5,066
Views
165
CrossRef citations to date
0
Altmetric
Mini Review

The therapeutic potential of chemical chaperones in protein folding diseases

&
Pages 197-202 | Received 13 Dec 2013, Accepted 16 Apr 2014, Published online: 12 May 2014

Abstract

Several neurodegenerative diseases are caused by defects in protein folding, including Alzheimer, Parkinson, Huntington, and prion diseases. Once a disease-specific protein misfolds, it can then form toxic aggregates which accumulate in the brain, leading to neuronal dysfunction, cell death, and clinical symptoms. Although significant advances have been made toward understanding the mechanisms of protein aggregation, there are no curative treatments for any of these diseases. Since protein misfolding and the accumulation of aggregates are the most upstream events in the pathological cascade, rescuing or stabilizing the native conformations of proteins is an obvious therapeutic strategy. In recent years, small molecules known as chaperones have been shown to be effective in reducing levels of misfolded proteins, thus minimizing the accumulation of aggregates and their downstream pathological consequences. Chaperones are classified as molecular, pharmacological, or chemical. In this mini-review we summarize the modes of action of different chemical chaperones and discuss evidence for their efficacy in the treatment of protein folding diseases in vitro and in vivo.

Introduction

The misfolding of proteins is a common event in cells. This can be caused by chance, by environmental factors or by mutations that make the native protein conformation less stable. When proteins misfold, they expose hydrophobic segments that are normally buried in the core of their native conformation. This exposure promotes the formation of intermolecular binding and subsequent aggregation. It is generally accepted that these aggregates are toxic and their accumulation is the cause of neurodegenerative disorders like Alzheimer disease (AD), Parkinson disease (PD), Huntington disease (HD), and prion disease.Citation1

To avoid accumulation of these neurotoxic species, cells have evolved a protein quality control (PQC) system which supervises protein folding and eliminates misfolded proteins before they can exert toxic effects.Citation2 Unfortunately, as we age, the delicate balance of the synthesis, folding, and degradation of proteins can be altered and the load of misfolded protein may overwhelm the PQC system allowing the accumulation of toxic protein aggregates. Under this cellular stress, the unfolded protein response (UPR) is activated.Citation2 Through this response, the cell tries to restore its normal function by stopping protein synthesis and increasing the production of chaperones involved in protein folding. If this response is not sufficient to stop the accumulation of unfolded protein, the UPR directs the cell toward apoptosis. Improving the efficiency of the PQC system, therefore, is one approach to combating protein folding diseases.

Chaperones are one of the major players of the PQC system. As traditionally defined, they oversee the correct folding and assemble of proteins, thus preventing their degradation or aggregation and ensuring their appropriate trafficking and function. The overall proteostatic function of chaperones makes them prime candidates for therapeutic agents for neurodegenerative disease. They are classified into 3 groups: molecular, pharmacological, and chemical.

Molecular chaperones are proteins that interact with the non-native state of other proteins to assist them in their folding or unfolding and their assembly or disassembly. They are not present in the final functional protein structure. They represent the first and most potent line of defense against protein misfolding and the aggregation process.Citation3 For example, overexpression of heat shock proteins (Hsps), the major molecular chaperones in cells, has been shown to be neuroprotective in neurodegenerative diseases,Citation4 and this has led to studies of Hsps as potential therapies.Citation5

Pharmacological chaperones are low molecular weight compounds which specifically bind proteins and induce refolding or structure stabilization, restoring protein function.Citation6 They can be enzyme or receptor ligands or molecules which selectively bind to a particular native conformation of a protein to increase its stability.

Chemical chaperones can be divided into 2 groups: osmolytes and hydrophobic compounds.Citation7 They are also low molecular weight compounds but they have a nonspecific mode of action and in some cases cannot bind directly to the proteins. Unlike the pharmacological chaperones, these molecules usually only have effect at high concentrations (molar), so they have been largely neglected as therapeutic agents. However, recently some of these molecules are receiving increasing attention as potential treatments for neurodegenerative conditions given their complex mechanisms of action which likely act at different levels of the neuropathology cascade.

In this mini-review we will focus on the role and therapeutic potential of chemical chaperones in protein folding diseases, including prion disease.

Osmolyte Chaperones

Cellular osmolytes are ancient members of stress responses. They play an important role for organisms exposed to stress conditions such as fluctuating salinity, desiccation, or extreme temperatures.Citation8 The major osmolytes in eukaryotes are restricted to a few classes of low molecular weight compounds: free amino acids and amino acid derivatives (e.g., glycine, taurine, β-alanine), polyols (e.g., glycerol, sucrose), and methyl-amines (e.g., trimethylamine N-oxide [TMAO]).

