Skip to main content

SYSTEMATIC REVIEW article

Front. Physiol., 30 October 2018
Sec. Integrative Physiology
This article is part of the Research Topic Biology of Aging: Impactful Interventions to Extend Health-span View all 19 articles

Repurposing Proteostasis-Modifying Drugs to Prevent or Treat Age-Related Dementia: A Systematic Review

\r\nDaniel S. HeardDaniel S. Heard1Camilla S. L. TuttleCamilla S. L. Tuttle2Nicola T. Lautenschlager,Nicola T. Lautenschlager1,3Andrea B. Maier,*Andrea B. Maier2,4*
  • 1North West Mental Health, Melbourne Health, Melbourne, VIC, Australia
  • 2@AgeMelbourne, Department of Medicine and Aged Care, University of Melbourne, Melbourne, VIC, Australia
  • 3Academic Unit for Psychiatry of Old Age, Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
  • 4@AgeAmsterdam, Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands

Background: Dementia has a significant impact on quality of life of older individuals. Impaired proteostasis has been implicated as a potential cause of dementia, that can be therapeutically targeted to improve patient outcomes. This review aimed to collate all current evidence of the potential for targeting proteostasis with repurposed drugs as an intervention for age-related dementia and cognitive decline.

Methods: PubMed, Web of Science and Embase databases were searched from inception until 4th July 2017 for studies published in English. Interventional studies of repurposed proteostasis-modifying drugs in Alzheimer's disease (AD), Parkinson's disease (PD), Lewy Body disease, vascular dementia, and cognitive aging, in either animal models or humans with change in cognition as the outcome were included. The SYRCLE and Cochrane tools were used to assess risk of bias for included studies.

Results: Overall 47 trials, 38 animal and 9 human, were isolated for inclusion in this review. Drugs tested in animals and humans included lithium, rapamycin, rifampicin, and tyrosine kinase inhibitors. Drugs tested only in animals included Macrophage and Granulocyte-Macrophage Colony Stimulating Factors, methylene blue, dantrolene, geranylgeranylacetone, minocycline and phenylbutyric acid. Lithium (n = 10 animal, n = 6 human) and rapamycin (n = 12 animal, n = 1 human) were the most studied proteostasis modifying drugs influencing cognition. Nine of ten animal studies of lithium showed a statistically significant benefit in Alzheimer's models. Rapamycin demonstrated a significant benefit in models of vascular dementia, aging, and Alzheimer's, but may not be effective in treating established Alzheimer's pathology. Lithium and nilotinib had positive outcomes in human studies including Alzheimer's and Parkinson's patients respectively, while a human study of rifampicin in Alzheimer's failed to demonstrate benefit. Microdose lithium showed a strongly significant benefit in both animals and humans. While the risk of bias was relatively low in human studies, the risk of bias in animal studies was largely unclear.

Conclusion: Overall, the collective findings support the hypothesis that targeting proteostasis for treatment of dementia may be beneficial, and therefore future studies in humans with repurposed proteostasis modifying drugs are warranted. Larger human clinical trials focusing on safety, efficacy, tolerability, and reproducibility are required to translate these therapeutics into clinical practice.

Introduction

Dementias including Alzheimer's disease, vascular dementia, Parkinson's disease and Lewy body disease, have a significant impact on global health due to the increasing number of older individuals suffering from this disease (Prince et al., 2015). Developing effective methods for preventing, delaying or treating dementia are pressing priorities. The highest risk factor for dementia is chronological age, with an annual incidence of Alzheimer's disease doubling every 5 years past the age of 65 years (Bermejo-Pareja et al., 2008). Dementia subtypes share several pathological processes including abnormal accumulation of misfolded proteins such as; amyloid beta (Aβ) and tau in Alzheimer's disease, and alpha-synuclein in Parkinson's disease and Lewy Body disease (Ganguly et al., 2017). Loss of proteostasis is an important feature during the aging process (López-Otín et al., 2013), suggesting the age-related decline in the ability to refold or degrade damaged proteins may contribute to the exponential rise in dementia incidence observed with increasing age (Yerbury et al., 2016).

Several drugs already approved for their use in humans are known to enhance proteostasis including; lithium, mTOR inhibitors (sirolimus/rapamycin, everolimus), and tyrosine kinase inhibitors (nilotinib). The concept of modifying aging with a repurposed drug to prevent multiple diseases of aging will soon be tested in the Targeting Aging with Metformin (TAME) trial. TAME will examine Metformin's ability to prevent diseases of aging in non-diabetic elderly, including cognitive impairment (Barzilai et al., 2016), via targeting the deregulated nutrient sensing associated with aging. Applying a similar strategy to target the loss of proteostasis could be effective in preventing and/or treating age-related dementia.

This systematic review will examine the evidence for targeting proteostasis with repurposed drugs as an intervention for age-related dementia and cognitive decline.

Methods

Protocol Registration and Search Strategy

The protocol of this systematic review was registered at PROSPERO International prospective register of systematic reviews (Reg #: CRD42018091645). PubMed, Web of Science and Embase databases were used for this search from inception until 4th July 2017. The complete search strategy is presented in Supplementary Data 1. Key search terms included; “vascular dementia,” “Alzheimer* disease” “Lewy Body Disease,” “Parkinson* disease,” “cognitive aging,” “autophag*,” “lysosom*,” “proteasome endopeptidase complex,” “molecular chaperone*,” “unfolded protein response,” “insulin*,” “mTOR,” “GSK-3,” “akt,” “PI3K,” “AMPK,” “sirtuin*,” “sirolimus,” “everolimus,” “temsirolimus,” “rapamycin,” “metformin,” “DPP-4,” “GLP-1,” “nicotinamide,” “NAD,” “spermidine,” “imatinib,” “nilotinib,” “dasatinib,” “bosutinib,” “ponatinib,” “bafetinib,” “lithium,” “heat-shock protein,” “calori* restriction,” “carbohydrate restricted diet,” “protein restricted diet”. In addition to the database search a “snowballing” method was used to identify relevant articles out of the reference section and PubMed citations of each included article. After duplicates were removed studies were then screened for inclusion using Covidence systematic review software (Veritas Health Innovation, Melbourne, Australia).

