Elsevier

Brain Research

Volume 1070, Issue 1, 27 January 2006, Pages 232-241
Brain Research

Research Report
Focal ischemia induces expression of protease-activated receptor1 (PAR1) and PAR3 on microglia and enhances PAR4 labeling in the penumbra

https://doi.org/10.1016/j.brainres.2005.10.100Get rights and content

Abstract

Thrombin significantly influences neurodegenerative processes after ischemia. The current literature suggests that the effects are mediated via protease-activated receptors 1, 3 and 4 (PAR1, 3, 4). Therefore, we investigated with immunohistochemical methods whether focal cerebral ischemia altered the expression of PARs in the rodent brain. For this purpose, we used the model of endothelin-induced occlusion of the middle cerebral artery and the model of transcranial permanent occlusion of the middle cerebral artery in mice. In contrast to the exclusively neuronal staining in the brain parenchyma of naïve animals, PAR1 and PAR3 occurred in addition on microglial cells in the penumbra after transient and after permanent focal ischemia. Although microglia activation could be detected for several weeks after the insult, PAR1 and PAR3 were traceable on microglia only 12 and 48 h after the insult, but not on day 7 post-ischemia. PAR4 was expressed, both in naïve and in ischemic animals, exclusively in neuronal cells. However, at the border zone and within the infarct area, enhanced immunohistochemical PAR4 signals were recognized. From our data, we conclude that PAR1 and PAR3 could be involved in thrombin-modulated initiation of post-ischemic inflammation and PAR4 may be associated with neuronal degeneration.

Introduction

The serine protease thrombin is not only involved in the development of ischemic damage by mediating clot formation, but it also affects brain tissue directly. For example, thrombin can induce neuronal cell damage after ischemic insults (Hua et al., 2003, Kitaoka et al., 2002, Kitaoka et al., 2003, Lee et al., 1996, Striggow et al., 2000, Xi et al., 1999). On the other hand, this serine protease can also mediate neuroprotection under certain circumstances: low doses or application as a preconditioning stimulus led to increased neuronal survival (Masada et al., 2000, Striggow et al., 2000).

Protective as well as toxic effects of thrombin have been linked to PARs (for an overview, see Rohatgi et al., 2004, Xi et al., 2003). Our laboratory and others have shown that all PARs (PAR1–4) are widely expressed in the naïve rodent brain (Striggow et al., 2001, Weinstein et al., 1995). PAR1, PAR3 and PAR4 can be activated by thrombin, whereas PAR2 is activated by trypsin and tryptase (Ishihara et al., 1997, Nystedt et al., 1994, Vu et al., 1991, Xu et al., 1998). PARs possess a 7-trans-membrane domain structure and are activated by proteolytic cleavage of the N-terminus resulting in the exposure of a tethered ligand, which ultimately binds to the second extracellular loop and causes receptor activation (for an overview, see Wang and Reiser, 2003). Immunohistochemical analysis of ex vivo slices suggested an expression of PARs exclusively on neuronal structures in the unlesioned rodent brain (Striggow et al., 2001). However, in vitro studies demonstrated that the other major cell types of the central nervous system also express PARs, and functional responses after PAR activation could be elicited in astrocytes and microglial cells (Möller et al., 2000, Suo et al., 2002, Suo et al., 2003, Ubl et al., 1998, Wang et al., 2002a, Wang et al., 2002b). As astrocyte as well as microglia activation is a significant part of the post-ischemic pathophysiology (Danton and Dietrich, 2003, Mergenthaler et al., 2004), it is of interest to investigate whether PARs may be associated with these post-lesional astrocyte and microglia reactions.

For this purpose, we performed endothelin-induced middle cerebral artery occlusion (eMCAO) in rats, which is a technique to induce transient focal ischemia. However, to also detect changes in receptor expression under non-reperfusion conditions, we additionally used the method of transcranial electrocoagulation of the middle cerebral artery (MCA) in mice. In both models, unilateral damage of the cerebral cortex is induced. Additionally, in the rat eMCAO model, striatal areas are affected.

In the current study, we investigated whether the expression pattern of the thrombin receptors PAR1, PAR3 and PAR4 changed after transient and permanent focal ischemia and whether such possible changes could provide a clue about the role of PARs in the pathophysiology of such insults.

Section snippets

Ischemia

To check whether the ischemic conditions in the current work indeed induced damage, in 2–3 rats per time-point, we quantified the infarct area after Nissl staining on systematic randomly sampled slices and calculated the infarct volume by linear trapezoidal extrapolation. The average infarct volume increased with survival time, being approximately 30 mm3 at 12 h post-ischemia, 43 mm3 at 48 h post-ischemia and 77 mm3 at 7 days of survival. Fig. 1A shows a representative example of the infarct

