G Model

ARTICLE IN PRESS

BRB 8757 1–6

Brain Research Bulletin xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Brain Research Bulletin journal homepage: www.elsevier.com/locate/brainresbull

1

Review

2

RIFAMPICIN: An antibiotic with brain protective function

3

Q1

Burak Yulug a,∗ , Lütfü Hanoglu a , Ertugrul Kilic b , Wolf Rüdiger Schabitz c a

Department of Neurology, University of Istanbul-Medipol, Istanbul, Turkey Department of Physiology, Brain Research Laboratory, University of Istanbul-Medipol, Istanbul, Turkey c Department of Neurology, Bethel-EvKB, Bielefeld, Germany

4

b

5 6 7

a r t i c l e

8 23

i n f o

a b s t r a c t

9

Article history: Received 11 March 2014 Received in revised form 8 May 2014 Accepted 27 May 2014 Available online xxx

10 11 12 13 14

Besides its well known antibiotic activity rifampicin exerts multiple brain protective functions in acute cerebral ischemia and chronic neurodegeneration. The present mini-review gives an update of the unique activity of rifampicin in different diseases including Parkinson’s disease, meningitis, stroke, Alzheimer’s disease and optic nerve injury. © 2014 Elsevier Inc. All rights reserved.

15

22

Keywords: Rifampicin Stroke Parkinson’s disease Alzheimer’s disease Optic nerve injury Meningitis

24

Contents

16 17 18 19 20 21

1.

25 26 27 28 29 30 31

2.

32

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.1. Stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.2. Meningitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.3. Optic nerve injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.4. Parkinson’s disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.5. Alzheimer’s disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.6. The effects of rifampicin on glucocorticoid receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

00 00 00 00 00 00 00 00 00

33

1. Introduction

34

35Q3 36 37 38 39 40 41

Q2

The overwhelming progress in basic neuroscience research led to many therapeutic advances for neurological diseases. Despite these achievements treatment opportunities for several diseases such as stroke or dementia are still devastating. Therefore, research focused on the development of novel candidates causally interacting in brain disease pathophysiology. Of these candidates, antibiotics are particularly interesting because they exert in addition

∗ Corresponding author. Tel.: +90 506 406 97 14; fax: +90 232 446 80 90. E-mail address: [email protected] (B. Yulug).

to the substance immanent antibiotic activity an array of brain protective functions including the prevention of mitochondrial mediated cytochrome c release, microglial activation, glutamate neurotoxicity, and oxidative stress (Kim and Suh, 2009; Hashimoto, 2008; Mao, 2005; Rothstein et al., 2005; Tomiyama et al., 1996a). Rifampicin is a semisynthetic derivative of the rifampycins, a class of broad-spectrum antibiotics that are fermentation products of Nocardia meditterranei. The common structure of the rifampycins is a napthohydroquinone chromophore spanned by an aliphatic ansa chain (Tomiyama et al., 1996a). Rifampicin reaches maximal serum concentration in 1–4 h after application and its plasma half-time is 2–5 h (Acocella, 1978). The lipophilic ansa chain is mainly responsible for the transport of the drug across the blood-brain barrier (BBB) into the brain parenchyma (Mindermann et al., 1993). In the light

http://dx.doi.org/10.1016/j.brainresbull.2014.05.007 0361-9230/© 2014 Elsevier Inc. All rights reserved.

Please cite this article in press as: Yulug, B., et al., RIFAMPICIN: An antibiotic with brain protective function. Brain Res. Bull. (2014), http://dx.doi.org/10.1016/j.brainresbull.2014.05.007

42 43 44 45 46 47 48 49 50 51 52 53 54 55

G Model BRB 8757 1–6

B. Yulug et al. / Brain Research Bulletin xxx (2014) xxx–xxx

2 56 57 58 59

60

61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91

92

93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116

ARTICLE IN PRESS

of increasing evidences of oxidative stress and recently defined immunological cell death pathways in both acute and chronic neurodegenerative diseases the present review gives an update on the brain protective functions of rifampicin in the major diseases. 1.1. Stroke Stroke is the third leading cause of death and adult morbidity in developed countries (Sudlow and Warlow, 1997; Rothwell, 2001). Many molecular mechanisms including the role of massive inflammation and oxidative injury have been proposed to explain the pathogenesis of this clinically devastating disease (Bowen et al., 2006; Dirnagl et al., 1999; Hal et al., 1997). Poststroke reperfusion leads to free radical accumulation and release which damages intracellular and extracellular membranes ultimately resulting in cell dysfunction and death. The latter one damages intracellular and extracellular membranes ultimately resulting in cell dysfunction and death (Calleja et al., 1998). This concept was confirmed by various studies establishing the neuroprotective efficacy of free-radical scavengers after cerebral ischemia (Hal et al., 1997). The structural feature of rifampicin suggests that this drug may function as a free radical scavenger with its naphthohydroquinone ring which may contribute to the function of a hydroxyl radical scavenger activities in inhibiting neurotoxicity (Tomiyama et al., 1996a; Hal et al., 1997). Furthermore, rifampicin has been shown to downregulate the expression of pro-apoptotic Bax and upregulate the expression of anti-apoptotic Bcl-2, Bcl-XL, and of anti-apoptotic gene products such as XIAP, cIAP2, FLIPs, which play essential roles to block ischemia-mediated cell death (Gollapudi et al., 2003; Kilic et al., 2004; Jing et al., 2014; Notarianni, 2013). In this respect, we have earlier shown that rifampicin efficiently reduced brain injury and increased the number of viable neurons after permanent and transient focal cerebral ischemia in mice (Jing et al., 2014). All these findings suggest that rifampicin may be particularly suited for the treatment of stroke, where both kinds of injury interfere or overlap with each other. Rifampicine may therefore be an ideal candidate because it counteracts infections in acute stroke and acts at the same time as neuroprotectant 1.2. Meningitis Despite an effective antibiotic therapy, bacterial meningitis is still associated with high rates of mortality and permanent sequelae in children and adults (Mustafa et al., 1989). It is widely known that bacterial compounds release the production of reactive oxygen species (ROS), proinflammatory cytokines, excitatory amino acids and induce meningeal inflammation (Leib et al., 1996a,b; Mawatari et al., 1996; Tumani et al., 2000). Additionally, the functional relationship between oxidative injury and excitotoxicity has been established in several in vitro studies (Reynolds and Hastings, 1995). Radical scavengers were, for example, shown to attenuate early pathophysiological changes in bacterial meningitis (Koedel and Pfister, 1999; Böttcher et al., 2000). However, the inflammatory host response after initiation of therapy with antibiotics currently used for meningitis may contribute to early mortality and longterm sequelae in bacterial meningitis (Nau et al., 1999; Böttcher et al., 2000). Antibiotics can lead to the release of proinflammatory components of the bacterial cell wall into cerebrospinal fluid (CSF) during bacterial lysis which can secondary cause a burst of meningeal inflammation (Böttcher et al., 2000; Mustafa et al., 1989; Nau et al., 1997) and injury to the host including loss of membrane function, DNA damage, and cell death (Leib et al., 1996a; Koedel and Pfister, 1999; Böttcher et al., 2000). In this respect, both in vitro and in vivo studies have demonstrated that antibiotics that act by inhibiting protein synthesis, such as rifampin and rifabutin,

