Elsevier

Brain, Behavior, and Immunity

Volume 87, July 2020, Pages 429-443
Brain, Behavior, and Immunity

A role for glia maturation factor dependent activation of mast cells and microglia in MPTP induced dopamine loss and behavioural deficits in mice

https://doi.org/10.1016/j.bbi.2020.01.013Get rights and content

Highlights

  • GMF mediates MPTP activated MCs and oxidant-antioxidant imbalance in MPTP treated midbrain.

  • GMF dependent MCs activation reduces TH-immunoreactivity in VTA, SN STR of the MPTP treated midbrain.

  • GMF mediated MCs activation reduces TH, DAT and VMAT2 proteins expression in the SN of MPTP treated midbrain.

  • GMF mediated MCs activation increases GFAP, IBA1, calpain 1 and ICAM 1 expression SN of MPTP treated midbrain.

  • GMF mediated MCs activation reduces motor behavioral performance in rotarod test and hang test of MPTP treated mice.

Abstract

The molecular mechanism mediating degeneration of nigrostriatal dopaminergic neurons in Parkinson’s disease (PD) is not yet fully understood. Previously, we have shown the contribution of glia maturation factor (GMF), a proinflammatory protein in dopaminergic neurodegeneration mediated by activation of mast cells (MCs). In this study, methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigrostriatal neurodegeneration and astro-glial activations were determined by western blot and immunofluorescence techniques in wild type (WT) mice, MC-deficient (MC-KO) mice and GMF-deficient (GMF-KO) mice, with or without MC reconstitution before MPTP administration. We show that GMF-KO in the MCs reduces the synergistic effects of MC and Calpain1 (calcium-activated cysteine protease enzyme)-dependent dopaminergic neuronal loss that reduces motor behavioral impairments in MPTP-treated mouse. Administration of MPTP increase in calpain-mediated proteolysis in nigral dopaminergic neurons further resulting in motor decline in mice. We found that MPTP administered WT mice exhibits oxidative stress due to significant increases in the levels of malondialdehyde, superoxide dismutase and reduction in the levels of reduced glutathione and glutathione peroxidase activity as compared with both MC-KO and GMF-KO mice. The number of TH-positive neurons in the ventral tegmental area, substantia nigra and the fibers in the striatum were significantly reduced while granulocyte macrophage colony-stimulating factor (GM-CSF), MC-Tryptase, GFAP, IBA1, Calpain1 and intracellular adhesion molecule 1 expression were significantly increased in WT mice. Similarly, tyrosine hydroxylase, dopamine transporters and vesicular monoamine transporters 2 proteins expression were significantly reduced in the SN of MPTP treated WT mice. The motor behavior as analyzed by rotarod and hang test was significantly reduced in WT mice as compared with both the MC-KO and GMF-KO mice. We conclude that GMF-dependent MC activation enhances the detrimental effect of astro-glial activation-mediated oxidative stress and neuroinflammation in the midbrain, and its inhibition may slowdown the progression of PD.

Introduction

Parkinson’s disease (PD) is the most common age associated movement disorder. Clinically, it is characterized by the progressive loss of dopaminergic neurons in the substantia nigra (SN) and ventral tegmental area (VTA) leading to depletion of dopaminergic neurotransmission to the striatum (STR) with resulting motor dysfunction in mid to later stages of PD (Dufty et al., 2007, Esteves et al., 2010, Samantaray et al., 2015, Spillantini et al., 1997). However, the exact pathological mechanism of PD is not fully understood. Accumulating evidences suggest that intracellular mitochondrial dysfunction dependent reactive oxygen species (ROS) production, neuroinflammation, impairment in ubiquitin-proteasomal and autophagy-lysosomal systems along with accumulation of α-synuclein contribute to PD progression (Gandhi and Wood, 2005, Selvakumar et al., 2014, Selvakumar et al., 2015, Selvakumar et al., 2018b, Selvakumar et al., 2019). Neuroinflammation is a result of a distinct cascade of events in the brain (Xanthos and Sandkühler, 2014). However, chronic neuroinflammation can result in detrimental effects involving changes in the brain parenchyma, breach of blood brain barrier (BBB), and neuronal hyperexcitability that ultimately leads to neuronal death (Bañuelos-Cabrera et al., 2014, Hendriksen et al., 2017, Lyman et al., 2014, Maas et al., 2008). Neuroinflammation plays a crucial role in PD pathogenesis, due to age, genetics and other sporadic factors that influence immune alterations that can lead to astro-glial activation associated with dopaminergic neurodegeneration (Chung et al., 2010, Kanaan et al., 2010). The neuronal loss is exacerbated when the central inflammatory response is induced with the active peripheral inflammation in PD (Liu and Bing, 2011).

