.

    Bacterial chaperones CsgE and CsgC differentially modulate human α-synuclein amyloid formation via transient contacts

    PLoS One

    (2015)
  • I.A. Clark et al.

    Amyloid beta: one of three danger-associated molecules that are secondary inducers of the proinflammatory cytokines that mediate Alzheimer's disease

    Br. J. Pharmacol.

    (2015)
  • W.S. Cleveland et al.

    Local regression models

  • P.A. Cox et al.

    Diverse taxa of cyanobacteria produce beta-N-methylamino-L-alanine, a neurotoxic amino acid

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • J.F. Cryan et al.

    Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour

    Nat. Rev. Neurosci.

    (2012)
  • K. Del Tredici et al.

    Lewy pathology in the submandibular gland of individuals with incidental Lewy body disease and sporadic Parkinson's disease

    Acta Neuropathol.

    (2010)
  • R. Diaz Heijtz et al.

    Normal gut microbiota modulates brain development and behavior

    Proc. Natl. Acad. Sci. U. S. A.

    (2011)
  • J.D. Doecke et al.

    Blood-based protein biomarkers for diagnosis of Alzheimer disease

    Arch. Neurol.

    (2012)
  • D. Erny et al.

    Host microbiota constantly control maturation and function of microglia in the CNS

    Nat Neurosci

    (2015)
  • C.B. Forsyth et al.

    Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson's disease

    PLoS One

    (2011)
  • P. Forsythe et al.

    Voices from within: gut microbes and the CNS

    Cell Mol. Life Sci.

    (2013)
  • R.P. Friedland

    Mechanisms of molecular mimicry involving the microbiota in neurodegeneration

    J. Alzheimers Dis.

    (2015)
  • Cited by (854)

    View all citing articles on Scopus
    View full text