Eligibility Criteria

Type of Studies

The search was designed to retrieve all published research studies that investigated the effect of modifying protein homeostasis or deregulated nutrient sensing (DNS) on cognitive function in age-related neurodegenerative disease and normal aging populations. To be included in this review the study had to report on one or more neuropsychological tests measuring change in cognitive function. To meet the criteria of modifying protein homeostasis or deregulated nutrient sensing, the intervention had to be previously demonstrated to modulate these pathways, or data had to be provided proving the intervention's effect on these pathways. Animal in vivo models and human trials were included in this review. The following Dementia populations/models were specifically targeted; Alzheimer's disease, Vascular Dementia, Parkinson's disease and Lewy Body Disease. In addition, normal aging populations, defined as a population not suffering from dementia and over the age of 18 years for human studies, as well as populations likely to have a higher pace of aging such as animal models with diabetes or obesity were included. Randomized controlled trials (RCTs) and non-randomized studies comparing outcomes to either retrospective or prospective controls met the inclusion criteria. Studies were excluded if they met the following criteria; observational studies, exercise as the sole intervention, in vitro data only, conference abstracts, reviews, editorials, letters to the editor, case reports with ≤5 population size, or published in a language other than English.

Outcome

In animals (using mice as an example), cognitive tests would include spatial memory tests (Morris water maze [MWM], radial arm water maze [RAWM], Barnes maze), associative learning tasks (passive avoidance, fear conditioning), alternation tasks (Y-Maze/T-Maze), recognition memory tasks (Novel Object Recognition), attentional tasks (3 and 5 choice serial reaction time), set-shifting tasks, and reversal learning tasks. In human studies examples of neuropsychological measures would be cognitive testing batteries commonly used in clinical or research settings to examine cognitive function, such as the Mini-Mental State Examination (MMSE), Rowland Universal Dementia Assessment Scale (RUDAS), Neuropsychiatry Unit Cognitive Assessment Tool (NUCOG), Montreal Cognitive Assessment (MOCA), Clinical Dementia Rating Scale Sum of Boxes (CDR-SoB), Addenbrooke's Cognitive Examination (ACE) or Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-Cog).

Study Selection

Two review authors (DH and CT) independently screened the titles and abstracts and subsequently the full text articles of potentially relevant studies against the inclusion and exclusion criteria. A third reviewer (ABM) resolved any disagreements between the authors.

Included studies were separated into the following four groups for data extraction (1) proteostasis–repurposed drug, (2) proteostasis–novel intervention (defined as a novel molecule, botanical extract, or dietary manipulation), (3) DNS–repurposed drug and (4) DNS–novel intervention. Where an intervention is thought to modify both pathways (for example the mTOR inhibitor, rapamycin) it was included in the loss of proteostasis group. The current paper presents the results of the 1st group: proteostasis—repurposed drugs.

Data Extraction and Quality Assessment

The following variables were extracted independently by two reviewers (DH and CT): author, year of publication, study design, species, animal model/population (dementia subtype or normal aging), sample size, age, sex, baseline cognition/stage of disease, duration of study, cognitive outcome, drug, comparator, setting, hallmark(s) of aging targeted by the intervention, and journal citation. For binary outcomes the number of events and total number in group, percentage of events or ratios with confidence intervals; for continuous outcomes, mean or median, standard deviation, standard error, confidence intervals or interquartile range, and number of participants; other reported results such as mean difference and p-values of measures of cognitive function.

Risk of bias was assessed by two reviewers (DH, CT) using the Cochrane Risk of Bias tool (Higgins et al., 2011) for human studies and SYRCLE's risk of bias tool for animal studies. The SYRCLE RoB tool is an adaptation of the Cochrane tool for use in systematic reviews of laboratory animal studies (Hooijmans et al., 2014).

Registered Human Trials

To establish the progress of repurposed drugs into human studies which have not yet been completed, clinicaltrials.gov was searched for registered studies of the drugs identified in our search in Alzheimer's disease, Parkinson's disease, Lewy Body Disease and Vascular Dementia.

Results

Study Selection and Characteristics

The literature search and selection process for this review is illustrated in Figure 1. After exclusion of duplicates the remaining 1,687 studies were screened for Title and Abstracts of which 413 underwent full text screening. An additional 22 studies were identified via snowballing. Overall, 47 articles specifically investigating a proteostasis intervention on Dementia and cognitive aging were included in this review. The repurposed drugs used in these intervention studies are outlined in Tables 1, 2 and Supplementary Table 1. The following drugs were found testing the modification of cognition in animal and human studies; lithium (n = 10 animal, n = 6 human), rapamycin (n = 12 animal, n = 1 human), rifampicin (n = 1 animal, n = 1 human), tyrosine kinase inhibitors (bosutinib n = 1 animal, nilotinib n = 1 human), Macrophage Colony Stimulating Factor (M-CSF; n = 1 animal), Granulocyte Macrophage Colony Stimulating Factor (GM-CSF; n = 1 animal), methylene blue (n = 4, animal), geranylgeranylacetone (GGA; n = 2 animal), dantrolene (n = 3 animal), minocycline (n = 2 animal) and phenylbutyric acid (n = 1 animal). Doxycycline was tested in a single human trial only. Of these drugs only lithium, rapamycin, rifampicin, and the tyrosine kinase inhibitors have been tested in both animal and human studies (Figure 2).

FIGURE 1
www.frontiersin.org

Figure 1. Study selection process.

TABLE 1
www.frontiersin.org

Table 1. Characteristics of animal studies testing the effect of lithium (a), rapamycin (b), rifampicin (c), bosutinib (d) on cognition.

TABLE 2
www.frontiersin.org

Table 2. Characteristics of human studies testing the effect of lithium (a), rapamycin (b), rifampicin & doxycycline (c) and nilotinib (d) on cognition.

FIGURE 2
www.frontiersin.org

Figure 2. Proteostasis Drugs and Cognitive Outcomes: From Animals to Humans. A schematic overview of trials investigating the influence of proteostasis drugs on cognitive outcomes. One bar is equal to one trial, green indicates a positive result for at least one cognitive outcome and black is indicative of no positive outcomes. X represents a registered trial. Trials are arranged in chronological order.

