Elsevier

Neuroscience

Volume 237, 1 May 2013, Pages 118-129
Neuroscience

BDNF receptor blockade hinders the beneficial effects of exercise in a rat model of Parkinson’s disease

https://doi.org/10.1016/j.neuroscience.2013.01.060Get rights and content

Abstract

Physical exercise is known to produce beneficial effects to the nervous system. In most cases, brain-derived neurotrophic factor (BDNF) is involved in such effects. However, little is known on the role of BDNF in exercise-related effects on Parkinson’s disease (PD). The aim of this study was to investigate the effects of intermittent treadmill exercise-induced behavioral and histological/neurochemical changes in a rat model of unilateral PD induced by striatal injection of 6-hydroxydopamine (6-OHDA), and the role of BDNF in the exercise effects. Adult male Wistar rats were divided into two main groups: (1) injection of K252a (a blocker of BDNF receptors), and (2) without BDNF receptor blockade. These groups were then subdivided into four groups: control (CLT), sedentary (SED, non-exercised with induction of PD), exercised 3×/week during four weeks before and four weeks after the induction of PD (EXB + EXA), and exercised 3×/week during four weeks after the induction of PD (EXA).

One month after 6-OHDA injections, the animals were subjected to rotational behavioral test induced by apomorphine and the brains were collected for immunohistochemistry and immunoblotting assays, in which we measured BDNF and tyrosine hydroxylase (TH) in the substantia nigra pars compacta (SNc) and the striatum (caudate-putamen, CPu).

Our results showed a significant reduction of rotational asymmetry induced by apomorphine in the exercised parkinsonian rats. BDNF decreased in the SNc of the SED group, and exercise was able to revert that effect. Exercised groups exhibited reduced damage to the dopaminergic system, detected as a decreased drop of TH levels in SNc and CPu. On the other hand, BDNF blockade was capable of substantially reducing TH expression postlesion, implying enhanced dopaminergic cell loss.

Our data revealed that physical exercise is capable of reducing the damage induced by 6-OHDA, and that BDNF receptors are involved in that effect.

Highlights

► Exercise decreases tyrosine hydroxylase deficits in a model of Parkinson’s disease. ► BDNF receptor blockade increases dopaminergic deficits in parkinsonian-exercised rats. ► BDNF appears to be involved in the neuroprotection of dopaminergic neurons.

Introduction

The etiology of Parkinson’s disease (PD) is still not well understood. Postmortem examinations reveal a loss of the dopaminergic neurons in the substantia nigra pars compacta (SNc), and the consequent loss of dopamine in the striatum in the brains of PD patients (Blum et al., 2001). Research on the pathogenesis of the PD has been mainly based on the development of animal models that reproduce the loss of dopaminergic neurons in the SNc. The most used animal model of PD is the injection of dopamine analog 6-hydroxydopamine (6-OHDA) into the caudate-putamen (striatum – CPu) or SNc, and which selectively destroys catecholaminergic neurons, resulting in a decrease of dopamine levels into CPu. That neurotoxin is found in the brains of PD patients (Jellinger et al., 1995) and the injection of 6-OHDA in the central–lateral portion of the CPu is the animal model that most closely resembles the human disease (Tillerson et al., 2002, Schober, 2004). In addition, the intrastriatal injection is less aggressive than other injection sites, and produces a slow evolution of symptoms, which seems to be more suitable for studies that aim therapeutic strategies (Blandini et al., 2008). The analysis of tyrosine hydroxylase (TH) levels is usually employed to follow the progression of the neurodegenerative process in dopaminergic fibers and cell bodies. TH activity is also known to be progressively decreased following the loss of dopamine neurons of the SNc in PD patients (Haavik and Toska, 1998).

With an increasing life expectancy worldwide and hence the number of people with neurodegenerative diseases, several efforts have been made in an attempt to understand the etiology of PD and to develop therapeutic approaches. Exercise is a simple and widely practiced behavior that activates molecular and cellular cascades that support and maintain brain plasticity (Cotman and Berchtold, 2002), and is inversely related to neurodegenerative diseases. However, the extent of the lesion and the type of the exercise (voluntary or forced) may affect the degree of neuroprotection and behavioral improvement induced by physical exercise (Hirsch and Farley, 2009, Alonso-Frech et al., 2011).