Emerging evidence suggest that the brain’s resident immunocompetent macrophages such as astro-glial cells are the major source of inflammatory mediators. Microglia also respond to proinflammatory signals that are released by other type of non-neuronal cells such as mast cells (MCs) (Kempuraj et al., 2018c). Different types of extra and intracellular insults promote neurodegeneration through products, such as proteases, calpains, cytokines, chemokines, ROS and others factors released by astro-glial cells (Crocker et al., 2003, Smith et al., 2012). Furthermore, calpains are unique family of calcium-activated cysteine proteases that include isoforms calpain 1 and alpain 2. Both calpains consist of large and unique subunits with common small regulatory subunits (Crocker et al., 2003, Sorimachi et al., 1997). Activated MCs show increased calpain 1 activity and its deficiency reduces IκB-NF-κB pathway activation without affecting MAPKs or NFAT pathways in MCs (Wu et al., 2014).

MCs are principally bone marrow derived hematopoietic progenitors that enter the brain through BBB, to actively participate in the neuroinflammatory process through their actively stored and newly synthesized inflammatory mediators such as cytokines, chemokines, proteases and ROS (Dong et al., 2014, Hendriksen et al., 2017, Kempuraj et al., 2017, Nelissen et al., 2013, Skaper et al., 2012). During the inflammatory condition, MC release their own mediators acting as enzymatic catalysts and recruiters to initiate, amplify, and propagate to other immune and neuronal cells (Dong et al., 2014, Hendriksen et al., 2017). MC releases granulocyte macrophage colony-stimulating factor (GM-CSF), a monomeric glycoprotein during activation (Kempuraj et al., 2017, Kempuraj et al., 2019, Wodnar-Filipowicz et al., 1989). In addition, MC-Tryptase is an enzyme that secreted by MC along with histamine during the MC activation (Kempuraj et al., 2018, Kempuraj et al., 2017, Payne and Kam, 2004). However, the molecular interaction between the neuronal cells and MCs particularly in PD progression is still not fully understood (Kempuraj et al., 2018c, Skaper et al., 2014).

Intracellular small molecules are crucial players in several intracellular modifications, which are involved in a large variety of pathological conditions. In recent years, studies have focused on the potential therapeutic use of small agents with strong biological properties. Glia maturation factor (GMF), a 17-kDa neuroinflammatory protein that was first discovered, purified and sequenced in our laboratory (Kaplan et al., 1991, Lim et al., 1989, Zaheer et al., 1993). GMF consists of a 141-amino acid polypeptide chain is abundant in the brain and predominantly expressed by the brain parenchyma cells such as astrocytes, microglia and neurons (Lim et al., 1988, Lim et al., 1989, Lim and Zaheer, 2006). GMF is upregulated under cellular stress condition and is phosphorylated by protein kinases A and C, casein kinase, and ribosomal S6 kinase at multiple phosphorylation sites (Thangavel et al., 2011, Zaheer et al., 2006, Zaheer and Lim, 1997). In the present study, we investigated the role of GMF in the activation of MCs to release MCs proteases that activate the microglial Calpain 1 expression in a MPTP model of PD. Our findings suggest that the presence of GMF in the MCs makes them more reactive to MPTP toxicity by inducing Calpain 1 release from the microglial cells that leads to dopaminergic depletion in nigrostriatal region in the mice midbrain.

Section snippets

Mouse primary mast cell culture

Bone marrow-derived MCs (BMMCs) were grown by culturing mouse femur bone marrow cells acquired from WT mice as well as from GMF-knockout (GMF-KO) mice as described previously (Kempuraj et al., 2019, Kim et al., 2011, Sayed et al., 2011, Tagen et al., 2009). Briefly, bone marrow from 5 to 10 mice were pooled together in order to get more number of MCs in the culture. Bone marrow cells were grown in high glucose DMEM (GIBCO, Life Technologies, Grand Island, NY) supplemented with IL-3 (10 ng/ml),

GMF-dependent activation of mast cells amplifies motor behavioral impairments in MPTP treated SN of midbrain

To determine the effect of GMF mediated MC activation on motor behavioral deficits in acute MPTP-treated mice, we performed motor behavioral studies. The latency of falling on the rotarod apparatus and neuromuscular strength and motor functions in hang test at 7 days after the last MPTP administration is shown in Fig. 1. In MPTP-treated mice, robust motor deficits, primary evidence for PD like symptoms is manifested by significantly decreased latency of fall (retention time) in rotarod test at