Lithium and Cognitive Aging

Lithium was the most investigated proteostasis modulator for cognitive aging. Overall 16 studies (6 humans, 6 mice, 2 rats, 1 drosophila, and 1 zebrafish) investigating the influence of lithium on cognitive aging were found. The majority of these studies utilized an Alzheimer's animal model or were conducted in an Alzheimer's population (Tables 1, 2). The findings of these studies were largely positive with all animal studies, bar one (Caccamo et al., 2007), reporting the use of lithium as having a statistically significant beneficial impact on at least one cognitive outcome irrespective of treatment duration (Table 3). The findings of the animal studies are consistent with the lithium human studies (Table 4), with three randomized controlled studies showing a statistically significant benefit of the use on lithium on either the ADAS-Cog or MMSE (Leyhe et al., 2009; Forlenza et al., 2011; Nunes et al., 2013). The other three studies, two of which were open-label (Pomara et al., 1983; Macdonald et al., 2008) and one RCT (Hampel et al., 2009), did not show cognitive benefits after treatment with lithium.

TABLE 3
www.frontiersin.org

Table 3. Results of animal studies testing the effect of lithium (a), rapamycin (b), rifampicin (c) and bosutinib (d) on cognition.

TABLE 4
www.frontiersin.org

Table 4. Results of human studies testing the effect of lithium (a), rapamycin (b), rifampicin & doxycycline (c) and nilotinib (d) on cognition.

Rapamycin and Cognitive Aging

Rapamycin has been identified as the second most frequently investigated (n = 13 studies) proteostasis modulator. Twelve animal studies (10 mice, 2 rats), predominately using an Alzheimer's disease model, investigated the influence of rapamycin on cognitive aging (Table 1). Overall, nine out of twelve animal studies reported a statistically significant benefit on at least one cognitive outcome, irrespective of treatment duration (Table 3). One human study investigating safety, efficacy and tolerability of rapamycin in humans (Kraig et al., 2018), did not report any significant benefit to overall cognition in an older population (Table 4).

Rifampicin, Tetracycline Antibiotics, Tyrosine Kinase Inhibitors and Cognitive Aging

Rifampicin, tetracycline antibiotics and tyrosine kinase inhibitors have been tested in both animal and human models (Tables 1, 2). The two rifampicin studies (1 mouse, 1 human; Tables 3, 4) investigating its therapeutic effects for Alzheimer disease had opposite findings, with positive cognitive outcomes in mice (Umeda et al., 2016), but no benefit found in the human study (Molloy et al., 2013).

Studies of tetracycline antibiotics showed similar results to rifampicin, with minocycline showing cognitive benefits in two studies using rat and chicken models of Alzheimer disease (Supplementary Table 1, Supplementary Table 2), while another tetracycline, doxycycline, showed no benefit in human AD patients, either alone or in combination with rifampicin (Molloy et al., 2013).

The tyrosine kinase inhibitor, bosutinib, was reported in mouse models of Alzheimer's disease as statistically beneficial to cognitive function, and another tyrosine kinase inhibitor, nilotinib, was found to improve scores on the MMSE and SCOPA-Cog in an open-label study in patients with Parkinson's disease, however the statistical significance was not reported (Tables 3, 4).

Other Proteostasis-Modifying Drugs

There were six proteostasis modulators that have been tested to improve cognitive outcomes in animal models but are yet to be studied in human populations—M-CSF, GM-CSF, methylene blue, GGA, dantrolene, and phenylbutyric acid (Supplementary Table 1). Both of the M/GM-CSF studies indicated beneficial outcomes with the use of these therapeutics in mouse models of Alzheimer's disease, as was the case with methylene blue, GGA and phenylbutyric acid (Supplementary Table 2). Studies of dantrolene to improve cognitive outcomes showed a statistically significant improvement in one mouse model of Alzheimer's (Peng et al., 2012) but no benefit to cognition in another (Wu et al., 2015), with one study in aged rats indicating a trend toward benefit on Morris water maze performance.

Risk of Bias

Table 5 shows the SYRCLE risk of bias ratings for animal studies. The majority of animal studies had an unclear risk of bias, as specific details of randomization and blinding were often not provided. Most studies provided information on the baseline characteristics of animals, and some studies did specify that the investigator performing behavioral assessments of the animals was blinded to the treatment status of the animal, indicating low risk of bias where this was the case. Overall the risk of bias was similar across studies regardless of the drug being tested.

TABLE 5
www.frontiersin.org

Table 5. SYRCLE Risk of Bias for animal studies.

Table 6 shows the Cochrane risk of bias rating for human studies. There was significant heterogeneity among lithium studies, with (Pomara et al., 1983; Macdonald et al., 2008) scoring high risk of bias across most or all domains due to an open label design and not reporting all quantitative outcome data. (Hampel et al., 2009) had an intermediate risk of bias as investigators were aware of patient treatment status. Studies by Leyhe et al. (2009), Forlenza et al. (2011) and Nunes et al. (2013) scored a lower risk of bias due to randomized, double-blind designs, though details of sequence generation and allocation concealment were not reported. Studies of rapamycin (Kraig et al., 2018) and rifampicin (Molloy et al., 2013) scored a low risk of bias across most domains. The study using nilotinib (Pagan et al., 2016) was rated as having a high risk of bias across most domains, due to an open label design.

TABLE 6
www.frontiersin.org

Table 6. Cochrane Risk of Bias for human studies.

Registered Human Trials

Figure 2 shows the progress of the drugs identified in our search from animal into human studies, including planned or ongoing studies registered on clinicaltrials.gov. Nilotinib has two phase 2 studies registered, one in Alzheimer's disease and one in Parkinson's disease. Lithium has one phase 4 study registered, while GM-CSF and phenylbutyric acid each have one phase 2 study registered in Alzheimer's disease.

Discussion

In this review we have summarized all current animal and human research studies that have investigated the effect of proteostasis modulators on cognitive function. Overall, the therapeutic alteration of proteostasis pathways using repurposed drugs is a promising approach to the treatment of dementia and age-related cognitive decline with a reasonable research translation between animal and human studies showing similar conclusions observed across the studies.

Lithium

Lithium was the most studied proteostasis modifying drug identified in this review, and the furthest progressed in translation to treatment of age-related dementia. All except one of the animal studies included in this review showed a benefit to at least one cognitive outcome despite heterogeneity in species, model, dose, stage of disease at intervention and duration of treatment. Three of six human studies also found benefit, and consistent with the results in mice the largest effect was observed with microdoses and long duration of treatment (Nunes et al., 2013). The only negative RCT used a high dose for a short duration (Hampel et al., 2009). Taken together, these studies show lithium has a consistent benefit in Alzheimer's disease in animal models and humans, which appears more pronounced with lower doses, longer durations of treatment, and when commenced at an earlier stage of disease.