Animal studies with PD models have shown that several plasticity processes are triggered by exercise and are involved in neuroprotection mechanisms, such as enhanced angiogenesis (Al-Jarrah et al., 2010), increased anti-inflammatory (Cadet et al., 2003) and decreased inflammatory responses (Wu et al., 2011), improved mitochondrial functions (Lau et al., 2011), and increased neurogenesis in the striatum (Tajiri et al., 2010) and in the SNc (Steiner et al., 2006). Plasticity responses improve the neurochemical deficits, especially TH levels, and both cognitive and motor symptoms (Sutoo and Akiyama, 2003, Cohen et al., 2003, Petzinger et al., 2007, Yoon et al., 2007, O’Dell et al., 2007, Tajiri et al., 2010, Lau et al., 2011, Wu et al., 2011). In models, clinical studies with PD patients have shown that exercise decreases the risk of PD (Chen et al., 2005), improves life quality (Herman et al., 2007), motor coordination (Fisher et al., 2008) and balance (Toole et al., 2000, Goodwin et al., 2008).

It is possible that the neuroprotective effects of exercise are promoted by neurotrophins, such as the brain-derived neurotrophic factor (BDNF). This neurotrophin has a critical role in cell differentiation, neuronal survival, migration, dendritic arborization, synaptogenesis, and synaptic plasticity, acting through tyrosine kinase receptor B (TrkB) (Cotman and Berchtold, 2002).

BDNF expression has been shown to be decreased in animal models of PD (Tajiri et al., 2010, Lau et al., 2011, Wu et al., 2011) and postmortem studies in humans with PD (Howells et al., 2000). Exercise, on the other hand, is able to restore BDNF levels in animals (Tajiri et al., 2010, Lau et al., 2011, Wu et al., 2011) and parkinsonian patients (Ahlskog, 2011). In vitro, BDNF has neuroprotective effects against the neurotoxicity induced by 6-OHDA (Stahl et al., 2011). An in vivo study demonstrated similar results, as Wu and collaborators (2011) showed that BDNF injection in the CPu of mice prior to the injection of lipopolysaccharide (LPS) promoted an improvement of TH levels similar to what was observed in the exercised group. On the other hand, enhanced TH deficits were observed after intracerebroventricular administration of a BDNF receptor blocker in exercised mice prior to the administration of LPS (Wu et al., 2011) as well as in TrkB knockout mice (Baydyuk et al., 2011).

The above-mentioned studies reinforce the idea that BDNF is involved in PD neuroprotection induced by exercise, although they may not be the ideal models to study therapeutic strategies in the early stages of PD. For example, intraperitoneal injections of LPS promote a quick systemic and intense inflammation that activates microglial response in the SNc and other brain regions (Dutta et al., 2008, Wu et al., 2011), resulting in a severe loss of 76% of the TH-positive cells (Wu et al., 2011).

Furthermore, there are no studies in the literature showing the beneficial effects of intermittent exercise in PD animal models. An intermittent exercise routine, as proposed by this study, seems more feasible to PD patients and nonathletes that normally exercise 3 or 4 times a week (Herman et al., 2007). The present exercise protocol may therefore be closer to the reality of PD patients.

The aim of this study was to investigate the possible effects of intermittent treadmill exercise-induced behavioral and histological/neurochemical changes in a rat model of unilateral PD induced by striatal injection of 6-OHDA, and the role of BDNF in these effects.

Section snippets

Experimental procedures

We used two different exercise interventions: (i) exercised before and after the induction of PD – to associate with the findings that physically active individuals have some protection against the occurrence of PD (Sasco et al., 1992, Chen et al., 2005), and (ii) exercised only after the induction of PD – to analyze if physical exercise has beneficial effects after disease is installed (Herman et al., 2007, Fisher et al., 2008). TH immunostaining and immunoblotting were used as indicators of

Behavioral testing

One month after 6-OHDA injections the behavioral testing showed changes in rotational behavior induced by apomorphine [F(3,72) = 17.577; p = 0.000001]. We have not observed any significant rotational asymmetric behavior in the control groups (CLT, 7 ± 1 turns; CLT + K252a, 8 ± 2 turns) and exercised groups (EXB + EXA, 17 ± 3 turns; EXA, 33 ± 2 turns). However, the SED group and the exercised + K252a-treated groups (EXB + EXA + K252a and EXA + K252a) exhibited an increase in the rotational asymmetry following

Discussion

The purpose of this study was to investigate the impact of intermittent exercise protocols on the 6-OHDA model of PD and to analyze the possible involvement of BDNF, a neurotrophin that has a critical role in the mechanisms of exercise-induced neuroprotection (Blum et al., 2001, Mattson et al., 2008, Nguyen et al., 2008, Zigmond et al., 2009, Tajiri et al., 2010, Wu et al., 2011). In general, the intermittent treadmill exercise employed in our study has produced an increase of BDNF levels in

Conclusion

In summary, we showed in this study that intermittent exercise training can lead to improvement of the dopaminergic system and recovery of motor behavior in 6-OHDA-injected rats. We also demonstrated that those beneficial effects seem to involve the BDNF–TrkB system as a major neuroprotective system.