Naïve animals

Former results indicated that in the naïve rodent brain PAR1 is expressed in the neuronal membrane (Striggow et al., 2001). In the current study, immunohistochemistry demonstrated the PAR1 signal also as a thin granular ring in rat and mice brain tissue. These cells were identified as neurons by NeuN double labeling (Fig. 1C, rat brain slice). Such membrane-bound localization of PAR1 was also shown in other cell types (Chevessier et al., 2001). Additionally, we found a pronounced PAR1 signal in

eMCAO

Male Sprague–Dawley rats (250–300 g) were used. The animals were fed with laboratory chow (Altromin, Germany) and water ad libitum and maintained in a thermoregulated environment (17–21°C) during a 12/12 h light/dark cycle. The rats were anesthetized with sodium pentobarbitone (40–50 mg/kg) and placed in a stereotaxic frame. A scalp incision of approximately 1 cm in length was made from a point between the eyes, along the midline towards the back of the skull. The periosteum was then removed

Acknowledgments

This work was supported by Deutsche Forschungsgemeinschaft (DFG grant RE 847/3). The technical support from K. Krautwald is greatly acknowledged.

References (41)

  • F. Chevessier et al.

    Expression of the thrombin receptor (PAR-1) during rat skeletal muscle differentiation

    J. Cell. Physiol.

    (2001)
  • G.H. Danton et al.

    Inflammatory mechanisms after ischemia and stroke

    J. Neuropathol. Exp. Neurol.

    (2003)
  • B. Gabryel et al.

    Role of astrocytes in the pathogenesis of ischemic brain injury

    Neurotox. Res.

    (2001)
  • P. Grabham et al.

    Thrombin receptor activation stimulates astrocyte proliferation and reversal of stellation by distinct pathways: involvement of tyrosine phosphorylation

    J. Neurochem.

    (1995)
  • J.R. Hamilton et al.

    Increased expression of protease-activated receptor-2 (PAR2) and PAR4 in human coronary artery by inflammatory stimuli unveils endothelium-dependent relaxation to PAR2 and PAR4 agonists

    Circ. Res.

    (2001)
  • Y. Hua et al.

    Thrombin exacerbates brain edema in focal cerebral ischemia

    Acta Neurochir., Suppl.

    (2003)
  • H. Ishihara et al.

    Protease-activated receptor 3 is a second thrombin receptor in humans

    Nature

    (1997)
  • T. Kitaoka et al.

    Delayed argatroban treatment reduces edema in a rat model of intracerebral hemorrhage

    Stroke

    (2002)
  • T. Kitaoka et al.

    Effect of delayed argatroban treatment on intracerebral hemorrhage-induced edema in the rat

    Acta Neurochir., Suppl.

    (2003)
  • K.R. Lee et al.

    Edema from intracerebral hemorrhage: the role of thrombin

    J. Neurosurg.

    (1996)
  • Cited by (39)

    • Protease-activated receptor 1 (PAR1) inhibits synaptic NMDARs in mouse nigral dopaminergic neurons

      2020, Pharmacological Research
      Citation Excerpt :

      PAR1 function in normal brain has been less examined, but cumulative evidence supports its role in the regulation of synaptic transmission as well as in learning and memory processes and complex behaviors [24–27]. PAR1 is ubiquitously expressed in the brain, either in neurons or in astrocytes and microglia, although with strong differences between areas and cellular populations [4,28–30]. Substantia nigra pars compacta (SNpc) is among brain areas with highest PAR1 expression [31], with PAR1 localization demonstrated in dopamine (DA) neurons from rodent (rat and mouse) and human midbrain [22,31,32].

    • PAR1 activation affects the neurotrophic properties of Schwann cells

      2017, Molecular and Cellular Neuroscience
      Citation Excerpt :

      It has been proposed that PAR1 is a novel regulator of synaptic responses and a modulator of astrocyte-neuron interactions underlying memory formation (Almonte et al., 2013). Furthermore PAR1 expression, especially in glial cells, is modulated under various pathological conditions (for review, see Luo et al., 2007; Sokolova and Reiser, 2008) suggesting that PAR1 may be involved in glial response to brain damage (Striggow et al., 2001; Boven et al., 2003; Pompili et al., 2004, 2006, 2011; Ishida et al., 2006; Henrich-Noack et al., 2006). Under pathological conditions, PAR1 has been shown to mediate either neuronal death or survival (Ramachandran et al., 2012; Zhen et al., 2016).

    • Serine proteases, serine protease inhibitors, and protease-activated receptors: Roles in synaptic function and behavior

      2011, Brain Research
      Citation Excerpt :

      However, when coexpressed with PAR4, PAR3 forms heterodimers with PAR4 and seems to act as a cofactor for PAR4 activation and signaling through Gαq/11 coupling at low thrombin concentrations (as reviewed in Coughlin, 2000; Macfarlane et al., 2001; and Ramachandran and Hollenberg, 2008). Reminiscent of the studies of PAR1, PAR4 expression was highly upregulated in both neurons and glia in ischemia models (Henrich-Noack et al., 2006, 2010; Rohatgi et al., 2004a). Moreover, reduced infarct volumes were recently observed in PAR4 knockout mice in a transient ischemia/reperfusion model (Mao et al., 2010).

    View all citing articles on Scopus
    View full text