Mechanism of action

Under denaturing environmental stresses, the intracellular milieu is enriched with organic osmolytes and these osmolytes increase the stability of proteins without affecting their activity.Citation8 Particular osmolytes appear to be involved in particular stress conditions. For example, polyols protect cells against extreme temperature and dehydration, amino acids protect against extracellular environments that are high in salt concentration, and methylamines are present in urea-rich cells, protecting against the deleterious effect of urea on protein structure.Citation8

Despite these differences, osmolytes share the same non-selective mechanism for stabilization of protein structure; they alter solvent properties. A peptide backbone is hydrophilic but amino acid side chains vary in their hydrophobicity. Thus there is a balance of backbone and side chain interaction in a given solvent that determines the free energy of a folded or unfolded state. Osmolytes sequester water molecules, leaving a hydrophobic environment around the protein. This increases the free energy of the protein’s unfolded state more than its folded state, thereby shifting the folding–unfolding equilibrium toward the folded state, where the hydrophilic protein backbone minimizes its exposure to the hydrophobic surroundings.Citation9-Citation11

In addition to their direct effect on the protein conformation and folding, osmolytes modulate the function of molecular chaperones,Citation12,Citation13 further improving the efficiency of the PQC system.

Evidence for therapeutic effects in vitro

The effect of osmolyte chaperones on neurodegenerative-related protein misfolding has been well studied in vitro.

Experiments with α-synuclein, the protein associated with PD, demonstrated that the methylamine TMAO induced conversion of the unfolded protein into its native state.Citation14 Polyols and TMAO also modulated the aggregation properties of Aβ peptide, which accumulates in AD, accelerating the conformational change from random coil to β-sheet.Citation15,Citation16

Within prion diseases, TMAO and glycerol prevented conversion of the cellular form of prion protein (PrPC) into its infective form (PrPSc) in scrapie-infected mouse neuroblastoma cells, without affecting the existing population of PrPSc.Citation17 In a PrP H187R (PrP187R) cell model, which is a prototype of familial Creutzfeldt Jakob disease (CJD), glycerol also reduced the lysosomal accumulation of PrP187R and facilitated its transport to the cell surface.Citation18 Molecular dynamics simulations studying the conversion of PrP at low pH revealed that TMAO prevented residues that are key to conversion from assuming an extended sheet structure.Citation19

Evidence for therapeutic effects in vivo

Oral administration of 2% of trehalose solution (starting at 21 d of age and continuing until the day the mice were killed) has been shown to improve motor dysfunction and extend lifespan in a transgenic mouse model of HD by minimizing the aggregation propensity of the disease-associated polyglutamine-containing protein Huntingtin.Citation20

Although not a naturally occurring chaperone, the well-studied osmolyte DMSO significantly prolonged disease incubation time and delayed the accumulation of PrPSc in hamsters intracranially inoculated with prions. This was achieved with oral administration of a 7.5% solution (starting at day 0 or 14 dpi). It should be noted however, that clear adverse effects such as weight loss were observed.Citation21

Future therapeutic potential

Despite their high efficiency as anti-aggregation agents and beneficial effects in animal models of neurodegenerative diseases, very few of these osmolytic chaperones have entered into clinical trials because of issues related to their toxicity (the majority of these chemical chaperones require high concentrations for effectiveness) and their lack of specificity. Instead, the group of hydrophobic chaperones, which are much less toxic, are being increasingly examined as therapeutic agents.

Hydrophobic Chaperones

Several molecules have been classified as hydrophobic chaperones, including sodium 4-phenylbutyrate (PBA) and bile acids. The general mechanism of action proposed for chemical chaperones involves the interaction of hydrophobic regions of the chaperone with exposed hydrophobic segments of the unfolded protein. This interaction protects the protein from aggregation. However, whether PBA and bile acids truly demonstrate this behavior is unclear. They do reduce aggregate accumulation in vivo and in vitro and revert endoplasmic reticulum (ER) stress, but it has been recently demonstrated that these molecules have more complex mechanisms of action which influence many levels of regulation.

4-phenylbutyrate

Sodium 4-phenylbutyrate (PBA) is an orally bioavailable, blood brain barrier (BBB) permeable, short-chain fatty acid that has been approved by the Food and Drug Administration (FDA) for treatment of urea cycle disorders. PBA has potential benefit for a wide variety of diseases like cancer, cystic fibrosis, thalassemia, spinal muscular atrophy as well as protein folding diseases such as type 2 diabetes mellitus, amyotrophic lateral sclerosis (ALS), HD, AD, and PD.Citation22-Citation30

Although PBA has been classically described as a chemical chaperone, based on its effects on ER stress and aggregate accumulation, the actual molecular mechanisms involved in its beneficial effects are not completely clear.

Mechanisms of action

Two primary mechanisms of action have been proposed for PBA: chemical chaperone and histone deacetylase (HDAC) inhibitor. Modifications to PBA structure that remove HDAC inhibitory activity do not abolish its effects on protein aggregation or ER stress, suggesting that the protective effects are not based on HDAC inhibition alone but rather may involve direct chaperone-like interactions.Citation31 At the same time, the HDAC inhibitory property of PBA allows it to regulate the transcription of many genes involved in the UPR system; PBA induces the synthesis of molecular chaperonesCitation32 while downregulating general protein synthesis.Citation33 Also of note, epigenetic regulators related to histone acetylation are critical regulators of neuronal gene expression. It has been recently suggested that HDAC inhibitory activity may be related to the enhancement of synaptic plasticity, learning, and memory, indicating that compounds like PBA might be a useful class of therapeutic agents for neurodegenerative disorders.Citation34 Indeed, PBA has been studied as a therapeutic agent in vitro and in vivo.