release smaller quantities of lipoteiochic acids (LTA)/teichoic acids (TA) than b-lactam antibiotics (Nau et al., 1999; Stuertz et al., 1998). Nau et al. compared by their interesting study the effects of rifampicin with those of ceftriaxone on mortality, neuronal damage, and LTA-TA concentrations in serum and CSF in a model of experimental meningitis in mice (Nau et al., 1999). After intracerebral infection with S. pneumoniae they applied 2-mg doses of rifampicin or ceftriaxone and demonstrated that rifampicin not only reduced overall mortality during the first 24 h but led to a significant decrease of serum and cerebrospinal fluid concentrations of proinflammatory bacterial compounds compared to ceftriaxone-treated mice. This study confirmes previous data from a rabbit model of pneumococcal meningitis comparing the effect of rifampicin versus ceftriaxone on ROS production of CSF phagocytes, on CSF malondialdehyde (MDA) concentrations, and on neuronal damage. The study suggests that CSF leukocytes from rifampicin-treated rabbits produced less ROS than leukocytes from animals treated with ceftriaxone (Böttcher et al., 2000). Böttcher et al. (2000) interestingly found that the CSF malondialdehyde concentrations and the density of apoptotic neurons in the dentate gyrus were lower after rifampicin than ceftriaxone-treated animals providing further evidence that minimizing the release of proinflammatory bacterial compounds may improve outcome in bacterial meningitis. Favoring the therapeutic role of antibiotics that do not interfere with cell wall synthesis, these findings were suggested with two earlier studies showing that rifabutin and quinupristin–dalfopristin inhibited the rise of tumor necrosis factor in CSF compared with ceftriaxone therapy (Schmidt et al., 1997; Trostdorf et al., 1999). This was suggested by recent human studies showing that intensified treatment with high dose rifampicin could be associated with better survival in patients with tuberculous meningitis (Yulug et al., 2004; Ruslami et al., 2013) In conlusion, with its radical scavenging and immunomodulating efficacy, rifampicin represents an interesting therapeutic candidate for reducing early mortality in bacterial meningitis. 1.3. Optic nerve injury Optic nerve injury models provide valuable opportunities to discover and study mechanisms of axotomy induced retinal ganglion cell apoptosis and allowed the evaluation of several potential strategies for neuroprotection in neurodegenerative diseases. It has been shown that transection of the optical nerve (ON) triggers a highly coordinated response of injury-associated genes, which finally leads to apoptosis of retinal ganglion cells (RGC) (Kilic et al., 2004; Isenmann et al., 1997; Klocker et al., 1998). In neurodegenerative disease, it is widely known that production of reactive oxygen species overwhelm endogenous antioxidant defense mechanisms hereby inducing neuronal cell death. In this respect, antioxidant agents were shown to suppress apoptosis induced by various insults, including axotomy (Kilic et al., 2004; Castagne and Clarke, 1996). The free radical–scavenger activity of rifampicin has been shown by electron spin resonance spectrometric analysis and stable free radical alpha, alpha-diphenylbeta-picrylhydrazyl (DPPH) reduction (Kilic et al., 2004; Karunakar et al., 2003; Namba et al., 1992). Beyond the antioxidative properties of rifampicin, several other neuroprotective mechanisms have been discussed. Gollapudi et al. (2003) who reported that rifampicin-mediated inhibition of apoptosis and activation of caspase-3 and capase-8 occurred at least in part via Glucocorticoid receptor (GR) activation. Additionally, it has been already shown that rifampicin may block programmed cell death through various pro- and anti-apoptotic proteins (Gollapudi et al., 2003; Kilic et al., 2004; Jing et al., 2014; Notarianni, 2013). In the light of these findings, we have earlier evaluated the possible neuroprotective effect of rifampicin comparing the vehicle and rifampicin treated mice after axotomy of mice (Kilic et al.,

Please cite this article in press as: Yulug, B., et al., RIFAMPICIN: An antibiotic with brain protective function. Brain Res. Bull. (2014), http://dx.doi.org/10.1016/j.brainresbull.2014.05.007

117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151

152

153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180

G Model BRB 8757 1–6

ARTICLE IN PRESS B. Yulug et al. / Brain Research Bulletin xxx (2014) xxx–xxx

190

2004). After continious administration of 5 mg/kg rifampicin during the subsequent 14 days of axotomy we have evaluated the survival of RGCs by fluorescence microscopy and demonstrated that in the rifampicin treated group, retinal ganglion cell survival was significantly increased after axotomy as compared with vehicle treated and phosphate-buffered saline treated control animals. These findings altogether suggest that rifampicin is able to prevent neuronal cell degeneration including anti-oxidative and anti-apoptotic mechanisms suggesting a potential role for future treatment of neurodegenerative disorders.