The apparent superiority of lower doses of lithium is encouraging, as at standard psychiatric dose it can have renal and thyroid-related side effects that limit tolerability and is toxic at levels only slightly above the therapeutic range (Timmer and Sands, 1999). Higher lifetime exposure to natural microlevels of lithium in drinking water is associated with a reduced incidence of dementia (Kessing et al., 2017), a finding which adds plausibility to the idea that microdoses of lithium rather than the current therapeutic doses might be beneficial for the treatment of dementia.

A possible explanation for lithium's non-linear dose response involves the interaction between lithium's autophagy-enhancing inhibition of inositol triphosphate receptor (IP3R) signaling, and its autophagy-reducing inhibition of GSK-3B, a tau-phosphorylating kinase (Sarkar et al., 2008). Genetic reduction of IP3R signaling in drosophila rescues Alzheimer's disease phenotypes in the same way lithium does (McBride et al., 2010), suggesting lithium's inhibition of IP3R signaling via reducing the formation of inositol triphosphate (IP3) may be sufficient to cause its benefits. This is mechanistically plausible, as inhibition of IP3R signaling enhances autophagy through decreasing calcium release from the endoplasmic reticulum, enhancing proteostasis (Sarkar et al., 2005). However; lithium's inhibition of GSK3B has also been postulated to have beneficial effects in Alzheimer's disease via reducing tau phosphorylation and neurofibrillary tangle formation as well as inhibiting autophagy via increasing mTOR signaling (Sarkar et al., 2008). Although, this latter theory is less likely to explain the positive influence of microdoses of lithium as lower levels of lithium do not inhibit GSK-3B in mice (Nunes et al., 2015). However, this GSK-3B mechanism may explain the reduced efficacy of lithium observed with increasing doses. Increased mTOR signaling due to GSK3B inhibition counteracts the autophagy-enhancing effects of reduced IP3R activation beyond a certain dose (Sarkar et al., 2009). If increased mTOR activity limits the effective dose of lithium, this raises the question of whether an mTOR inhibitor such as rapamycin combined with lithium would have beneficial synergistic effects in dementia or cognitive aging. A synergistic benefit of lithium and rapamycin has been demonstrated in a drosophila Huntington's disease model (Sarkar et al., 2008), but to our knowledge this has not been explored in models of Alzheimer's disease.

Rapamycin

Predictably, rapamycin the known inhibitor of mTOR was also one of the most studied proteostasis modulators investigated, more so in animal models then in humans. Overall, the trials indicated a positive therapeutic effect of rapamycin on cognitive outcomes. Cognitive benefits via rapamycin was demonstrated in models of normal aging (Majumder et al., 2011, 2012; Neff et al., 2013; Wang et al., 2014), vascular dementia (Jahrling et al., 2017), and transgenic Alzheimer's disease models (Spilman et al., 2010; Majumder et al., 2011; Lin et al., 2013; Zhang et al., 2017), which suggests rapamycin has potential to rescue cognitive decline caused by a range of pathologies. This breadth of efficacy is a promising characteristic, as autopsy studies have demonstrated that mixed pathology is common in dementia sufferers (Nelson et al., 2016).

Majumder et al. (2011) reported a possible mechanism explaining their findings that rapamycin while effective in preventing cognitive decline before pathology develops, fails as a treatment once Alzheimer's pathology is established. They found rapamycin induces autophagy strongly both before and after Alzheimer's pathology is present. However, increased autophagy induction fails to reduce levels of amyloidβ in mice with established disease and leads to accumulation of enlarged autophagosomes containing undigested material. This finding suggests deficient substrate clearance and is consistent with previous findings that autophagy in Alzheimer's disease is principally defective at the stage of autolysosomal proteolysis (Nixon and Yang, 2011; Bordi et al., 2016). Therefore, to be effective in established Alzheimer's dementia rapamycin may need to be combined with a drug that can enhance autophagy at the stage of autolysosomal digestion.

Rapamycin's demonstrated ability to improve phenotypes of aging in animals has recently led to human trials assessing safety and efficacy in older adults (Kraig et al., 2018). Despite demonstrating low-dose rapamycin can be used safely in older adults, they were unable to show significant enhancement of cognition. However, larger trials are required to determine the potential benefit of rapamycin's for cognitive aging in humans.

Rifampicin, Tetracycline Antibiotics and Tyrosine Kinase Inhibitors

Rifampicin was more effective when started earlier in the disease process, and when used at a higher dose (Umeda et al., 2016). Cohorts treated with the lower dose at a later stage of disease did not show an improved cognitive benefit, a finding which is relevant to interpreting the DARAD study of rifampicin in Alzheimer's disease by Molloy et al. (2013).

In the DARAD study, rifampicin was tested in Alzheimer's patients at a low dose and showed either no benefit or on some measures a significant worsening of cognition compared to placebo. A possible explanation for this failure is an insufficient dose and treatment duration. Data supporting this view is provided by Iizuka et al. (2017), who in an observational study determined that a minimum dose rifampicin of 450 mg/day for at least 12 months was required before any cognitive improvement was observed (Iizuka et al., 2017).

Doxycycline also failed to produce benefit for human Alzheimer's disease patients in the DARAD study (Molloy et al., 2013), despite positive animal studies with closely related tetracycline antibiotic minocycline (Choi et al., 2007; Gibbs and Gibbs, 2013). It is difficult to know what implications this has for minocycline's repurposing potential, however currently there are no upcoming human studies registered on clinicaltrials.gov for either doxycycline or minocycline in dementia.

Lonskaya et al examined bosutinib in two mouse models of established Alzheimer's disease, and demonstrated statistically significant benefits after 3 weeks of treatment. Beneficial effects on cognition were also observed in human Parkinson's disease patients by Pagan et al. although the small size and open label design of the study mean the results require confirmation in larger randomized trials. These promising findings have led to significant interest in repurposing these drugs, to the extent phase 2 clinical trials are currently assessing the effect of these drugs in Alzheimer's and Parkinson's disease cohorts.

Other Proteostasis-Modifying Drugs

Three other proteostasis-modifying drugs have excited interest in translating promising animal study findings into humans—GM-CSF, methylene blue and phenylbutyric acid have registered phase 2 studies on clinicaltrials.gov to test their use in Alzheimer's disease.