Evidence for therapeutic effects in vitro

PBA can inhibit the aggregation of recombinant α-synuclein aggregation in vitro, likely through its chaperone effect on hydrophobic interaction.Citation35 PBA also has neuroprotective effects in cell culture studies, likely mediated through HDAC inhibition. In neuronal cell culture models of AD, treatment with PBA led to neuroprotection associated with a decrease in tau phosphorylation. It also modulated the processing of the amyloid precursor protein (APP).Citation36,Citation37 In a cell model of PD, PBA increased the expression of genes important for moderating oxidative stress and protein aggregation, thereby reverting the pathology.Citation28

Evidence for therapeutic effects in vivo

There have been encouraging results from PBA treatment of several animal models of neurodegenerative disease.

Oral administration of 0.5% PBA solution to AD mouse models significantly reduced the number of plaques in the hippocampus and reversed cognitive deficits.Citation38,Citation39 It has been suggested that at least part of this neuroprotective effect is due to the chaperone-like activity of PBA.Citation36

Neuroprotective effects of PBA were also assessed on rotenone-induced PD mouse models. Protection against neurodegeneration was observed when PBA at a dose of 120 mg/kg was intraperitoneally injected 30 min before each oral administration of rotenone.Citation35 Additionally, oral administration of PBA (200 mg/kg/day 3h after injection with MPTP) in an acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD inhibited the expression of proinflammatory molecules and attenuated the production of reactive oxygen species from activated microglia.Citation40

In HD mouse models, daily intraperitoneal injections of PBA (100 mg/kg/day) ameliorated the neurodegenerative phenotype, significantly extending survival and attenuating both gross brain and neuronal atrophy.Citation27,Citation41 Through its HDAC inhibitory activity, PBA increased mRNA for components of the ubiquitin-proteosomal pathway and downregulated caspases implicated in apoptotic cell death. Of note, suberoylanilide hydroxamic acid, another HDAC inhibitor, has been also able to ameliorate motor deficits in a mouse model of HD.Citation42

Future therapeutic potential

While PBA can be utilized with a good efficiency by oral supplementation and treatment seems to have no severe side effects, the main drawback to its therapeutic use is the high dosage required. If a direct translation of the effective dosage in AD mice (200 mg/kg) is done, up to 15 g/day would be required for AD treatment in humans. This concentration was the maximum tolerated in a human tolerability study of HD patients.Citation43 It is possible that modifications of this molecule can be made in order to improve its efficiency. In one study, 4-PBA derivatives carrying a p-nitro, p-amino, or p-methoxy group on the benzene ring were able to influence anti-aggregation activity, although the effects were mild.Citation31

Lipids and detergents (bile acids)

Bile acids (BAs) are acidic steroids that are synthesized from cholesterol in the liver. They are secreted into the intestine where they can be dehydroxylated by bacteria to become secondary BAs like deoxycholic acid (DCA) or ursodeoxycholic acid (UDCA). Secondary BAs can then be returned to the liver and conjugated with amino acids to generate conjugated BAs such as tauroursodeoxycholic acid (TUDCA) which is UDCA conjugated with the amino acid taurine. BAs are the major constituents of bile. While their main role is the solubilization of dietary fats and fat-soluble vitamins to improve absorption from the intestinal lumen, in recent years, neuroprotective functions have been attributed to BAs.Citation44,Citation45

Mechanisms of action

TUDCA and UDCA can reduce the accumulation of toxic aggregates in different experimental models of neurodegenerative diseases. As such, they have been classified as chemical chaperones. However, as is the case for PBA, the observed treatment benefits may also be the result of other functions attributed to these bile acids.Citation46

The cytoprotective effects of UDCA and TUDCA have been attributed to the reduction of reactive oxygen species formation,Citation47 the prevention of mitochondrial dysfunction,Citation48 and the inhibition of apoptosis through both the intrinsicCitation49 and the extrinsic pathway.Citation50

These steroids can also activate specific nuclear receptors and G protein–coupled receptors influencing the expression of genes that encode proteins involved in the regulation of glucose, fatty acid, lipoprotein synthesis, energy metabolism, and the regulation of their own synthesis.Citation51

In vitro data

There is in vitro evidence to support a chaperone activity for BAs. Studies of millimolar concentrations of TUDCA and UDCA have demonstrated their inhibitory effects on the thermal aggregation of different proteins.Citation52,Citation53 This effect may have been secondary to the direct stabilization of the protein in question or through direct interaction with other molecular chaperones, enhancing their function.Citation52,Citation54 However, this chaperone effect is not observed for all proteins; when neurodegenerative disease-related proteins like Aβ peptide were tested, TUDCA was found ineffective at preventing aggregation in vitro,Citation55 although these latter experiments were performed with lower concentrations of TUDCA (micromolar). Also of note, the cytotoxic steroid DCA actually enhanced the in vitro aggregation of prion protein (PrP).Citation56

Despite the lack of effect on aggregation of Aβ peptide in vitro, TUDCA strongly inhibited apoptosis in a cell culture model of AD in which cells expressed the Dutch mutation (E22Q) in amyloid precursor protein.Citation55 Thus the ability of BAs to directly interact with and influence protein misfolding could be protein specific and they may exert protective effects via other mechanisms when used in cellular environments.