191

1.4. Parkinson’s disease

181 182 183 184 185 186 187 188 189

3

proven regulatory effect on human glucocorticoid receptor and gene expression (Calleja et al., 1998), the immunomodulatory role of rifampicin has been suggested by a recent study showing that rifampicin may suppress nuclear factor-kappa B that may be responsible for decreasing toll-like receptor 2 (TLR2) (Kim et al., 2009) and phosphorylation of mitogen-activated protein kinases (MAPKs) (Bi et al., 2013; Molloy et al., 2013). All these findings support the potential role of rifampicin as a novel anti-inflammatory drug for the treatment of PD. 1.5. Alzheimer’s disease

192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244

Parkinson’s disease (PD) leads to degeneration of dopaminergic neurons in the substantia nigra pars compacta and the deposition of Lewy bodies in surviving neurons (Langston, 2002; Heisters, 2011; Eriksen et al., 2005; Babi and Mahovi, 2008; Bi et al., 2013). Many different factors and pathways have been hypothesized in the pathogenesis of neuronal death in PD including oxidative stress, mitochondrial dysfunction, neuroinflammation, and apoptosis (Bi et al., 2013; Olanow, 2007). It has been already shown that 1methyl-4-phenylpyridinium (MPP+) exerts its neurotoxic actions by inhibition of the complex I activity and induction of lipid peroxidation that has been shown to be associated with activation of apoptosis (Bi et al., 2013; Watanabe et al., 2005). In the light of these findings, we have earlier evaluated the neuroprotective effects of rifampicin on dopaminergic neuron survival in an in vivo model of MPP+ intoxication (Kilic et al., 2004). We found that rifampicin at different concentrations significantly increased the number of surviving dopaminergic neurons which was suggested by a similar study by Oida et al. (Bi et al., 2013; Oida et al., 2006). Dopaminergic toxicity of MPP may be associated with an upregulation of oligomeric species of alpha-synuclein (Bi et al., 2013; Xu et al., 2007) suggesting the role of alpha-synuclein aggregation by mitochondrial dysfunction in PD pathogenesis. This was in agreement with other studies demonstrating that upregulation of alphasynuclein increases cytotoxicity and neurodegeneration (Bennett, 2005). In this respect, Xu et al. demonstrated that rifampicin pretreatment led to a dose-dependent increase in cell viability and reduction in alpha-synuclein expression (Xu et al., 2007) as suggested by previous data showing that rifampicin inhibits alphasynuclein fibrillation and disaggregates existing fibrils (Li et al., 2004). Rifampicin’s neuroprotective effect was further suggested by earlier in vitro studies showing its free radical scavenging activity (Tomiyama et al., 1996a). These studies supported the important role of mitochondria as the major source of ROS responsible for oxidative stress-mediated cell death in PD (Beal, 1992; Adams et al., 2001). In a study by Chen et al. rifampicin pretreatment protects PC12 cells against rotenone-induced cell death by suppressing rotenone-induced apoptosis and mitochondrial oxidative stress (Chen et al., 2010). However the molecular mechanism underlying this neuroprotection remain unknown. In this respect, a very recent proteomic analysis study has shown that rifampicin exerted protective cellular responses via the up regulation of glucose-regulated protein 78 which is a chaperone protein localized in the endoplasmic reticulum and plays important role in cytoprotection and cell survival under endoplasmic stress in response to accumulation of misfolded proteins (Bi et al., 2011) Increasing evidence has demonstrated that also neuroinflammation, which is characterized by activated microglia and infiltrating T cells at the sites of neuronal injury, plays an important role in the pathophysiology of PD (Bi et al., 2013; Qian et al., 2010; Lu et al., 2010). Investigations using lypopolysacharide-stimulated BV2 microglial cells demonstrated that rifampicin significantly inhibited the LPS-induced expression of pro-inflammatory mediators (Bi et al., 2013; Dutta et al., 2008). Besides rifampicin’s

Alzheimer’s disease (AD) is a progressive neurodegenerative disease, one primary cause of a progressive decline of cognitive and memory functions ultimatively leading to dementia. The neuropathological correlates of the disease are the occurrence of extracellular senile plaques and intracellular neurofibrillary tangles leading to neuronal death and brain atrophy (Schaeffer et al., 2011; Philipson et al., 2010; Hardy and Higgins, 1992; Estus et al., 1997). Beta amyloid (A␤) was shown to be a key factor in AD pathogenesis inducing and maintaining pathophysiological processes such as neuroinflammation, excitotoxixity, oxidative stress, tau hyperphosphorilation and A␤ aggregation (Morrison and Lyketsos, 2005; Dyrks et al., 1992). The generation of A␤ from Amyloid-Precursor-Protein (APP) is a complex pathway and involving enzymatic cleavage of transmembrane APP protein. This may result in amyloid accumulation secondary to an imbalance between protein fragment production and its clearance (Schaeffer et al., 2011; Philipson et al., 2010; Hardy and Higgins, 1992; Estus et al., 1997; Cummings, 2004; Morrison and Lyketsos, 2005). Besides its well known pathophysiological role by various neurodegenerative diseases, earlier studies further demonstrated that free radical production is also involved in the generation of A␤ aggregation (Dyrks et al., 1992; Hensley et al., 1994) suggesting a potential inhibiting role of radical scavengers on A␤ related neurotoxicity. Tomiyama et al. reported that rifampicin inhibited aggregation and fibril formation of synthetic A␤ 1–40 peptide and related neurotoxicity in a dose-dependent manner on rat pheochromocytoma PC12 cells (Tomiyama et al., 1994, 1996b). This suggested that at least one mechanism of rifampicin-mediated inhibition of A␤ aggregation may include scavenging of free radicals and supported a potential therapeutic role of rifampicin for Alzheimer’s disease. Rifampicin may exert its neuroprotective effects in addition to scavenging free radicals by inhibition of amyloid fibril formation (Tomiyama et al., 1997). In accordance with these studies recent evidence strongly suggests that impaired clearance of A␤ across the BBB might lead to the formation of A␤ brain deposits and Alzheimer’s disease progression (Endoh et al., 1999; Cirrito et al., 2005; Vogelgesang et al., 2002; Kuhnke et al., 2007; Lam et al., 2001). Thus, the efflux transporter permeability glycoprotein (P-gp) may play an important role for the elimination of A␤1–40 and A␤1–42 from the brain across the BBB (Endoh et al., 1999; Cirrito et al., 2005; Vogelgesang et al., 2002; Kuhnke et al., 2007; Lam et al., 2001). Abuznait et al. demonstrated that decreased intracellular accumulation of A␤1–40 is associated with P-gp up-regulation caused by rifampicin (Lam et al., 2001). These findings were consistent with their results on an in vitro concentration-dependent increase in Pgp expression and activity by various drugs, including rifampicin (Abuznait et al., 2011a). Moreover, a very recent mice study has shown that the upregulation of Low density lipoprotein receptorrelated protein 1 (LRP1) and P-gp at the BBB by rifampicin and caffeine enhanced brain A␤ clearance and suggested the presence of a possible transporter/receptor that plays an important role in A␤ clearance which is upregulated by rifampicin (Abuznait et al., 2011b). All these findings together support a possible relationship between P-gp dysregulation and cognitive improvement in