Dantrolene is a drug of interest due to similarities it shares with lithium, in that it enhances autophagy by reducing calcium efflux from the endoplasmic reticulum (Wang et al., 2017). Unlike lithium however it acts by inhibition of the ryanodine receptor rather than IP3R signaling, raising the possibility of a complementary mechanism of action (Vervliet et al., 2017). This suggests dantrolene is worth testing in human trials to determine whether it can provide similar benefits to lithium, and in combination with lithium in animal models for potential synergistic effects. Currently however, no human trials are registered on clinicaltrials.gov for dantrolene in dementia.

Limitations

Our study has several limitations. Our search strategy was based primarily on key terms related to the mechanisms of proteostasis, with the addition of a selection of drugs well known to modulate these processes. Therefore, our search may have missed studies that examine proteostasis-modifying drugs not named in our search and not mentioning proteostasis related key terms. However, we addressed this by adding relevant articles by snowballing.

Secondly, because the present study's focus is restricted to approved drugs, it does not provide an adequate overview of the translational pipeline where a repurposed drug is used as the basis for novel molecules that proceed into later stage studies.

Third, we cannot exclude publication bias, particularly in animal studies which are unlikely to be registered beforehand and may be less likely to be published if results are negative.

Conclusions

The results of this review support the concept of a translational approach to repurposing proteostasis modifying drugs for the treatment of age-related dementia and cognitive decline. However, larger clinical trials assessing the influence of these drugs particularly, lithium and rapamycin are required before they are ready for the clinic. In addition, animal models assessing whether the combination of proteostasis modulators can act in synergy to improve cognitive outcomes are required. A translational strategy based on systematic screening of rational drug combinations starting in simple model organisms such as C. elegans may provide a pipeline of novel candidate therapies to advance into human studies.

Author Contributions

DH: Search strategy, screening, data extraction, drafting manuscript; CT: Search strategy screening, data extraction, drafting manuscript; NL: Search strategy, drafting manuscript; AM: Search strategy, conflict resolution, drafting manuscript.

Funding

An unrestricted grant by the University of Melbourne supported the work.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

Thanks to Patrick Condron from the Brownless Biomedical Library, University of Melbourne, for his assistance with our search strategy.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fphys.2018.01520/full#supplementary-material

References

Barzilai, N., Crandall, J. P., Kritchevsky, S. B., and Espeland, M. A. (2016). Metformin as a tool to target aging. Cell Metab. 23, 1060–1065. doi: 10.1016/j.cmet.2016.05.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Bermejo-Pareja, F., Benito-León, J., Vega, S., Medrano, M. J., and Román, G. C. (2008). Incidence and subtypes of dementia in three elderly populations of central Spain. J. Neurol. Sci. 264, 63–72. doi: 10.1016/j.jns.2007.07.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Boissonneault, V., Filali, M., Lessard, M., Relton, J., Wong, G., and Rivest, S. (2009). Powerful beneficial effects of macrophage colony-stimulating factor on beta-amyloid deposition and cognitive impairment in Alzheimer's disease. Brain 132, 1078–1092. doi: 10.1093/brain/awn331

PubMed Abstract | CrossRef Full Text | Google Scholar

Bordi, M., Berg, M. J., Mohan, P. S., Peterhoff, C. M., Alldred, M. J., Che, S., et al. (2016). Autophagy flux in CA1 neurons of Alzheimer hippocampus: Increased induction overburdens failing lysosomes to propel neuritic dystrophy. Autophagy 12, 2467–2483. doi: 10.1080/15548627.2016.1239003

PubMed Abstract | CrossRef Full Text | Google Scholar

Boyd, T. D., Bennett, S. P., Mori, T., Governatori, N., Runfeldt, M., Norden, M., et al. (2010). GM-CSF upregulated in rheumatoid arthritis reverses cognitive impairment and amyloidosis in Alzheimer mice. J. Alzheimers. Dis. 21, 507–518. doi: 10.3233/JAD-2010-091471

PubMed Abstract | CrossRef Full Text | Google Scholar

Caccamo, A., Oddo, S., Tran, L. X., and LaFerla, F. M. (2007). Lithium reduces Tau phosphorylation but not Aβ or working memory deficits in a transgenic model with both plaques and tangles. Am. J. Pathol. 170, 1669–1678. doi: 10.2353/ajpath.2007.061178

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, Y., Kim, H. S., Shin, K. Y., Kim, E. M., Kim, M., Kim, H. S., et al. (2007). Minocycline attenuates neuronal cell death and improves cognitive impairment in Alzheimer's Disease models. Neuropsychopharmacology 32, 2393–2404. doi: 10.1038/sj.npp.1301377

PubMed Abstract | CrossRef Full Text | Google Scholar

Deiana, S., Harrington, C. R., Wischik, C. M., and Riedel, G. (2009). Methylthioninium chloride reverses cognitive deficits induced by scopolamine: comparison with rivastigmine. Psychopharmacology 202, 53–65. doi: 10.1007/s00213-008-1394-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Fiorentini, A., Rosi, M. C., Grossi, C., Luccarini, I., and Casamenti, F. (2010). Lithium improves hippocampal neurogenesis, neuropathology and cognitive functions in APP mutant mice. Zars, T., ed. PLoS ONE 5:e14382. doi: 10.1371/journal.pone.0014382

PubMed Abstract | CrossRef Full Text | Google Scholar

Forlenza, O. V., Diniz, B. S., Radanovic, M., and Santos, F. S. (2011). Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br. J. Psychiatry 198, 351–356. doi: 10.1192/bjp.110.080044

PubMed Abstract | CrossRef Full Text | Google Scholar

Ganguly, G., Chakrabarti, S., Chatterjee, U., and Saso, L. (2017). Proteinopathy, oxidative stress and mitochondrial dysfunction: cross talk in Alzheimer's disease and Parkinson's disease. Drug Des. Devel. Ther. 11, 797–810. doi: 10.2147/DDDT.S130514

PubMed Abstract | CrossRef Full Text | Google Scholar

Gibbs, M. E., and Gibbs, C. L. (2013). Deleterious effects of soluble beta amyloid on cognition, antagonism by saline and noradrenaline, a role for microglia. Neuroscience 230, 62–71. doi: 10.1016/j.neuroscience.2012.10.070

PubMed Abstract | CrossRef Full Text | Google Scholar

Halloran, J., Hussong, S., Burbank, R., Podlutskaya, N., Fischer, K., Sloane, L., et al. (2012). Chronic inhibition of mTOR by rapamycin modulates cognitive and non-cognitive components of behavior throughout lifespan in mice. Neuroscience 223, 102–113. doi: 10.1016/j.neuroscience.2012.06.054

PubMed Abstract | CrossRef Full Text | Google Scholar

Hampel, H., Ewers, M., Bürger, K., Annas, P., Mörtberg, A., Bogstedt, A., et al. (2009). Lithium trial in Alzheimer's Disease: a randomized, single-blind, placebo-controlled, multicenter 10-week study. J. Clin. Psychiatry 70, 922–931.

