Elsevier

Brain, Behavior, and Immunity

Volume 44, February 2015, Pages 121-127
Brain, Behavior, and Immunity

Plasma inflammatory biomarkers for Huntington’s disease patients and mouse model

https://doi.org/10.1016/j.bbi.2014.09.011Get rights and content

Highlights

  • Increased plasma IL-6, MMP-9, VEGF, and TGF-β1 in HD patients.

  • Decreased IL-18 plasma levels in HD patients.

  • Plasma IL-6 reversely correlates with independence scale and functional capacity.

  • Alterations of the inflammatory markers were found in R6/2 mice at different ages.

  • These inflammatory markers may serve as the potential biomarkers for HD.

Abstract

Huntington’s disease (HD), caused by expanded CAG repeats encoding a polyglutamine tract in the huntingtin (HTT) protein, presents with a predominant degeneration of neurons in the striatum and cortex. Lines of evidence have observed neuroinflammation, particularly microglial activation, is involved in the pathogenesis of HD. Given that HTT is also expressed in peripheral inflammatory cells, it is possible that inflammatory changes detected in peripheral plasma may be biologically relevant and parallel the neuroinflammatory process of HD patients. By examining the expression levels of 13 microglia-derived inflammatory markers in the plasma of 5 PreHD carriers, 15 HD patients and 16 healthy controls, we found plasma levels of IL-6, MMP-9, VEGF and TGF-β1 were significantly increased in HD patients when compared with the controls, while plasma level of IL-18 were significantly reduced in HD patients compared with controls. Plasma level of IL-6 was reversely correlated with the UHDRS independence scale and functional capacity. To understand the temporal correlation between these inflammatory markers and HD progression, their levels were further tested in plasma from R6/2 mouse HD model at different ages. In rotarod test, R6/2 HD mice started to manifest HD phenotype at 7.5 weeks of age. Higher plasma VEGF levels of R6/2 mice than those of age-matched wild-type (WT) littermates were noted from 7 (presymptomatic stage) to 13 weeks of age (late symptomatic stage). The plasma IL-6 levels of R6/2 mice were higher than those of the WT littermates from 9 (early symptomatic stage) to 13 weeks of age. R6/2 mice demonstrated higher MMP-9 and TGF-β1 levels than their WT littermates from 11 (middle symptomatic stage) to 13 weeks of age. In contrast, the plasma IL-18 level was lower than those in WT littermates since 11 weeks of age. These altered expressions of inflammatory markers may serve as the potential biomarkers for HD onset and progression. Specific inhibition/activation of these inflammatory markers may be the targets of HD drug development.

Introduction

Huntington’s disease (HD) is an autosomal-dominant, progressive neurodegenerative disorder, caused by an unstable CAG trinucleotide repeat expansion encoding a polyglutamine tract in the huntingtin (HTT) protein (MacDonald et al., 1993). The polyglutamine expansion causes a conformational change in the HTT which forms aggregates in both the nucleus and/or cytoplasm of affected neurons and leads to deleterious neuronal functions (Di Prospero and Fischbeck, 2005). Impaired proteasome activity (Valera et al., 2005), transcriptional dysregulation (Cha, 2007), oxidative stress (Stack et al., 2008), mitochondrial and metabolic dysfunction (Browne, 2008), abnormal protein–protein interaction (Giorgini and Muchowski, 2005), neuroinflammation (Bjorkqvist et al., 2008, Dalrymple et al., 2007, Hsiao et al., 2013, Hsiao et al., 2014, Moller, 2010), and microglial activation (Hsiao and Chern, 2010, Pavese et al., 2006, Sapp et al., 2001, Shin et al., 2005, Tai et al., 2007) have been shown to play important roles in the pathogenesis of HD.

Lines of evidence have observed the activation of microglia during the asymptomatic stage and its correlation with disease severity in HD patients (Pavese et al., 2006, Sapp et al., 2001, Tai et al., 2007). Positron emission tomography (PET) has shown early and significant microglial activation in HD patients (Pavese et al., 2006) and presymptomatic HD gene carriers (Tai et al., 2007). Increased microglia-secreted inflammatory mediators, such as IL-6, IL-8, IL-10, matrix metallopeptidase 9 (MMP-9), and chemokine Csingle bondC motif ligand 2 (CCL2) mRNA, have been demonstrated in the brain tissue of post-mortem HD patients (Silvestroni et al., 2009). Increased levels of IL-4, IL-6, IL-8, and TNF-α have been detectable in plasma and cerebrospinal fluid (CSF) of HD patients (Bjorkqvist et al., 2008). Plasma levels of the chemokines eotaxin-3, macrophage inflammatory protein-1β (MIP-1β), eotaxin, monocyte chemotactic protein-1 (MCP-1) and MCP-4 are significantly elevated in HD compared with controls (Wild et al., 2011). Intranuclear aggregates have been shown in microglials in the striatum of R6/2 mice, which leads to microglial activation and subsequent inflammatory factors secretion and neuronal damage (Shin et al., 2005). Altered microglial morphology was also found in the YAC128 mouse model of HD (Franciosi et al., 2012). Thus activated microglia could critically regulate processes of neuronal death and survival by secreting glutamate, neurotrophic factors, and pro- and anti-inflammatory cytokines. An imbalance between neurotoxic and neuroprotective factors may ultimately be responsible for neuronal dysfunction and cell death for HD.