Please cite this article in press as: Yulug, B., et al., RIFAMPICIN: An antibiotic with brain protective function. Brain Res. Bull. (2014), http://dx.doi.org/10.1016/j.brainresbull.2014.05.007

245 246 247 248 249 250 251 252 253

254

255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308

G Model

ARTICLE IN PRESS

BRB 8757 1–6

B. Yulug et al. / Brain Research Bulletin xxx (2014) xxx–xxx

4

Table 1 Summary of positive trials of rifampicin treatment for neuroprotection. Disease

309 310 311 312 313 314 315 316 317 318 319

Outcome

Study

Source

Animal

Human

Stroke

↓ Infarct volumes ↓ Apoptotic cell death

+



Yulug et al. (2004)

Menengitis

↓ Neuronal damage ↓ Proinflamatory mediators ↓ ROS production ↓ Mortality ↑ Clinical survival

+

+

Nau et al. (1999) Böttcher et al. (2000) Stuertz et al. (1998) Schmidt et al. (1997) Trostdorf et al. (1999) Ruslami et al. (2013) Alvarez-Uria et al. (2013)

Optic nerve injury

↓ Apoptotic retinal ganglion cell death

+



Kilic et al. (2004)

Parkinson’s disease

↑ Dopaminergic cell survival ↓ Alpha synuclein expression ↑ GRP78 expression ↓ MPP+-induced apoptosis

+



Kilic et al. (2004) Oida et al. (2006) Xu et al. (2007) Li et al. (2004) Chen et al. (2010) Jing et al. (2014)

Alzheimer’s disease

↓ Amyloid Beta aggregation ↑ Amyloid Beta clearance ↑ P-gp Improved clinical SADAScog score

+

+

Tomiyama et al. (1994, 1997) Abuznait et al. (2011a) Qosa et al. (2012) Loeb et al. (2004) Namba et al. (1992)

patients with Alzheimer’s disease suggesting that in addition to oxidative injury and fibril aggregation, targeting P-gp by rifampicin could be an effective strategy in decreasing the progression of Alzheimer’s disease. In contrast to the rapidly increasing preclinical data, only a few clinical studies evaluated the effects of rifampicin in patients with Alzheimer’ disease. Namba et al. reported by their recent study that non-demented elderly leprosy patients that were on rifampicine treatment for years showed an unusual absence of senile plaques in their brains compared with age-matched controls (Namba et al., 1992). Another study by Loeb et al. showed that oral daily doses of rifampin 300 mg for 3 months in patients with

mild to moderate AD improved cognitive function measured with the Standardized Alzheimer’s Disease Assessment Scale-Cognitive Subscale (SADAScog score) (Qosa et al., 2012). However, these promising pilot results could not be replicated in a recent study were patients were treated twelve months with rifampicin (Loeb et al., 2004). Overall, despite strong preclinical evidences this controversial clinical findings together suggest that more clinical research and related functional neuroimaging data (etc. amyloidPET) with combined clinical assessment scores is needed in order to investigate the clinical relevance of the neuroprotective effect of Q4 rifampicin (Table 1 and Fig. 1).

Anti-apoptotic

Anti-inflamatory

•Stimulates anti-apoptotic Bcl-2, Bcl-XL, XIAP, CIAP2 and FLIP activities

•Reduces pro-inflamatory cytokines and bacterial compounds

•Reduces pro-apoptotic Bax, Caspase 3 and caspase 8 activities

•Reduces NF B, MAPKs and TLR-2 activities •Increases GR agonist activity

•Increases eNOS activity

Rifampicin

Anti-oxidant

Anti-Alzheimer

•Reduces ROS production

•Reduces A β aggregation

•Reduces mitochondrial oxidative stress

•Reduces amyloid fibril formation •Increases permeability glycoprotein

Anti-Parkinson •Reduces MPP + toxicity •Reduces alpha-synuclein aggregation •Increases glucose-regulated protein 78 Fig. 1. Multiple mechanisms of rifampicin.

Please cite this article in press as: Yulug, B., et al., RIFAMPICIN: An antibiotic with brain protective function. Brain Res. Bull. (2014), http://dx.doi.org/10.1016/j.brainresbull.2014.05.007