Discussion

In the present study, we demonstrate the sequential action of GMF-dependent murine MC activation and calpain 1 expression involved in the dopaminergic neurodegeneration in the SN and STR of midbrain. Murine MC Proteases and calpain 1 mediate the activation of astro-glial cells and this further leads to dopaminergic neurodegeneration and motor behavioral deficits in WT, MC-KO and GMF-KO mice. The salient findings in the current study include a significant oxidative stress, increased calpain 1,

Conclusions

In summary, the present study demonstrates that reconstitution of MC from WT mice into WT mice, MC-KO mice and GMF-KO mice followed by MPTP administration enhances nigrostriatal dopaminergic degeneration in the midbrain. Administration of MPTP following MC reconstitution enhances oxidative stress by altering levels of MDA, GSH and activities of SOD and GPx, reduces TH, DAT and VMAT2 protein expression. In addition, MPTP reduces number of TH positive dopaminergic neurons in the SN, VTA and STR

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

AZ is a recipient of the Department of Veterans Affairs Research Career Scientist award and the U. S. Department of Veteran Affairs, Office of Research and Development-Biomedical Laboratory and Research and Development (ORD-BLRD) service-Veteran Affairs Merit award I01BX002477 and National Institutes of Health, United States grants AG048205 and NS073670 to AZ supported this work.

References (88)

  • M.M. Khan et al.

    Protection of MPTP-induced neuroinflammation and neurodegeneration by Pycnogenol

    Neurochem. Int.

    (2013)
  • Y. Kitamura et al.

    Decrease of mast cells in W/Wv mice and their increase by bone marrow transplantation

    Blood

    (1978)
  • R. Lim et al.

    Endogenous immunoreactive glia maturation factor-like molecule in cultured rat Schwann cells

    Brain Res.

    (1988)
  • R. Lim et al.

    Impaired motor performance and learning in glia maturation factor-knockout mice

    Brain Res.

    (2004)
  • M. Lyman et al.

    Neuroinflammation: The role and consequences

    Neurosci. Res.

    (2014)
  • A.I.R. Maas et al.

    Moderate and severe traumatic brain injury in adults

    Lancet Neurol.

    (2008)
  • L. Meagher et al.

    Measurement of mRNA for E-selectin, VCAM-1 and ICAM-1 by reverse transcription and the polymerase chain reaction

    J. Immunol. Methods

    (1994)
  • J. Miklossy et al.

    Role of ICAM-1 in persisting inflammation in Parkinson disease and MPTP monkeys

    Exp. Neurol.

    (2006)
  • R. Paus et al.

    Neuroimmunoendocrine circuitry of the ‘brain-skin connection’

    Trends Immunol.

    (2006)
  • S. Phani et al.

    VTA neurons show a potentially protective transcriptional response to MPTP

    Brain Res.

    (2010)
  • H.-R. Rodewald et al.

    Widespread immunological functions of mast cells: fact or fiction?

    Immunity

    (2012)
  • G.P. Selvakumar et al.

    Escin attenuates behavioral impairments, oxidative stress and inflammation in a chronic MPTP/probenecid mouse model of Parkinson's disease

    Brain Res.

    (2014)
  • J.A. Smith et al.

    Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases

    Brain Res. Bull.

    (2012)
  • S. Sugama et al.

    Age-related microglial activation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic neurodegeneration in C57BL/6 mice

    Brain Res.

    (2003)
  • S.J. Wertheimer et al.

    Intercellular adhesion molecule-1 gene expression in human endothelial cells. Differential regulation by tumor necrosis factor-alpha and phorbol myristate acetate

    J. Biological Chem.

    (1992)
  • M.J. West

    Stereological methods for estimating the total number of neurons and synapses: issues of precision and bias

    Trends Neurosci.

    (1999)
  • H.Q. Yan et al.

    Delayed increase of tyrosine hydroxylase expression in rat nigrostriatal system after traumatic brain injury

    Brain Res.

    (2007)
  • A. Zaheer et al.

    Protein kinase A (PKA)- and protein kinase C-phosphorylated glia maturation factor promotes the catalytic activity of PKA

    J. Biol. Chem.

    (1997)
  • A. Zameer et al.

    Increased ICAM-1 and VCAM-1 expression in the brains of autoimmune mice

    J. Neuroimmunol.

    (2003)
  • H. Akiyama et al.