In vivo data

The neuroprotective action of BAs has been tested in several animal models of neurodegenerative diseases. In transgenic mouse models of Familial Amyloidotic Polyneuropathy, TUDCA significantly reduced transthyretin (TTR) toxic aggregates without affecting TTR aggregation in vitro,Citation57 again suggesting that this effect was not due to a direct stabilization of the native structure of the protein but rather an effect on other cellular processes such as apoptotic or oxidative mechanisms involved in the early stages of pathology. Feeding APP/PS1 transgenic mice with a diet supplemented with TUDCA reduced the accumulation of Aβ deposits and ameliorated memory deficits. This effect was associated with but not limited to a reduction of γ-secretase activity.Citation58 Administration of TUDCA led to a significant reduction in neuropathology and improved the locomotor and sensorimotor deficits of a transgenic HD mouse model.Citation44 Additionally, intraperitoneal injection of TUDCA in mouse model of PD prevented dopaminergic cell death through the activation of pro-survival pathways.Citation59

Future therapeutic potential

Bile acids, in particular TUDCA and UDCA, have received increasing attention as potential treatments for neurodegenerative conditions given their anti-amyloidogenic activity and their ability to modulate apoptotic pathways.Citation60 TUDCA and UDCA have therapeutic advantages over other compounds with chaperone activity. These BAs are orally bioavailable, BBB permeable, and have a low toxicity profile. In addition, TUDCA has been FDA-approved for use in humans to treat primary biliary cirrhosis.

Translation to Clinical Trials

There have been many efforts to develop small molecules that can inhibit the aggregation of proteins. Numerous anti-amyloidogenic compounds have been discovered either by screening large libraries of organic molecules or by rationally designing compounds based on the structure and dynamics of the protein involved.Citation61,Citation62 The use of these molecules can partially overcome and in some cases complete reverse the pathology in vitro and in vivo using models of neurodegenerative diseases. Unfortunately, those compounds which have been tested in clinical trials have had discouraging results to date.Citation63

There are several factors that could explain why these compounds have failed in humans so far. A major problem is that no effective preclinical diagnoses are readily available for these diseases. It may be that once clinical symptoms appear, the level of protein aggregation and accumulation is too high to be affected by prevention of further misfolding. Compounds designed to act exclusively at the initiation of aggregation will be effective only if given very early in disease. Chaperones, as protein stabilizing molecules, may have less effect on pre-formed aggregates than on the monomer proteins themselves. Designing chaperones that more directly affect the aggregates is challenging because the structure of the toxic aggregates and their mechanism(s) of conversion remain unclear. As newer research begins to reveal more of this structural information, there may be more success in inhibitory molecule design. For example, the extent of β sheet content within a toxic oligomeric species may direct the planar attributes of the inhibitory molecule.Citation64 Finally, if an effective anti-aggregation molecule is discovered or successfully synthesized, this drug must cross the BBB and cannot be toxic at the effective concentration.

The most effective treatment regimen may involve a combination of drugs, some which stop protein accumulation, some which promote clearance of toxic aggregates, and others which act downstream to offset the neurodegenerative cascade. In this context, compounds which themselves have a multi-modal/multi-target mechanism would be ideal. TUDCA, UDCA, and PBA may fulfill this role, in that they are not only able to counterattack the misfolding step but also modulate apoptosis, gene expression, and probably other biological processes involved in the neuropathogenesis. These natural compounds also have the advantage of being orally bioavailable, being BBB permeable, having a low toxicity profile, and already being FDA-approved drugs. Clinical trials are currently underway to study the tolerability and efficacy of TUDCA and UDCA in patients with ALS, TTR amyloidosis, and HD.

Conclusion

As our life expectancy and population numbers continue to increase, so will the prevalence and socioeconomic burden of neurodegenerative diseases. Given this, it is of some urgency to find viable therapeutic strategies. Since the accumulation of misfolded host proteins in the brain is the putative cause for many of these diseases, stabilizing native protein structure is a logical approach. TUDCA and UDCA, which can have anti-aggregation effects as well as effects on apoptosis regulation, may represent safe and effective compounds which act at more than one level of the neuropathogenic cascade. More studies with these promising compounds should be pursued to fully understand their mechanism(s) of action and to expand the repertoire of treatable neurodegenerative diseases.