Acknowledgments

This study was supported by FAPESP, CAPES, University of São Paulo – NAPNA and CNPq (Brazil). Thanks are also due to Adilson S. Alves for technical assistance. C.C.R., A.F.B.F. and G.P.C.K. were the recipients of fellowships from FAPESP.

References (50)

  • M.P. Mattson et al.

    Mitochondria in neuroplasticity and neurological disorders

    Neuron

    (2008)
  • S.J. O’Dell et al.

    Running wheel exercise enhances recovery from nigrostriatal dopamine injury without inducing neuroprotection

    Neuroscience

    (2007)
  • C.C. Real et al.

    Exercise-induced plasticity of AMPA-type glutamate receptor subunits in the rat brain

    Brain Res

    (2010)
  • R. Salgado-Delgado et al.

    Internal desynchronization in a model of night-work by forced activity in rats

    Neuroscience

    (2008)
  • K. Stahl et al.

    Cytoprotective effects of growth factors: BDNF more potent than GDNF in an organotypic culture model of Parkinson’s disease

    Brain Res

    (2011)
  • B. Steiner et al.

    Enriched environment induces cellular plasticity in the adult substantia nigra and improves motor behavior function in the 6-OHDA rat model of Parkinson’s disease

    Exp Neurol

    (2006)
  • D. Sutoo et al.

    Regulation of brain function by exercise

    Neurobiol Dis

    (2003)
  • N. Tajiri et al.

    Exercise exerts neuroprotective effects on Parkinson’s disease model of rats

    Brain Res

    (2010)
  • M.J. Zigmond et al.

    Triggering endogenous neuroprotective processes through exercise in models of dopamine deficiency

    Parkinsonism Relat Disord

    (2009)
  • S.Y. Wu et al.

    Running exercise protects the substantia nigra dopaminergic neurons against inflammation-induced degeneration via the activation of BDNF signaling pathway

    Brain Behav Immun

    (2011)
  • M.C. Yoon et al.

    Treadmill exercise suppresses nigrostriatal dopaminergic neuronal loss in 6-hydroxydopamine-induced Parkinson’s rats

    Neurosci Lett

    (2007)
  • J.E. Ahlskog

    Does vigorous exercise have a neuroprotective effect in Parkinson disease?

    Neurology

    (2011)
  • M. Al-Jarrah et al.

    Endurance exercise training promotes angiogenesis in the brain of chronic/progressive mouse model of Parkinson’s disease

    NeuroRehabilitation

    (2010)
  • F. Alonso-Frech et al.

    Exercise and physical therapy in early management of Parkinson disease

    Neurologist

    (2011)
  • R.M. Arida et al.

    Exercise paradigms to study brain injury recovery in rodents

    Am J Phys Med Rehabil

    (2011)
  • Cited by (87)

    • Exercise in Parkinson’s disease

      2022, Exercise to Prevent and Manage Chronic Disease Across the Lifespan
    • The effects of treadmill exercise in animal models of Parkinson's disease: A systematic review

      2021, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      We observed that the most frequent protocol was light to moderate intensity, performed for 4 weeks, 5 days/week, between 30−40 min/day, with a velocity of 10 and 20 m/min, and started 1–2 days after the PD induction. Furthermore, protocols with short durations, 14 days (Shin et al., 2017a; Shin et al., 2017b), low intensity (2−3 m/min (Yoon et al., 2007)), and a frequency of every other day (Real et al., 2017; Real et al., 2019; Real et al., 2013; Ferreira et al., 2020; Garcia et al., 2017; Binda et al., 2020), were also able to induce improvement in the dopaminergic system, as well as other systems. Thus, the results of the studies summarized herein show that it is important to perform some type of exercise to reverse or mitigate the damage caused by PD induction in animal models, but it is not clear which protocol is the best.

    • Promoting brain health through physical activity among adults exposed to early life adversity: Potential mechanisms and theoretical framework

      2021, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      For instance, experimental studies conducted in rodents have shown that aerobic exercise increases BDNF levels in the striatum (Marais et al., 2009), hippocampus (Vaynman et al., 2004; Wrann et al., 2013), and PFC (D.-C. Wang et al., 2020). Moreover, the neuroprotective effects of aerobic exercise in animal models of Parkinson’s Disease and AD are dependent in part on increased production of BDNF (Real et al., 2013; Xiong et al., 2015), with blockade of BDNF receptors attenuating exercise-induced improvements in brain health (Real et al., 2013; Vaynman et al., 2004). Observational studies in humans have demonstrated that both acute and habitual aerobic exercise are associated with increases in peripheral BDNF levels (Dinoff et al., 2016; Szuhany et al., 2015).

    View all citing articles on Scopus
    View full text