Although the pathology of HD is mainly in the striatum, a few studies have identified substantial biochemical deficits in peripheral tissues (Chang et al., 2012, Chen et al., 2007, Dalrymple et al., 2007, Leoni et al., 2008, Maglione et al., 2005, Nagata et al., 2004, Sawa et al., 1999, Underwood et al., 2006). Given that neuroinflammation plays a role in the pathogenesis of HD and it is practically difficult to obtain brain tissues from HD patients, we aimed to identify potential peripheral inflammatory changes by comparing the plasma levels of a panel of microglia-derived inflammatory markers between HD patients and age/gender-matched control subjects. The panel of microglia-derived inflammatory markers excluded inflammatory markers, alterations of which in HD have been previously reported in literature. In addition, we also measured plasma IL-6 that has been shown to be increased in HD patients to examine if similar result could be seen in our patients (Bjorkqvist et al., 2008). Although it is important to understand the temporal relation between alterations of these markers and the development of HD phenotypes, the limited number of HD patients and lengthy disease course make it hard to clarify this important issue in human samples. The R6/2 mouse model of HD (Mangiarini et al., 1996) carries the mutation in a fragment of the human HTT gene has been widely used to investigate the disease pathogenesis and test potential therapeutic strategies for HD (Li et al., 2005, Morton and Morton, 2013), which addresses importance of finding biomarkers of this model to test the efficacy of potential treatments in preclinical studies. Therefore, we checked the levels of these inflammatory markers in plasma from R6/2 HD mouse model at different ages in order to know if alterations of inflammatory markers can be recapitulated in presymptomatic, early, and late disease stage and if they can also be served as useful biomarkers for R6/2 mice. Our findings successfully demonstrated the potentials of these inflammatory markers as indicators for disease progression in HD patients and also in HD mice.

Section snippets

Ethics statement

This study was performed under a protocol approved by the Institutional Review Boards of Chang Gung Memorial Hospital and all examinations were performed after obtaining written informed consents.

Study population and sample collection

Twenty subjects with HD, including 5 pre-symptomatic HD gene (preHD) carriers and 15 symptomatic HD patients, and 16 healthy controls were recruited in this study. Each group displayed similar gender distribution, age, body weight, body mass index (BMI) and preprandial blood sugar. Unified Huntington’s

Determination of potential inflammatory cytokines in the plasma from HD patients

Microglial activation plays an important role in the neurodegeneration of HD. By examining 13 microglia-derived inflammatory factors in HD patients, preHD carriers and control subjects and adjusting the data for age, gender, use of dopamine antagonist, SSRI, and amantadine, we found plasma levels of IL-6 (HD vs preHD vs control: 1.61 ± 0.21 vs 1.05 ± 0.25 vs 0.95 ± 0.09 pg/mL, P = 0.036), MMP-9 (HD vs preHD vs control: 99.07 ± 11.68 vs 89.96 ± 38.41 vs 52.05 ± 10.54 ng/mL, P = 0.048), VEGF (HD vs preHD vs

Discussion

Collectively, our data show that immune activation in HD is widespread and detectable in peripheral plasma across disease stages. Increased plasma IL-6, MMP-9, VEGF, TGF-β1, and decreased IL-18 may contribute to the pathogenesis in HD. We confirmed these findings in HD mouse models, demonstrating that there are temporal correlations between clinical stages and above inflammatory markers in HD. These alterations in plasma of HD patients as well as HD mice suggest a significant role played by

Disclosure statement

The authors do not have any actual or potential conflicts of interest.

Author’s contribution

Conception and design: Chen C.M. Analysis and interpretation of data: Chen C.M., Chang K.H., Chen Y.C. Acquisition of data: Chen C.M., Chang K.H., Wu Y.R. Drafting the article or revising it critically for important intellectual content: Chang K.H., Chen C.M., Chen Y.C., Final approval of the version to be published: Chang K.H., Chen C.M.

Acknowledgments

This work was supported by grants CMRPG3C0391, CMRPG3D005, and CMRPG3D009 from Chang Gung Memorial Hospital, AS-100-TP2-B02-4 from Academia Sinica, and 102-2314-B-182A-087-MY3 from the National Science Council, Executive Yuan, Taipei, Taiwan.

References (74)

  • S.R. Lee et al.

    PPARgamma agonist pioglitazone reduces matrix metalloproteinase-9 activity and neuronal damage after focal cerebral ischemia

    Biochem. Biophys. Res. Commun.

    (2009)
  • D.Q. Li et al.