PubMed Abstract | Google Scholar

Higgins, J. P. T., Altman, D. G., Gøtzsche, P. C., Jüni, P., Moher, D., Oxman, A. D., et al. (2011). The Cochrane Collaboration's tool for assessing risk of bias in randomised trials BMJ 343:d5928. doi: 10.1136/bmj.d5928

PubMed Abstract | CrossRef Full Text | Google Scholar

Hochgräfe, K., Sydow, A., Matenia, D., Cadinu, D., Könen, S., Petrova, O., et al. (2015). Preventive methylene blue treatment preserves cognition in mice expressing full-length pro-aggregant human Tau. Acta Neuropathol. Commun. 3:25. doi: 10.1186/s40478-015-0204-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Hooijmans, C. R., Rovers, M. M., de Vries, R. B., Leenaars, M., Ritskes-Hoitinga, M., and Langendam, M. W. (2014). SYRCLE's risk of bias tool for animal studies. BMC Med. Res. Methodol. 14:43. doi: 10.1186/1471-2288-14-43

PubMed Abstract | CrossRef Full Text | Google Scholar

Hopp, S. C., D'Angelo, H. M., Royer, S. E., Kaercher, R. M., Adzovic, L., and Wenk, G. L. (2014). Differential rescue of spatial memory deficits in aged rats by L-type voltage-dependent calcium channel and ryanodine receptor antagonism. Neuroscience 280, 10–18. doi: 10.1016/j.neuroscience.2014.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoshino, T., Suzuki, K., Matsushima, T., Yamakawa, N., Suzuki, T., and Mizushima, T. (2013). Suppression of Alzheimer's disease-related phenotypes by geranylgeranylacetone in mice. PLoS ONE 8:e76306. doi: 10.1371/journal.pone.0076306

PubMed Abstract | CrossRef Full Text | Google Scholar

Iizuka, T., Morimoto, K., Sasaki, Y., Kameyama, M., Kurashima, A., Hayasaka, K., et al. (2017). preventive effect of rifampicin on alzheimer disease needs at least 450 mg daily for 1 year: An FDG-PET follow-up study. Dement. Geriatr. Cogn. Dis. Extra. 7, 204–214. doi: 10.1159/000477343

PubMed Abstract | CrossRef Full Text | Google Scholar

Jahrling, J. B., Lin, A. L., DeRosa, N., Hussong, S. A., Van Skike, C. E., Girotti, M., et al. (2017). mTOR drives cerebral blood flow and memory deficits in LDLR-/- mice modeling atherosclerosis and vascular cognitive impairment. J. Cereb. Blood Flow Metab. 38, 58–74. doi: 10.1177/0271678X17705973

PubMed Abstract | CrossRef Full Text | Google Scholar

Kessing, L. V., Gerds, T. A., Knudsen, N. N., Jørgensen, L. F., Kristiansen, S. M., Voutchkova, D., et al. (2017). Association of lithium in drinking water with the incidence of dementia. JAMA Psychiatry 74, 1005–1010. doi: 10.1001/jamapsychiatry.2017.2362

PubMed Abstract | CrossRef Full Text | Google Scholar

Kraig, E., Linehan, L. A., Liang, H., Romo, T. Q., Liu, Q., Wu, Y., et al. (2018). A randomized control trial to establish the feasibility and safety of rapamycin treatment in an older human cohort: Immunological, physical performance, and cognitive effects. Exp. Gerontol. 105, 53–69. doi: 10.1016/j.exger.2017.12.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Leyhe, T., Eschweiler, G. W., Stransky, E., Gasser, T., Annas, P., Basun, H., et al. (2009). Increase of BDNF Serum Concentration in Lithium Treated Patients with Early Alzheimer's Disease. Journal of Alzheimer's Disease 16, 649–656. doi: 10.3233/JAD-2009-1004

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, A. L., Jahrling, J. B., Zhang, W., DeRosa, N., Bakshi, V., Romero, P., et al. (2015). Rapamycin rescues vascular, metabolic and learning deficits in apolipoprotein E4 transgenic mice with pre-symptomatic Alzheimer's disease. J. Cereb. Blood Flow Metab. 37, 217–226. doi: 10.1177/0271678X15621575

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, A. L., Zheng, W., Halloran, J. J., Burbank, R. R., Hussong, S. A., Hart, M. J., et al. (2013). Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer's disease. J. Cereb. Blood Flow Metab. 33, 1412–1421. doi: 10.1038/jcbfm.2013.82

PubMed Abstract | CrossRef Full Text | Google Scholar

Lonskaya, I., Hebron, M. L., Desforges, N. M., Franjie, A., and Moussa, C. E. (2013). Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol. Med. 5, 1247–1262. doi: 10.1002/emmm.201302771

PubMed Abstract | CrossRef Full Text | Google Scholar

López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M., and Kroemer, G. (2013). The hallmarks of aging. Cell 53, 1194–1217. doi: 10.1016/j.cell.2013.05.039

CrossRef Full Text

Macdonald, A., Briggs, K., Poppe, M., Higgins, A., Velayudhan, L., and Lovestone, S. (2008). A feasibility and tolerability study of lithium in Alzheimer's disease. Int. J. Geriatr. Psychiatry 23, 704–711. doi: 10.1002/gps.1964

PubMed Abstract | CrossRef Full Text | Google Scholar

Majumder, S., Caccamo, A., Medina, D. X., Benavides, A. D., Javors, M. A., Kraig, E., et al. (2012). Lifelong rapamycin administration ameliorates age-dependent cognitive deficits by reducing IL-1beta and enhancing NMDA signaling. Aging Cell 11, 326–335. doi: 10.1111/j.1474-9726.2011.00791.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Majumder, S., Richardson, A., Strong, R., and Oddo, S. (2011). Inducing autophagy by rapamycin before, but not after, the formation of plaques and tangles ameliorates cognitive deficits. PLoS ONE 6:e25416. doi: 10.1371/journal.pone.0025416