Abbreviations:
PQC=

protein quality control

UPR=

unfolded protein response

Hsps=

heat shock proteins

TMAO=

trimethylamine N-oxide

PrPC=

cellular form of prion protein

PrPSc=

disease-associated prion protein

DMSO=

dimethyl sulfoxide

PBA=

4-phenylbutyrate

CJD=

Creutzfeldt Jakob disease

ALS=

amylotrophic lateral sclerosis

HD=

Huntington disease

AD=

Alzheimer’s disease

PD=

Parkinson disease

HDAC=

histone deacetylase

MPTP=

1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

BA=

bile acid, DCA, deoxycholic acid

UDCA=

ursodeoxycholic acid

TUDCA=

tauroursodeoxycholic acid

TTR=

transthyretin

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

10.4161/pri.28938

References

  • Jucker M, Walker LC. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 2013; 501:45 - 51; http://dx.doi.org/10.1038/nature12481; PMID: 24005412
  • Doyle SM, Genest O, Wickner S. Protein rescue from aggregates by powerful molecular chaperone machines. Nat Rev Mol Cell Biol 2013; 14:617 - 29; http://dx.doi.org/10.1038/nrm3660; PMID: 24061228
  • Muchowski PJ, Wacker JL. Modulation of neurodegeneration by molecular chaperones. Nat Rev Neurosci 2005; 6:11 - 22; http://dx.doi.org/10.1038/nrn1587; PMID: 15611723
  • Turturici G, Sconzo G, Geraci F. Hsp70 and its molecular role in nervous system diseases. Biochem Res Int 2011; 2011:618127; http://dx.doi.org/10.1155/2011/618127; PMID: 21403864
  • Nemirovsky A, Fisher Y, Baron R, Cohen IR, Monsonego A. Amyloid beta-HSP60 peptide conjugate vaccine treats a mouse model of Alzheimer’s disease. Vaccine 2011; 29:4043 - 50; http://dx.doi.org/10.1016/j.vaccine.2011.03.033; PMID: 21473952
  • Bernier V, Lagacé M, Bichet DG, Bouvier M. Pharmacological chaperones: potential treatment for conformational diseases. Trends Endocrinol Metab 2004; 15:222 - 8; http://dx.doi.org/10.1016/j.tem.2004.05.003; PMID: 15223052
  • Gaestel M, Alberti S. Molecular chaperones in health and disease. Berlin: Springer, 2006.
  • Yancey PH, Clark ME, Hand SC, Bowlus RD, Somero GN. Living with water stress: evolution of osmolyte systems. Science 1982; 217:1214 - 22; http://dx.doi.org/10.1126/science.7112124; PMID: 7112124
  • Lin TY, Timasheff SN. Why do some organisms use a urea-methylamine mixture as osmolyte? Thermodynamic compensation of urea and trimethylamine N-oxide interactions with protein. Biochemistry 1994; 33:12695 - 701; http://dx.doi.org/10.1021/bi00208a021; PMID: 7918496
  • Baskakov I, Bolen DW. Forcing thermodynamically unfolded proteins to fold. J Biol Chem 1998; 273:4831 - 4; http://dx.doi.org/10.1074/jbc.273.9.4831; PMID: 9478922
  • Street TO, Bolen DW, Rose GD. A molecular mechanism for osmolyte-induced protein stability. Proc Natl Acad Sci U S A 2006; 103:13997 - 4002; http://dx.doi.org/10.1073/pnas.0606236103; PMID: 16968772
  • Diamant S, Eliahu N, Rosenthal D, Goloubinoff P. Chemical chaperones regulate molecular chaperones in vitro and in cells under combined salt and heat stresses. J Biol Chem 2001; 276:39586 - 91; http://dx.doi.org/10.1074/jbc.M103081200; PMID: 11517217
  • Street TO, Krukenberg KA, Rosgen J, Bolen DW, Agard DA. Osmolyte-induced conformational changes in the Hsp90 molecular chaperone. Protein Sci 2010; 19:57 - 65; PMID: 19890989
  • Uversky VN, Li J, Fink AL. Trimethylamine-N-oxide-induced folding of alpha-synuclein. FEBS Lett 2001; 509:31 - 5; http://dx.doi.org/10.1016/S0014-5793(01)03121-0; PMID: 11734201
  • Yang DS, Yip CM, Huang TH, Chakrabartty A, Fraser PE. Manipulating the amyloid-beta aggregation pathway with chemical chaperones. J Biol Chem 1999; 274:32970 - 4; http://dx.doi.org/10.1074/jbc.274.46.32970; PMID: 10551864
  • Fung J, Darabie AA, McLaurin J. Contribution of simple saccharides to the stabilization of amyloid structure. Biochem Biophys Res Commun 2005; 328:1067 - 72; http://dx.doi.org/10.1016/j.bbrc.2005.01.068; PMID: 15707986
  • Tatzelt J, Prusiner SB, Welch WJ. Chemical chaperones interfere with the formation of scrapie prion protein. EMBO J 1996; 15:6363 - 73; PMID: 8978663
  • Gu Y, Singh N. Doxycycline and protein folding agents rescue the abnormal phenotype of familial CJD H187R in a cell model. Brain Res Mol Brain Res 2004; 123:37 - 44; http://dx.doi.org/10.1016/j.molbrainres.2004.01.006; PMID: 15046864