    Regulation of MMP-9 production by human corneal epithelial cells

    Exp. Eye Res.

    (2001)
  • J.Y. Li et al.

    The use of the R6 transgenic mouse models of Huntington’s disease in attempts to develop novel therapeutic strategies

    NeuroRx

    (2005)
  • C.Y. Lin et al.

    Neurovascular abnormalities in humans and mice with Huntington’s disease

    Exp. Neurol.

    (2013)
  • M.E. MacDonald et al.

    A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes

    Cell

    (1993)
  • L. Mangiarini et al.

    Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice

    Cell

    (1996)
  • J.P. Miller et al.

    Matrix metalloproteinases are modifiers of huntingtin proteolysis and toxicity in Huntington's disease

    Neuron

    (2010)
  • M. Motta et al.

    Altered plasma cytokine levels in Alzheimer’s disease: correlation with the disease progression

    Immunol. Lett.

    (2007)
  • J. Ojala et al.

    Expression of interleukin-18 is increased in the brains of Alzheimer’s disease patients

    Neurobiol. Aging

    (2009)
  • A. Rahman et al.

    Late onset vascular dysfunction in the R6/1 model of Huntington’s disease

    Eur. J. Pharmacol.

    (2013)
  • T. Salo et al.

    Transforming growth factor-beta 1 up-regulates type IV collagenase expression in cultured human keratinocytes

    J. Biol. Chem.

    (1991)
  • M.L. Springer et al.

    VEGF gene delivery to muscle: potential role for vasculogenesis in adults

    Mol. Cell

    (1998)
  • Y.F. Tai et al.

    Imaging microglial activation in Huntington’s disease

    Brain Res. Bull.

    (2007)
  • V. Tourjman et al.

    Antipsychotics’ effects on blood levels of cytokines in schizophrenia: a meta-analysis

    Schizophr. Res.

    (2013)
  • M.P. Vawter et al.

    TGFbeta1 and TGFbeta2 concentrations are elevated in Parkinson’s disease in ventricular cerebrospinal fluid

    Exp. Neurol.

    (1996)
  • X. Yue et al.

    Comparative study of the neurotrophic effects elicited by VEGF-B and GDNF in preclinical in vivo models of Parkinson’s disease

    Neuroscience

    (2014)
  • G. Battaglia et al.

    Early defect of transforming growth factor beta1 formation in Huntington’s disease

    J. Cell. Mol. Med.

    (2011)
  • E. Bettelli et al.

    Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells

    Nature

    (2006)
  • M. Bjorkqvist et al.

    A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington’s disease

    J. Exp. Med.

    (2008)
  • J. Bouchard et al.

    Cannabinoid receptor 2 signaling in peripheral immune cells modulates disease onset and severity in mouse models of Huntington’s disease

    J. Neurosci.

    (2012)
  • S.E. Browne

    Mitochondria and Huntington’s disease pathogenesis: insight from genetic and chemical models

    Ann. N. Y. Acad. Sci.

    (2008)
  • M. Castellanos et al.

    Plasma metalloproteinase-9 concentration predicts hemorrhagic transformation in acute ischemic stroke

    Stroke

    (2003)
  • K.H. Chang et al.

    Downregulation of genes involved in metabolism and oxidative stress in the peripheral leukocytes of Huntington’s disease patients

    PLoS One

    (2012)
  • A. Dalrymple et al.

    Proteomic profiling of plasma in Huntington’s disease reveals neuroinflammatory activation and biomarker candidates

    J. Proteome Res.

    (2007)
  • J.M. Damsker et al.

    Th1 and Th17 cells: adversaries and collaborators

    Ann. N. Y. Acad. Sci.

    (2010)
  • A.W. Deckel et al.

    Cerebral blood flow velocity decreases during cognitive stimulation in Huntington’s disease

    Neurology

    (1998)
  • N.A. Di Prospero et al.

    Therapeutics development for triplet repeat expansion diseases

    Nat. Rev. Genet.

    (2005)
  • Cited by (111)

    • Growth factors and their receptors: multitasking functionality in health and disease

      2023, Receptor Tyrosine Kinases in Neurodegenerative and Psychiatric Disorders
    • Anti-inflammatory effects of ellagic acid and vanillic acid against quinolinic acid-induced rat model of Huntington's disease by targeting IKK-NF-κB pathway

      2022, European Journal of Pharmacology
      Citation Excerpt :

      Furthermore positron emission tomography studies of HD brains also confirm significant microglial activation with increased levels of pro-inflammatory markers in striatum (Chang et al., 2015; Pavese et al., 2006; Politis et al., 2011). Nuclear factor kappa B (NFκB) pathway is the underlying pathway associated with microglial activation, which leads to oxidative stress and inflammation (Chang et al., 2015). It is reported that activation of IKK-NF-κB pathway in microglia takes place via kynurenine pathway and direct binding of mhtt with it. (

    View all citing articles on Scopus

    All authors are employees of Chang Gung Memorial Hospital and report no financial disclosures.

    View full text