    Expression of intercellular adhesion molecule (ICAM)-1 by a subset of astrocytes in Alzheimer disease and some other degenerative neurological disorders

    Acta Neuropathol.

    (1993)
  • M.A. Brown et al.

    Mast cells are important modifiers of autoimmune disease: with so much evidence, why is there still controversy?

    Front. Immunol.

    (2012)
  • Y.C. Chung et al.

    Paroxetine prevents loss of nigrostriatal dopaminergic neurons by inhibiting brain inflammation and oxidative stress in an experimental model of Parkinson’s disease

    J. Immunol.

    (2010)
  • R. Corti et al.

    Evolving concepts in the triad of atherosclerosis, inflammation and thrombosis

    J. Thromb. Thrombolysis

    (2004)
  • S.J. Crocker et al.

    Inhibition of Calpains prevents neuronal and behavioral deficits in an MPTP mouse model of Parkinson's disease

    J. Neurosci.

    (2003)
  • J.S. Dahlin et al.

    Detection of circulating mast cells in advanced systemic mastocytosis

    Leukemia

    (2016)
  • M. Diepenbroek et al.

    Overexpression of the calpain-specific inhibitor calpastatin reduces human alpha-Synuclein processing, aggregation and synaptic impairment in [A30P]αSyn transgenic mice

    Hum. Mol. Genet.

    (2014)
  • H. Dong et al.

    Mast cells and neuroinflammation

    Med. Sci. Monit. Basic Res.

    (2014)
  • S. Gandhi et al.

    Molecular pathogenesis of Parkinson's disease

    Hum. Mol. Genet.

    (2005)
  • N. Gaudenzio et al.

    Analyzing the functions of mast cells in vivo using 'Mast Cell Knock-in' mice

    J Vis Exp

    (2015)
  • H.J. Gundersen

    Stereology: the fast lane between neuroanatomy and brain function–or still only a tightrope?

    Acta Neurol. Scand. Suppl.

    (1992)
  • Y. Jin et al.

    Mast cells are early responders after hypoxia-ischemia in immature rat brain

    Stroke

    (2009)
  • A. Kaizaki et al.

    Maternal MDMA administration in mice leads to neonatal growth delay

    J. Toxicol. Sci.

    (2014)
  • N.M. Kanaan et al.

    Age-related accumulation of Marinesco bodies and lipofuscin in rhesus monkey midbrain dopamine neurons: Relevance to selective neuronal vulnerability

    J. Compar. Neurol.

    (2007)
  • R. Kaplan et al.

    Molecular cloning and expression of biologically active human glia maturation factor-beta

    J. Neurochem.

    (1991)
  • Cited by (18)

    • Analysis of m6A modification regulators in the substantia nigra and striatum of MPTP-induced Parkinson's disease mice

      2022, Neuroscience Letters
      Citation Excerpt :

      All the animal experiments in this study were approved by the Institutional Animal Care and Use Committee of Chongqing Medical University. The rotarod test was used to assess motor coordination and balance of mice as previously described with minor modifications [25]. In brief, mice were trained to stay on a rotating rotarod (Ugo Basile Srl, Gemonio, Varese, Italy) three days before performing the motor function test at a rotating speed of 30 rpm, with 3 consecutive trials performed daily.

    • Silibinin attenuates motor dysfunction in a mouse model of Parkinson's disease by suppression of oxidative stress and neuroinflammation along with promotion of mitophagy

      2021, Physiology and Behavior
      Citation Excerpt :

      Mice in the control group and in the MPTP vehicle group were administered orally with the same volume of 0.5% sodium carboxymethyl cellulose used as vehicle. In order to test the motor coordination of experimental animals, we carried out the rotarod test [33]. Mice were trained 3 days before test.

    • Andrographolide suppresses NLRP3 inflammasome activation in microglia through induction of parkin-mediated mitophagy in in-vitro and in-vivo models of Parkinson disease

      2021, Brain, Behavior, and Immunity
      Citation Excerpt :

      To study the role of Andro in-vivo, we have developed a sub-acute model of PD by MPTP intoxication for continuous 5 days. MPTP causes severe motor deficits together with loss dopamine and TH positive dopaminergic neurons in the SNpc of mice brain (Selvakumar et al., 2020). Moreover, MPTP administration showed increased expression of NLRP3 inflammasomes and its downstream targets IL-1β and IL-18 which was in accordance with previous reports (Cho et al., 2020; Lee et al., 2019).

    View all citing articles on Scopus
    View full text