320 321 322 323 324 325 326 327 328 329 330

G Model BRB 8757 1–6

ARTICLE IN PRESS B. Yulug et al. / Brain Research Bulletin xxx (2014) xxx–xxx

331

1.6. The effects of rifampicin on glucocorticoid receptors

370

Besides studies suggesting free radical scavenging as a potential neuroprotective mechanism of rifampicin, earlier studies have further shown that rifampicin can also activate glucocorticoid receptors (Calleja et al., 1998). Despite conflicting reports regarding the protective role of corticosteroids (Wallerath et al., 1999), the activation of glucocorticoid receptors has been shown not only to mediate neuroprotection but also exert to myocardioprotective effects mediated by nontranscriptional activation of endothelial nitric oxide synthase (eNOS) through the PI3K/Akt pathway (Hafezi-Moghadam et al., 2002). Additionally, high-dose corticosteroids given within 2 h of transient cerebral ischemia were shown to increase eNOS activity, augment regional cerebral blood flow and reduce cerebral infarct size abolished by treatment with the glucocorticoid receptor (GR) antagonist (Limbourg et al., 2002). This was suggested by Gollapudi et al. (2003) who reported rifampicinmediated inhibition of peripheral blood T lymphocyte apoptosis via GR activation which was associated by inhibition of activation of both caspase-3 and caspase-8 and reversed by GR antagonist (RU486). However rifampicin’s agonist activity on the glucocorticoid receptor may function also as an immunodepressor that can help to decrease host related neuronal damage by neuroinflamation (Calleja et al., 1998; Nau et al., 1999). Additionally, recent studies showed that rifampicin may lead to induction of gene transcription controlled by glucocorticoid receptor- binding elements (Calleja et al., 1998) which were shown to exist in isolated Müller and photoreceptor cells in intact salamander retina and in all cell types in the human eye (Psarra et al., 2003). Activated GRs can inhibit activator protein (AP-1), which is essential for the induction of photoreceptor apoptosis by light and GR-mediated inhibition may occur in the nucleus of retinal cells by a protein- protein interaction of both transcription factors. Thus, induction of GR activity has been shown to prevent light-induced retinal degeneration by interference with AP-1-dependent steps of apoptosis induction in mice (Wenzel et al., 2001). Furthermore regarding recent hypothesis suggesting the role of neuroinflamation related glucocorticoid receptor-signaling insufficiency in the development of Alzheimer’s disease, rifampicin may play an important role in the progress of the disease via the activation of glucocorticoid receptors (Alvarez-Uria et al., 2013).

371

2. Conclusion

332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369

372 373 374 375 376 377 378

379Q5

380

381

382 383 384 385 386 387 388 389 390

The antibiotic rifampicin exerts strong brain protective in various in vivo and in vitro models of neurodegenerative disease including Stroke, Parkinson’s, Alzheimer’s. Pilot clinical studies suggest that patients with neurodegenerative diseases may benefit from rifampicin treatment. These promising findings should be further explored in larger preclinical therapeutic studies and further on in randomized early phase clinical trials. Uncited reference Yerramasetti et al. (2002). References Abuznait, A.H., Cain, C., Ingram, D., Burk, D., Kaddoumi, A., 2011a. Up-regulation of Pglycoprotein reduces intracellular accumulation of beta amyloid: investigation of P-glycoprotein as a novel therapeutic target for Alzheimer’s disease. J. Pharm. Pharmacol. 63, 1111–1118. Abuznait, A.H., Patrick, S.G., Kaddoumi, A., 2011b. Exposure of LS-180 cells to drugs of diverse physicochemical and therapeutic properties up-regulates Pglycoprotein expression and activity. J. Pharm. Pharm. Sci. 14, 236–248. Acocella, G., 1978. Clinical pharmacokinetics of rifampicin. Clin. Pharmacokinet. 3, 108–127.