  • Bennion BJ, DeMarco ML, Daggett V. Preventing misfolding of the prion protein by trimethylamine N-oxide. Biochemistry 2004; 43:12955 - 63; http://dx.doi.org/10.1021/bi0486379; PMID: 15476389
  • Tanaka M, Machida Y, Niu S, Ikeda T, Jana NR, Doi H, Kurosawa M, Nekooki M, Nukina N. Trehalose alleviates polyglutamine-mediated pathology in a mouse model of Huntington disease. Nat Med 2004; 10:148 - 54; http://dx.doi.org/10.1038/nm985; PMID: 14730359
  • Shaked GM, Engelstein R, Avraham I, Kahana E, Gabizon R. Dimethyl sulfoxide delays PrP sc accumulation and disease symptoms in prion-infected hamsters. Brain Res 2003; 983:137 - 43; http://dx.doi.org/10.1016/S0006-8993(03)03045-2; PMID: 12914974
  • Dover GJ, Brusilow S, Charache S. Induction of fetal hemoglobin production in subjects with sickle cell anemia by oral sodium phenylbutyrate. Blood 1994; 84:339 - 43; PMID: 7517215
  • Camacho LH, Olson J, Tong WP, Young CW, Spriggs DR, Malkin MG. Phase I dose escalation clinical trial of phenylbutyrate sodium administered twice daily to patients with advanced solid tumors. Invest New Drugs 2007; 25:131 - 8; http://dx.doi.org/10.1007/s10637-006-9017-4; PMID: 17053987
  • Zeitlin PL, Diener-West M, Rubenstein RC, Boyle MP, Lee CK, Brass-Ernst L. Evidence of CFTR function in cystic fibrosis after systemic administration of 4-phenylbutyrate. Mol Ther 2002; 6:119 - 26; http://dx.doi.org/10.1006/mthe.2002.0639; PMID: 12095312
  • Wirth B, Brichta L, Hahnen E. Spinal muscular atrophy: from gene to therapy. Semin Pediatr Neurol 2006; 13:121 - 31; http://dx.doi.org/10.1016/j.spen.2006.06.008; PMID: 17027862
  • Cudkowicz ME, Andres PL, Macdonald SA, Bedlack RS, Choudry R, Brown RH Jr., Zhang H, Schoenfeld DA, Shefner J, Matson S, et al, Northeast ALS and National VA ALS Research Consortiums. Phase 2 study of sodium phenylbutyrate in ALS. Amyotroph Lateral Scler 2009; 10:99 - 106; http://dx.doi.org/10.1080/17482960802320487; PMID: 18688762
  • Gardian G, Browne SE, Choi DK, Klivenyi P, Gregorio J, Kubilus JK, Ryu H, Langley B, Ratan RR, Ferrante RJ, et al. Neuroprotective effects of phenylbutyrate in the N171-82Q transgenic mouse model of Huntington’s disease. J Biol Chem 2005; 280:556 - 63; PMID: 15494404
  • Zhou W, Bercury K, Cummiskey J, Luong N, Lebin J, Freed CR. Phenylbutyrate up-regulates the DJ-1 protein and protects neurons in cell culture and in animal models of Parkinson disease. J Biol Chem 2011; 286:14941 - 51; http://dx.doi.org/10.1074/jbc.M110.211029; PMID: 21372141
  • Cuadrado-Tejedor M, García-Osta A, Ricobaraza A, Oyarzabal J, Franco R. Defining the mechanism of action of 4-phenylbutyrate to develop a small-molecule-based therapy for Alzheimer’s disease. Curr Med Chem 2011; 18:5545 - 53; http://dx.doi.org/10.2174/092986711798347315; PMID: 22172064
  • Ozcan U, Yilmaz E, Ozcan L, Furuhashi M, Vaillancourt E, Smith RO, Görgün CZ, Hotamisligil GS. Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science 2006; 313:1137 - 40; http://dx.doi.org/10.1126/science.1128294; PMID: 16931765
  • Mimori S, Ohtaka H, Koshikawa Y, Kawada K, Kaneko M, Okuma Y, Nomura Y, Murakami Y, Hamana H. 4-Phenylbutyric acid protects against neuronal cell death by primarily acting as a chemical chaperone rather than histone deacetylase inhibitor. Bioorg Med Chem Lett 2013; 23:6015 - 8; http://dx.doi.org/10.1016/j.bmcl.2013.08.001; PMID: 24044874
  • Ren M, Leng Y, Jeong M, Leeds PR, Chuang DM. Valproic acid reduces brain damage induced by transient focal cerebral ischemia in rats: potential roles of histone deacetylase inhibition and heat shock protein induction. J Neurochem 2004; 89:1358 - 67; http://dx.doi.org/10.1111/j.1471-4159.2004.02406.x; PMID: 15189338
  • Wright JM, Zeitlin PL, Cebotaru L, Guggino SE, Guggino WB. Gene expression profile analysis of 4-phenylbutyrate treatment of IB3-1 bronchial epithelial cell line demonstrates a major influence on heat-shock proteins. Physiol Genomics 2004; 16:204 - 11; http://dx.doi.org/10.1152/physiolgenomics.00160.2003; PMID: 14583596