PubMed Abstract | CrossRef Full Text | Google Scholar

McBride, S. M., Choi, C. H., Schoenfeld, B. P., Bell, A. J., Liebelt, D. A., Ferreiro, D., et al. (2010). Pharmacological and genetic reversal of age dependent cognitive deficits due to decreased presenilin function. J. Neurosci. 30, 9510–9522. doi: 10.1523/JNEUROSCI.1017-10.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

Medina, D. X., Caccamo, A., and Oddo, S. (2011). Methylene blue reduces aβ levels and rescues early cognitive deficit by increasing proteasome activity. Brain Pathol. 21, 140–149. doi: 10.1111/j.1750-3639.2010.00430.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Molloy, D. W., Standish, T. I., Zhou, Q., Guyatt, G., DARAD Study Group. (2013). A 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. doi: 10.1002/gps.3846

PubMed Abstract | CrossRef Full Text | Google Scholar

Neff, F., Flores-Dominguez, D., Ryan, D. P., Horsch, M., Schröder, S., Adler, T., et al. (2013). Rapamycin extends murine lifespan but has limited effects on aging. J. Clin. Invest. 123, 3272–3291. doi: 10.1172/JCI67674

PubMed Abstract | CrossRef Full Text | Google Scholar

Nelson, P. T., Trojanowski, J. Q., Abner, E. L., Al-Janabi, O. M., Jicha, G. A., Schmitt, F. A., et al. (2016). “New old pathologies”: AD, PART, and cerebral age-related TDP-43 with sclerosis (CARTS). J. Neuropathol. Exp. Neurol. 75, 482–498. doi: 10.1093/jnen/nlw033

PubMed Abstract | CrossRef Full Text | Google Scholar

Nery, L. R., Eltz, N. S., Hackman, C., Fonseca, R., Altenhofen, S., Guerra, H. N., et al. (2014). Brain intraventricular injection of amyloid-β in zebrafish embryo impairs cognition and increases tau phosphorylation, effects reversed by lithium. Laks, J., ed. PLoS ONE 9:e105862. doi: 10.1371/journal.pone.0105862

PubMed Abstract | CrossRef Full Text | Google Scholar

Nixon, R. A., and Yang, D.-S. (2011). Autophagy failure in Alzheimer's disease – locating the primary defect. Neurobiol. Dis. 43, 38–45. doi: 10.1016/j.nbd.2011.01.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Nocjar, C., Hammonds, M. D., and Shim, S. S. (2007). chronic lithium treatment magnifies learning in rats. Neuroscience 150, 774–788. doi: 10.1016/j.neuroscience.2007.09.063

PubMed Abstract | CrossRef Full Text | Google Scholar

Nunes, M. A., Schöwe, N. M., Monteiro-Silva, K. C., Baraldi-Tornisielo, T., Souza, S. I., Balthazar, J., et al. (2015). Chronic Microdose Lithium Treatment Prevented Memory Loss and Neurohistopathological Changes in a Transgenic Mouse Model of Alzheimer's Disease. Holscher, C., ed. PLoS ONE 10:e0142267. doi: 10.1371/journal.pone.0142267

PubMed Abstract | CrossRef Full Text | Google Scholar

Nunes, M. A., Viel, T. A., and Buck, H. S. (2013). Microdose lithium treatment stabilized cognitive impairment in patients with Alzheimer's Disease. Curr. Alzheimer Res. 10, 104–107.

PubMed Abstract | Google Scholar

Pagan, F., Hebron, M., Valadez, E. H., Torres-Yaghi, Y., Huang, X., Mills, R. R., et al. (2016). Nilotinib effects in Parkinson's disease and dementia with lewy bodies. J. Parkinsons. Dis. 6, 503–517. doi: 10.3233/JPD-160867

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, J., Liang, G., Inan, S., Wu, Z., Joseph, D. J., Meng, Q., et al. (2012). Dantrolene ameliorates cognitive decline and neuropathology in Alzheimer triple transgenic mice. Neurosci. Lett. 516, 274–279. doi: 10.1016/j.neulet.2012.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Pomara, N., Banay-Schwartz, M., Block, R., Stanley, M., and Gershon, S. (1983). Elevation of RBC glycine and choline levels in geriatric patients treated with lithium. Am. J. Psychiatry 140, 911–913. doi: 10.1176/ajp.140.7.911

PubMed Abstract | CrossRef Full Text | Google Scholar

Prince, M., Wimo, A., Guerchet, M., Ali, G., Wu, Y., Prina, M., et al. (2015). The World Alzheimer Report 2015, The Global Impact of Dementia: An Analysis of Prevalence, Incidence, Cost and Trends. Alzheimer's Disease International. Available online at: https://www.alz.co.uk/research/world-report-2015

Rockenstein, E., Torrance, M., Adame, A., Mante, M., Bar-on, P., Rose, J. B., et al. (2007). Neuroprotective effects of regulators of the glycogen synthase kinase-3β signaling pathway in a transgenic model of alzheimer's disease are associated with reduced amyloid precursor protein phosphorylation. J. Neurosci. 27, 1981–1991. doi: 10.1523/JNEUROSCI.4321-06.2007

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarkar, S., Floto, R. A., Berger, Z., Imarisio, S., Cordenier, A., Pasco, M., et al. (2005). Lithium induces autophagy by inhibiting inositol monophosphatase. J. Cell Biol. 170, 1101–1111. doi: 10.1083/jcb.200504035

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarkar, S., Krishna, G., Imarisio, S., Saiki, S., O'Kane, C. J., and Rubinsztein, D. C. (2008). A rational mechanism for combination treatment of Huntington's disease using lithium and rapamycin. Hum. Mol. Genet. 17, 170–178. doi: 10.1093/hmg/ddm294

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarkar, S., Ravikumar, B., Floto, R. A., and Rubinsztein, D. C. (2009). Rapamycin and mTOR-independent autophagy inducers ameliorate toxicity of polyglutamine-expanded huntingtin and related proteinopathies. Cell Death Differ. 16, 46–56. doi: 10.1038/cdd.2008.110