5

Adams Jr., J.D., Chang, M.L., Klaidman, L., 2001. Parkinson’s disease – redox mechanisms. Curr. Med. Chem. 8, 809–814. Alvarez-Uria, G., Midde, M., Pakam, R., Naik, P.K., 2013. Initial antituberculous regimen with better drug penetration into cerebrospinal fluid reduces mortality in HIV infected patients with tuberculous meningitis: data from an HIV observational. Cohort Study. Tuberc. Res. Treat. 2013, 242604. Babi, T., Mahovi, D., 2008. Parkinson’s disease-challenges in new drug development. Coll. Antropol. 32, 1275–1281. Beal, M.F., 1992. Does impairment of energy metabolism result in excitotoxic neuronal death in neurodegenerative illnesses? Ann. Neurol. 31, 119–130. Bennett, M.C., 2005. The role of alpha-synuclein in neurodegenerative diseases. Pharmacol. Ther. 105, 311–331. Bi, W., Zhu, L., Wang, C., Liang, Y., Liu, J., Shi, Q., Tao, E., 2011. Rifampicin inhibits microglial inflammation and improves neuron survival against inflammation. Brain Res. 13 (1395), 12–20. Bi, W., Zhu, L., Jing, X., Liang, Y., Tao, E., 2013. Rifampicin and Parkinson’s disease. Neurol. Sci. 34, 137–141. Böttcher, T., Gerber, J., Wellmer, A., Smirnov, A.V., Fakhrjanali, F., Mix, E., Pilz, J., Zettl, U.K., Nau, R., 2000. Rifampin reduces production of reactive oxygen species of cerebrospinal fluid phagocytes and hippocampal neuronal apoptosis in experimental Streptococcus pneumoniae meningitis. J. Infect. Dis. 181, 2095–2098. Bowen, K.K., Naylor, M., Vemuganti, R., 2006. Prevention of inflammation is a mechanism of preconditioning-induced neuroprotection against focal cerebral ischemia. Neurochem. Int. 49, 127–135. Calleja, C., Pascussi, J.M., Mani, J.C., Maurel, P., Vilarem, M.J., 1998. The antibiotic rifampicin is a nonsteroidal ligand and activator of the human glucocorticoid receptor. Nat. Med. 4, 92–96. Castagne, V., Clarke, P.G., 1996. Axotomy-induced retinal ganglion cell death in development: its time course and its diminution by antioxidants. Proc. Biol. Sci. 263, 1193–1197. Chen, S., Sun, Y., Zeng, Z., Tao, E., 2010. Rifampicin inhibits apoptosis in rotenoneinduced differentiated PC12 cells by ameliorating mitochondrial oxidative stress. Neural. Regen. Res. 5, 251–256. Cirrito, J.R., Deane, R., Fagan, A.M., Spinner, M.L., Parsadanian, M., Finn, M.B., Jiang, H., Prior, J.L., Sagare, A., Bales, K.R., Paul, S.M., Zlokovic, B.V., Piwnica-Worms, D., Holtzman, D.M., 2005. P-glycoprotein deficiency at the blood-brain barrier increases amyloid-beta deposition in an Alzheimer disease mouse model. J. Clin. Invest. 115, 3285–3290. Cummings, J.L., 2004. Alzheimer’s disease. N. Engl. J. Med. 351, 56–67. Dirnagl, U., Iadecola, C., Moskowitz, M.A., 1999. Pathobiology of ischaemic stroke, an integrated view. Trends Neurosci. 22, 391–397. Dutta, G., Zhang, P., Liu, B., 2008. The lipopolysaccharide Parkinson’s disease animal model: mechanistic studies and drug discovery. Fundam. Clin. Pharmacol. 22, 453–464. Dyrks, T., Dyrks, E., Hartmann, T., Masters, C., Beyreuther, K., 1992. Amyloidogenicity of beta A4 and beta A4-bearing amyloid protein precursor fragments by metalcatalyzed oxidation. J. Biol. Chem. 267, 18210–18217. Endoh, M., Kunishita, T., Tabira, T., 1999. No effect of anti-leprosy drugs in the prevention of Alzheimer’s disease and beta-amyloid neurotoxicity. J. Neurol. Sci. 165 (1), 28–30. Eriksen, J.L., Wszolek, Z., Petrucelli, L., 2005. Molecular pathogenesis of Parkinson disease. Arch. Neurol. 62, 353–357. Estus, S., Tucker, H.M., van Rooyen, C., Wright, S., Brigham, E.F., Wogulis, M., Rydel, R.E., 1997. Aggregated amyloid-beta protein induces cortical neuronal apoptosis and concomitant “apoptotic” pattern of gene induction. J. Neurosci. 17, 7736–7745. Gollapudi, S., Jaidka, S., Gupta, S., 2003. Molecular basis of rifampicin-induced inhibition of Anti-CD95-Induced apoptosis of peripheral blood T lymphocytes: the role of CD95 ligand and FLIPs. J. Clin. Immunol. 23, 11–22. Hafezi-Moghadam, A., Simoncini, T., Yang, Z., Limbourg, F.P., Plumier, J.C., Rebsamen, M.C., Hsieh, C.M., Chui, D.S., Thomas, K.L., Prorock, A.J., Laubach, V.E., Moskowitz, M.A., French, B.A., Ley, K., Liao, J.K., 2002. Acute cardiovascular protective effects of corticosteroids are mediated by non-transcriptional activation of endothelial nitric oxide synthase. Nat. Med. 8, 473–479. Hal, E.D., Ginsberg, M., Bogousslavsky, J., 1997. Antioxidant pharmacotherapy. In: Cerebrovascular Disease: Pathophysiology, Diagnosis and Management Malden. Blackwell, MA, USA, pp. 710–720. Hardy, J.A., Higgins, G.A., 1992. Alzheimer’s disease: the amyloid cascade hypothesis. Science 256, 184–185. Hashimoto, K., 2008. Microglial activation in schizophrenia and minocycline treatment. Prog. Neuropsychopharmacol. Biol Psychiatry 32, 1758–1759. Heisters, D., 2011. Parkinson’s: symptoms, treatments and research. Br. J. Nurs. 20, 548–554. Hensley, K., Carney, J.M., Mattson, M.P., Aksenova, M., Harris, M., Wu, J.F., Floyd, R.A., Buttereld, D.A., 1994. A model for beta-amyloid aggregation and neurotoxicity based on free radical generation by the peptide: relevance to Alzheimer disease Proc. Proc. Natl. Acad. Sci. U. S. A. 91, 3270–3274. Isenmann, S., Wahl, C., Krajewski, S., Reed, J.C., Bähr, M., 1997. Up-regulation of Bax protein in degenerating retinal ganglion cells precedes apoptotic cell death after optic nerve lesion in the rat. Eur. J. Neurosci. 9, 1763–1772. Jing, X., Shi, Q., Bi, W., Zeng, Z., Liang, Y., Wu, X., Xiao, S., Liu, J., Yang, L., Tao, E., 2014. Rifampicin Protects PC12 Cells from Rotenone-Induced Cytotoxicity by Activating GRP78 via PERK-eIF2␣-ATF4 Pathway. PLoS One 9, e92110. Karunakar, N., Prabhakar, M.C., Krishna, D.R., 2003. Determination of antioxidant activity of some drugs using high-pressure liquid chromatography. Arzneimittelforschung 53, 254–259.

Please cite this article in press as: Yulug, B., et al., RIFAMPICIN: An antibiotic with brain protective function. Brain Res. Bull. (2014), http://dx.doi.org/10.1016/j.brainresbull.2014.05.007