  • Konsoula Z, Barile FA. Epigenetic histone acetylation and deacetylation mechanisms in experimental models of neurodegenerative disorders. J Pharmacol Toxicol Methods 2012; 66:215 - 20; http://dx.doi.org/10.1016/j.vascn.2012.08.001; PMID: 22902970
  • Inden M, Kitamura Y, Takeuchi H, Yanagida T, Takata K, Kobayashi Y, Taniguchi T, Yoshimoto K, Kaneko M, Okuma Y, et al. Neurodegeneration of mouse nigrostriatal dopaminergic system induced by repeated oral administration of rotenone is prevented by 4-phenylbutyrate, a chemical chaperone. J Neurochem 2007; 101:1491 - 504; http://dx.doi.org/10.1111/j.1471-4159.2006.04440.x; PMID: 17459145
  • Cuadrado-Tejedor M, Ricobaraza AL, Torrijo R, Franco R, Garcia-Osta A. Phenylbutyrate is a multifaceted drug that exerts neuroprotective effects and reverses the Alzheimer´s disease-like phenotype of a commonly used mouse model. Curr Pharm Des 2013; 19:5076 - 84; http://dx.doi.org/10.2174/1381612811319280006; PMID: 23448463
  • Wiley JC, Meabon JS, Frankowski H, Smith EA, Schecterson LC, Bothwell M, Ladiges WC. Phenylbutyric acid rescues endoplasmic reticulum stress-induced suppression of APP proteolysis and prevents apoptosis in neuronal cells. PLoS One 2010; 5:e9135; http://dx.doi.org/10.1371/journal.pone.0009135; PMID: 20161760
  • Ricobaraza A, Cuadrado-Tejedor M, Garcia-Osta A. Long-term phenylbutyrate administration prevents memory deficits in Tg2576 mice by decreasing Abeta. Front Biosci (Elite Ed) 2011; 3:1375 - 84; PMID: 21622143
  • Wiley JC, Pettan-Brewer C, Ladiges WC. Phenylbutyric acid reduces amyloid plaques and rescues cognitive behavior in AD transgenic mice. Aging Cell 2011; 10:418 - 28; http://dx.doi.org/10.1111/j.1474-9726.2011.00680.x; PMID: 21272191
  • Roy A, Ghosh A, Jana A, Liu X, Brahmachari S, Gendelman HE, Pahan K. Sodium phenylbutyrate controls neuroinflammatory and antioxidant activities and protects dopaminergic neurons in mouse models of Parkinson’s disease. PLoS One 2012; 7:e38113; http://dx.doi.org/10.1371/journal.pone.0038113; PMID: 22723850
  • Ferrante RJ, Kubilus JK, Lee J, Ryu H, Beesen A, Zucker B, Smith K, Kowall NW, Ratan RR, Luthi-Carter R, et al. Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington’s disease mice. J Neurosci 2003; 23:9418 - 27; PMID: 14561870
  • Hockly E, Richon VM, Woodman B, Smith DL, Zhou X, Rosa E, Sathasivam K, Ghazi-Noori S, Mahal A, Lowden PA, et al. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington’s disease. Proc Natl Acad Sci U S A 2003; 100:2041 - 6; http://dx.doi.org/10.1073/pnas.0437870100; PMID: 12576549
  • Hogarth P, Lovrecic L, Krainc D. Sodium phenylbutyrate in Huntington’s disease: a dose-finding study. Mov Disord 2007; 22:1962 - 4; http://dx.doi.org/10.1002/mds.21632; PMID: 17702032
  • Keene CD, Rodrigues CM, Eich T, Chhabra MS, Steer CJ, Low WC. Tauroursodeoxycholic acid, a bile acid, is neuroprotective in a transgenic animal model of Huntington’s disease. Proc Natl Acad Sci U S A 2002; 99:10671 - 6; http://dx.doi.org/10.1073/pnas.162362299; PMID: 12149470
  • Ramalho RM, Viana RJ, Low WC, Steer CJ, Rodrigues CM. Bile acids and apoptosis modulation: an emerging role in experimental Alzheimer’s disease. Trends Mol Med 2008; 14:54 - 62; http://dx.doi.org/10.1016/j.molmed.2007.12.001; PMID: 18218342
  • Geier A, Wagner M, Dietrich CG, Trauner M. Principles of hepatic organic anion transporter regulation during cholestasis, inflammation and liver regeneration. Biochim Biophys Acta 2007; 1773:283 - 308; http://dx.doi.org/10.1016/j.bbamcr.2006.04.014; PMID: 17291602
  • Rodrigues CM, Fan G, Wong PY, Kren BT, Steer CJ. Ursodeoxycholic acid may inhibit deoxycholic acid-induced apoptosis by modulating mitochondrial transmembrane potential and reactive oxygen species production. Mol Med 1998; 4:165 - 78; PMID: 9562975
  • Rodrigues CM, Fan G, Ma X, Kren BT, Steer CJ. A novel role for ursodeoxycholic acid in inhibiting apoptosis by modulating mitochondrial membrane perturbation. J Clin Invest 1998; 101:2790 - 9; http://dx.doi.org/10.1172/JCI1325; PMID: 9637713
  • Rodrigues CM, Solá S, Sharpe JC, Moura JJ, Steer CJ. Tauroursodeoxycholic acid prevents Bax-induced membrane perturbation and cytochrome C release in isolated mitochondria. Biochemistry 2003; 42:3070 - 80; http://dx.doi.org/10.1021/bi026979d; PMID: 12627974