PubMed Abstract | CrossRef Full Text | Google Scholar

Spilman, P., Podlutskaya, N., Hart, M. J., Debnath, J., Gorostiza, O., Bredesen, D., et al. (2010). Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease. PLoS ONE 5:e9979. doi: 10.1371/journal.pone.0009979

PubMed Abstract | CrossRef Full Text | Google Scholar

Stack, C., Jainuddin, S., Elipenahli, C., Gerges, M., Starkova, N., Starkov, A., et al. (2014). Methylene blue upregulates Nrf2/ARE genes and prevents tau-related neurotoxicity. Hum. Mol. Genet. 23, 3716–3732. doi: 10.1093/hmg/ddu080

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, Y., Zhang, J. R., and Chen, S. (2017). Suppression of Alzheimer's disease-related phenotypes by the heat shock protein 70 inducer, geranylgeranylacetone, in APP/PS1 transgenic mice via the ERK/p38 MAPK signaling pathway. Exp. Ther. Med. 14, 5267–5274. doi: 10.3892/etm.2017.5253

PubMed Abstract | CrossRef Full Text | Google Scholar

Sy, M., Kitazawa, M., Medeiros, R., Whitman, L., Cheng, D., Lane, T. E., et al. (2011). Inflammation induced by infection potentiates tau pathological features in transgenic mice. Am. J. Pathol. 178, 2811–2822. doi: 10.1016/j.ajpath.2011.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Timmer, R. T., and Sands, J. M. (1999). Lithium Intoxication. J. Am. Soc. Nephrol. 10, 666–674.

PubMed Abstract | Google Scholar

Toledo, E. M., and Inestrosa, N. C. (2010). Activation of Wnt signaling by lithium and rosiglitazone reduced spatial memory impairment and neurodegeneration in brains of an APPswe/PSEN1DE9 mouse model of Alzheimer's disease. Mol. Psychiatry 15, 272–285. doi: 10.1038/mp.2009.72

PubMed Abstract | CrossRef Full Text | Google Scholar

Umeda, T., Ono, K., Sakai, A., Yamashita, M., Mizuguchi, M., Klein, W. L., et al. (2016). Rifampicin is a candidate preventive medicine against amyloid-beta and tau oligomers. Brain 139(Pt 5): 1568–1586. doi: 10.1093/brain/aww042

PubMed Abstract | CrossRef Full Text | Google Scholar

Vervliet, T., Pintelon, I., Welkenhuyzen, K., Bootman, M. D., Bannai, H., Mikoshiba, K., et al. (2017). Basal ryanodine receptor activity suppresses autophagic flux. Biochem. Pharmacol. 132, 133–142. doi: 10.1016/j.bcp.2017.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., Zhou, S. L., Min, F. Y., Ma, J. J., Shi, X. J., Bereczki, E., et al. (2014). mTOR-mediated hyperphosphorylation of tau in the hippocampus is involved in cognitive deficits in streptozotocin-induced diabetic mice. Metab. Brain Dis. 29, 729–736. doi: 10.1007/s11011-014-9528-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., Li, G. J., Hu, H. X., Ma, C., Ma, D. H., Liu, X. L., et al. (2016). Cerebral mTOR signal and pro-inflammatory cytokines in Alzheimer's disease rats. Transl. Neurosci. 7, 151–157. doi: 10.1515/tnsci-2016-0022

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Shi, Y., and Wei, H. (2017). Calcium dysregulation in Alzheimer's Disease: a target for new drug development. J. Alzheimers. Dis. Parkinsonism 7:5 doi: 10.4172/2161-0460.1000374

PubMed Abstract | CrossRef Full Text | Google Scholar

Wiley, J. C., Pettan-Brewer, C., and Ladiges, W. C. (2011). Phenylbutyric acid reduces amyloid plaques and rescues cognitive behavior in AD transgenic mice. Aging Cell 10, 418–428. doi: 10.1111/j.1474-9726.2011.00680.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilson, E. N., Do Carmo, S., Iulita, M. F., Hall, H., Ducatenzeiler, A., Marks, A. R., et al. (2017). BACE1 inhibition by microdose lithium formulation NP03 rescues memory loss and early stage amyloid neuropathology. Transl. Psychiatry 7:e1190. doi: 10.1038/tp.2017.169

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Z., Yang, B., Liu, C., Liang, G., Eckenhoff, M. F., Liu, W., et al. (2015). Long-term dantrolene treatment reduced intraneuronal amyloid in aged Alzheimer triple transgenic mice. Alzheimer Dis. Assoc. Disord. 29, 184–191. doi: 10.1097/WAD.0000000000000075

PubMed Abstract | CrossRef Full Text | Google Scholar

Yerbury, J. J., Ooi, L., Dillin, A., Saunders, D. N., Hatters, D. M., Beart, P. M., et al. (2016). Walking the tightrope: proteostasis and neurodegenerative disease. J. Neurochem. 137, 489–505. doi: 10.1111/jnc.13575

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L., Wang, L., Wang, R., Gao, Y., Che, H., Pan, Y., et al. (2017). Evaluating the Effectiveness of GTM-1, Rapamycin, and Carbamazepine on Autophagy and Alzheimer Disease. Med. Sci. Monit. 23, 801–808. doi: 10.12659/MSM.898679

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, B., Yang, C., Ding, L. C., and Liu, N. (2014). 3-methyladenine, an autophagic inhibitor, attenuates therapeutic effects of sirolimus on scopolamine-induced cognitive dysfunction in a rat model. Int. J. Clin. Exp. Med. 7, 3327–3332.

PubMed Abstract | Google Scholar

Keywords: aging, alzheimer's disease, dementia, lithium, proteostasis, rapamycin

Citation: Heard DS, Tuttle CSL, Lautenschlager NT and Maier AB (2018) Repurposing Proteostasis-Modifying Drugs to Prevent or Treat Age-Related Dementia: A Systematic Review. Front. Physiol. 9:1520. doi: 10.3389/fphys.2018.01520

Received: 30 July 2018; Accepted: 09 October 2018;
Published: 30 October 2018.

Edited by:

Anis Larbi, Singapore Immunology Network (A*STAR), Singapore

Reviewed by:

Carsten Merkwirth, Ferring Research Institute, Inc., United States
Michael Petrascheck, The Scripps Research Institute, United States

Copyright © 2018 Heard, Tuttle, Lautenschlager and Maier. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Andrea B. Maier, andrea.maier@unimelb.edu.au

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.