391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476

G Model BRB 8757 1–6 6 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555

ARTICLE IN PRESS B. Yulug et al. / Brain Research Bulletin xxx (2014) xxx–xxx

Kilic, U., Kilic, E., Lingor, P., Yulug, B., Bähr, M., 2004. Rifampicin inhibits neurodegeneration in the optic nerve transection model in vivo and after 1methyl-4-phenylpyridinium intoxication in vitro. Acta Neuropathol. 108, 65–68. Kim, H.S., Suh, Y.H., 2009. Minocycline and neurodegenerative diseases. Behav. Brain Res. 196, 168–179. Kim, S.K., Kim, Y.M., Yeum, C.E., Jin, S.H., Chae, G.T., Lee, S.B., 2009. Rifampicin inhibits the LPS-induced expression of Toll-like Receptor 2 via the suppression of NFkappaB DNA-binding activity in RAW 264.7 cells. Kor. J. Physiol. Pharmacol. 13, 475–482. Klocker, N., Cellerino, A., Bähr, M., 1998. Free radical scavenging and inhibition of nitric oxide synthase potentiates the neurotrophic effects of brain-derived neurotrophic factor on axotomized retinal ganglion cells in vivo. J. Neurosci. 18, 1038–1046. Koedel, U., Pfister, H., 1999. Oxidative stress in bacterial meningitis. Brain Pathol. 9, 57–67. Kuhnke, D., Jedlitschky, G., Grube, M., Krohn, M., Jucker, M., Mosyagin, I., Cascorbi, I., Walker, L.C., Kroemer, H.K., Warzok, R.W., Vogelgesang, S., 2007. MDR1-PGlycoprotein (ABCB1) mediates transport of Alzheimer’s amyloid-beta peptides – implications for the mechanisms of Abeta clearance at the blood-brain barrier. Brain Pathol. 17, 347–353. Lam, F.C., Liu, R., Lu, P., Shapiro, A.B., Renoir, J.M., Sharom, F.J., Reiner, P.B., 2001. Beta-amyloid efflux mediated by p-glycoprotein. J. Neurochem. 76, 1121–1128. Langston, J.W., 2002. Parkinson’s disease: current and future challenges. Neurotoxicology 23, 443–450. Leib, S.L., Kim, Y.S., Chow, L.L., Sheldon, R.A., Täuber, M.G., 1996a. Reactive oxygen intermediates contribute to necrotic and apoptotic neuronal injury in an infant rat model of bacterial meningitis due to Group B Streptococci. J. Clin. Invest. 98, 2632–2639. Leib, S.L., Kim, Y.S., Ferriero, D.M., Täuber, M.G., 1996b. Neuroprotective effect of excitatory amino acid antagonist kynurenic acid in experimental bacterial meningitis. J. Infect. Dis. 173, 166–171. Li, J., Zhu, M., Rajamani, S., Uversky, V.N., Fink, A.L., 2004. Rifampicin inhibits alphasynuclein fibrillation and disaggregates fibrils. Chem. Biol. 11, 1513–1521. Limbourg, F.P., Huang, Z., Plumier, J.C., Simoncini, T., Fujioka, M., Tuckermann, J., Schütz, G., Moskowitz, M.A., Liao, J.K., 2002. Rapid nontranscriptional activation of endothelial nitric oxide synthase mediates increased cerebral blood flow and stroke protection by corticosteroids. J. Clin. Invest. 110, 1729–1738. Loeb, M.B., Molloy, D.W., Smieja, M., Standish, T., Goldsmith, C.H., Mahony, J., Smith, S., Borrie, M., Decoteau, E., Davidson, W., McDougall, A., Gnarpe, J., O’DONNell, M., Chernesky, M.A., 2004. randomized, controlled trial of doxycycline and rifampin for patients with Alzheimer’s disease. J. Am. Geriatr. Soc. 52, 381–387. Lu, L., Li, F., Wang, X., 2010. Novel anti-inflammatory and neuroprotective agents for Parkinson’s disease. CNS Neurol. Disord. Drug Targets 9, 232–240. Mao, J., 2005. Glutamate transporter: an unexpected target for some antibiotics. Mol. Pain 9, 5. Mawatari, K., Yasui, Y., Sugitani, K., Takadera, T., Kato, S., 1996. Reactive oxygen species involved in the glutamate toxicity of C6 glioma cells via Xc antiporter system. Neuroscience 73, 201–208. Mindermann, T., Landolt, H., Zimmerli, W., Rajacic, Z., Gratzl, O., 1993. Penetration of rifampicin into the brain tissue and cerebral extracellular space of rats. J. Antimicrob. Chemother. 31, 731–737. Molloy, D.W., Standish, T.I., Zhou, Q., Guyatt, G., The DARAD Study Group A, 2013. Multicenter, blinded, randomized, factorial controlled trial of doxycycline and rifampin for treatment of Alzheimer’s disease: the DARAD trial. Int. J. Geriatr. Psychiatry 28, 463–470. Morrison, A.S., Lyketsos, C., 2005. The pathophysiology in Alzheimer’s disease and directions in treatment. Adv. Stud. Nurs. 3 (8), 256–270. Mustafa, M.M., Ramilo, O., Mertsola, J., Risser, R.C., Beutler, B., Hansen, E.J., McCracken, G.H.J.R., 1989. Modulation of inflammation and cachectin activity in relation to treatment of experimental Haemophilus influenzae type b meningitis. J. Infect. Dis. 160, 818–825. Namba, Y., Kawatsu, K., Izumi, S., Ueki, A., Ikeda, K., 1992. Neurofibrillary tangles and senile plaques in brain of elderly leprosy patients. Lancet 340, 978. Nau, R., Zysk, G., Schmidt, H., Fischer, F.R., Stringaris, A.K., Stuertz, K., Brück, W., 1997. Trovafloxacin delays the antibiotic-induced inflammatory response in experimental pneumococcal meningitis. J. Antimicrob. Chemother. 39, 781–788. Nau, R., Wellmer, A., Soto, A., Koch, K., Schneider, O., Schmidt, H., Gerber, J., Michel, U., Brück, W., 1999. Rifampin reduces early mortality in experimental streptococcus pneumoniae meningitis. J. Infect. Dis. 179, 1557–1560. Notarianni, E., 2013. Hypercortisolemia and glucocorticoid receptor-signaling insufficiency in Alzheimer’s disease initiation and development. Curr. Alzheimer Res. 10, 714–731. Oida, Y., Kitaichi, K., Nakayama, H., Ito, Y., Fujimoto, Y., Shimazawa, M., Nagai, H., Hara, H., 2006. Rifampicin attenuates the MPTP-induced neurotoxicity in mouse brain. Brain Res. 1082, 196–204. Olanow, C.W., 2007. The pathogenesis of cell death in Parkinson’s disease. Mov. Disord. 22, S335–S342. Philipson, O., Lord, A., Gumucio, A., O’Callaghan, P., Lannfelt, L., Nilsson, L.N., 2010. Animal models of amyloid-beta-related pathologies in Alzheimer’s disease. FEBS J. 277, 1389–1409.