  • Azzaroli F, Mehal W, Soroka CJ, Wang L, Lee J, Crispe IN, Boyer JL. Ursodeoxycholic acid diminishes Fas-ligand-induced apoptosis in mouse hepatocytes. Hepatology 2002; 36:49 - 54; http://dx.doi.org/10.1053/jhep.2002.34511; PMID: 12085348
  • Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. Bile acids as regulatory molecules. J Lipid Res 2009; 50:1509 - 20; http://dx.doi.org/10.1194/jlr.R900007-JLR200; PMID: 19346331
  • Song S, Liang JJ, Mulhern ML, Madson CJ, Shinohara T. Cholesterol-derived bile acids enhance the chaperone activity of α-crystallins. Cell Stress Chaperones 2011; 16:475 - 80; http://dx.doi.org/10.1007/s12192-011-0259-5; PMID: 21380614
  • Berger E, Haller D. Structure-function analysis of the tertiary bile acid TUDCA for the resolution of endoplasmic reticulum stress in intestinal epithelial cells. Biochem Biophys Res Commun 2011; 409:610 - 5; http://dx.doi.org/10.1016/j.bbrc.2011.05.043; PMID: 21605547
  • de Almeida SF, Picarote G, Fleming JV, Carmo-Fonseca M, Azevedo JE, de Sousa M. Chemical chaperones reduce endoplasmic reticulum stress and prevent mutant HFE aggregate formation. J Biol Chem 2007; 282:27905 - 12; http://dx.doi.org/10.1074/jbc.M702672200; PMID: 17626021
  • Viana RJ, Nunes AF, Castro RE, Ramalho RM, Meyerson J, Fossati S, Ghiso J, Rostagno A, Rodrigues CM. Tauroursodeoxycholic acid prevents E22Q Alzheimer’s Abeta toxicity in human cerebral endothelial cells. Cell Mol Life Sci 2009; 66:1094 - 104; http://dx.doi.org/10.1007/s00018-009-8746-x; PMID: 19189048
  • Kim NH, Choi JK, Jeong BH, Kim JI, Kwon MS, Carp RI, Kim YS. Effect of transition metals (Mn, Cu, Fe) and deoxycholic acid (DA) on the conversion of PrPC to PrPres. FASEB J 2005; 19:783 - 5; PMID: 15758042
  • Macedo B, Batista AR, Ferreira N, Almeida MR, Saraiva MJ. Anti-apoptotic treatment reduces transthyretin deposition in a transgenic mouse model of Familial Amyloidotic Polyneuropathy. Biochim Biophys Acta 2008; 1782:517 - 22; http://dx.doi.org/10.1016/j.bbadis.2008.05.005; PMID: 18572024
  • Nunes AF, Amaral JD, Lo AC, Fonseca MB, Viana RJ, Callaerts-Vegh Z, D’Hooge R, Rodrigues CM. TUDCA, a bile acid, attenuates amyloid precursor protein processing and amyloid-β deposition in APP/PS1 mice. Mol Neurobiol 2012; 45:440 - 54; http://dx.doi.org/10.1007/s12035-012-8256-y; PMID: 22438081
  • Castro-Caldas M, Carvalho AN, Rodrigues E, Henderson CJ, Wolf CR, Rodrigues CM, Gama MJ. Tauroursodeoxycholic acid prevents MPTP-induced dopaminergic cell death in a mouse model of Parkinson’s disease. Mol Neurobiol 2012; 46:475 - 86; http://dx.doi.org/10.1007/s12035-012-8295-4; PMID: 22773138
  • Castro RE, Solá S, Ramalho RM, Steer CJ, Rodrigues CM. The bile acid tauroursodeoxycholic acid modulates phosphorylation and translocation of bad via phosphatidylinositol 3-kinase in glutamate-induced apoptosis of rat cortical neurons. J Pharmacol Exp Ther 2004; 311:845 - 52; http://dx.doi.org/10.1124/jpet.104.070532; PMID: 15190125
  • Kuwata K, Nishida N, Matsumoto T, Kamatari YO, Hosokawa-Muto J, Kodama K, Nakamura HK, Kimura K, Kawasaki M, Takakura Y, et al. Hot spots in prion protein for pathogenic conversion. Proc Natl Acad Sci U S A 2007; 104:11921 - 6; http://dx.doi.org/10.1073/pnas.0702671104; PMID: 17616582
  • Hosokawa-Muto J, Kamatari YO, Nakamura HK, Kuwata K. Variety of antiprion compounds discovered through an in silico screen based on cellular-form prion protein structure: Correlation between antiprion activity and binding affinity. Antimicrob Agents Chemother 2009; 53:765 - 71; http://dx.doi.org/10.1128/AAC.01112-08; PMID: 19015328
  • Katsuno M, Tanaka F, Sobue G. Perspectives on molecular targeted therapies and clinical trials for neurodegenerative diseases. J Neurol Neurosurg Psychiatry 2012; 83:329 - 35; http://dx.doi.org/10.1136/jnnp-2011-301307; PMID: 22323772
  • Ferreira NC, Marques IA, Conceição WA, Macedo B, Machado CS, Mascarello A, Chiaradia-Delatorre LD, Yunes RA, Nunes RJ, Hughson AG, et al. Anti-prion activity of a panel of aromatic chemical compounds: in vitro and in silico approaches. PLoS One 2014; 9:e84531; http://dx.doi.org/10.1371/journal.pone.0084531; PMID: 24400098

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.