Psarra, A.M., Bochaton-Piallat, M.L., Gabbiani, G., Sekeris, C.E., Tsacopoulos, M., 2003. Mitochondrial localization of glucocortocoid receptor in glial (Muller) cells in the salamander retina. Glia 41, 38–49. Qian, L., Flood, P.M., Hong, J.S., 2010. Neuroinflammation is a key player in Parkinson’s disease and a prime target for therapy. J. Neural. Trans. 117, 971–979. Qosa, H., Abuznait, A.H., Hill, R.A., Kaddoumi, A., 2012. Enhanced brain amyloid-␤ clearance by rifampicin and caffeine as a possible protective mechanism against Alzheimer’s disease. J. Alzheimers Dis. 31, 151–165. Reynolds, I.J., Hastings, T.G., 1995. Glutamate induces the production of reactive oxygen species in cultured forebrain neurons following NMDA receptor activation. J. Neurosci. 15, 3318–3327. Rothstein, J.D., Patel, S., Regan, M.R., Haenggeli, C., Huang, Y.H., Bergles, D.E., Jin, L., Dykes Hoberg, M., Vidensky, S., Chung, D.S., Toan, S.V., Bruijn, L.I., Su, Z.Z., Gupta, P., Fisher, P.B., 2005. Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature 433, 73–77. Rothwell, P.M., 2001. The high cost of not funding stroke research: a comparison with heart disease and cancer. Lancet 357, 1612–1616. Ruslami, R., Ganiem, A.R., Dian, S., Apriani, L., Achmad, T.H., van der Ven, A.J., Borm, G., Aarnoutse, R.E., van Crevel, R., 2013. Intensified regimen containing rifampicin and moxifloxacin for tuberculous meningitis: an open-label, randomised controlled phase 2 trial. Lancet Infect. Dis. 13, 27–35. Schaeffer, E.L., Figueiro, M., Gattaz, W.F., 2011. Insights into Alzheimer disease pathogenesis from studies in transgenic animal models. Clinics (Sao Paulo). 66 (Suppl. 1), 45–54. Schmidt, H., Zysk, G., Reinert, R.R., Brück, W., Stringaris, A., Chen, V., Stuertz, K., Fischer, F., Bartels, R., Schaper, K.J., Weinig, S., Nau, R., 1997. Rifabutin for experimental pneumococcal meningitis. Chemotherapy 43, 264–271. Stuertz, K., Schmidt, H., Eiffert, H., Schwartz, P., Mäder, M., Nau, R., 1998. Differential release of lipoteichoic and teichoic acids from Streptococcus pneumoniae as a result of exposure to beta-lactam antibiotics, rifamycins, trovafloxacin, and quinupristin/dalfopristin. Antimicrob. Agents Chemother. 42, 277–281. Sudlow, C.L., Warlow, C.P., 1997. “International Stroke Incidence Studies Colloboration” Comparable studies of the incidence of stroke and its pathological types: results from an international colloboration. Stroke 28, 491–499. Tomiyama, T., Asano, S., Suwa, Y., Morita, T., Kataoka, K., Mori, H., Endo, N., 1994. Rifampicin prevents the aggregation and neurotoxicity of amyloid beta protein in vitro. Biochem. Biophys. Res. Commun. 204, 76–83. Tomiyama, T., Shoji, A., Kataoka, K., Suwa, Y., Asano, S., Kaneko, H., Endo, N., 1996a. Inhibition of amyloid beta protein aggregation and neurotoxicity by rifampicin: Its possible function as a hydroxyl radical scavenger. J. Biol. Chem. 271, 839–6845. Tomiyama, T., Shoji, A., Kataoka, K., Suwa, Y., Asano, S., Kaneko, H., Endo, N., 1996b. Inhibition of Amyloid beta Protein Aggregation and Neurotoxicity by Rifampicin, Its possible function as a hydroxyl scavenger. J. Biol. Chem. 271, 6839–6844. Tomiyama, T., Kaneko, H., Kataoka, Ki., Asano, S., Endo, N., 1997. Rifampicin inhibits the toxicity of pre-aggregated amyloid peptides by binding to peptide fibrils and preventing amyloid–cell interaction. Biochem. J. 322 (Pt 3), 859–865. Trostdorf, F., Reinert, R.R., Schmidt, H., Nichterlein, T., Stuertz, K., Schmitz-Salue, M., Sadowski, I., Brück, W., Nau, R., 1999. Quinupristin/dalfopristin attenuates the inflammatory response and reduces the concentration of neuron-specific enolase in the cerebrospinal fluid of rabbits with experimental Streptococcus pneumoniae meningitis. J. Antimicrob. Chemother. 43, 87–94. Tumani, H., Smirnov, A., Barchfeld, S., Olgemöller, U., Maier, K., Lange, P., Brück, W., Nau, R., 2000. Inhibition of glutamine synthetase in rabbit pneumococcal meningitis is associated with neuronal apoptosis in the dentate gyrus. Glia 30, 11–18. Vogelgesang, S., Cascorbi, I., Schroeder, E., Pahnke, J., Kroemer, H.K., Siegmund, W., Kunert-Keil, C., Walker, L.C., Warzok, R.W., 2002. Deposition of Alzheimer’s betaamyloid is inversely correlated with P-glycoprotein expression in the brains of elderly non-demented humans. Pharmacogenetics 12, 535–541. Wallerath, T., Witte, K., Schafer, S.C., Schwarz, P.M., Prellwitz, W., Wohlfart, P., Kleinert, H., Lehr, H.A., Lemmer, B., Förstermann, U., 1999. Down-regulation of the expression of endothelial NO synthase is likely to contribute to glucocorticoidmediated hypertension. Proc. Natl. Acad. Sci. U. S. A. 96, 13357–13362. Watanabe, Y., Himeda, T., Araki, T., 2005. Mechanisms of MPTP toxicity and their implications for therapy of Parkinson’s disease. Med. Sci. Monit. 11, RA17–RA23. Wenzel, A., Grimm, C., Seeliger, M.W., Jaissle, G., Hafezi, F., Kretschmer, R., Zrenner, E., Remé, C.E., 2001. Prevention of photoreceptor apoptosis by activation of the glucocorticoid receptor. Invest. Ophthalmol. Vis. Sci. 42 (7), 1653–1659. Xu, J., Wei, C., Xu, C., Bennett, M.C., Zhang, G., Li, F., Tao, E., 2007. Rifampicin protects PC12 cells against MPP+-induced apoptosis and inhibits the expression of an alpha-synuclein multimer. Brain Res. 1139, 220–225. Yerramasetti, R., Gollapudi, S., Gupta, S., 2002. Rifampicin inhibits CD95-mediated apoptosis of Jurkat T cells via glucocorticoid receptors by modifying the expression of molecules regulating apoptosis. J. Clin. Immunol. 22, 37–47. Yulug, B., Kilic, U., Kilic, E., Bahr, M., 2004. Rifampicin attenuates brain damage in focal ischemia. Brain. Res. 16, 76–80.

Please cite this article in press as: Yulug, B., et al., RIFAMPICIN: An antibiotic with brain protective function. Brain Res. Bull. (2014), http://dx.doi.org/10.1016/j.brainresbull.2014.05.007

556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634

RIFAMPICIN: an antibiotic with brain protective function.

Besides its well known antibiotic activity rifampicin exerts multiple brain protective functions in acute cerebral ischemia and chronic neurodegenerat...
439KB Sizes 7 Downloads 3 Views