Next Article in Journal
Platelet Concentrates in Musculoskeletal Medicine
Next Article in Special Issue
The Aggregation Conditions Define Whether EGCG is an Inhibitor or Enhancer of α-Synuclein Amyloid Fibril Formation
Previous Article in Journal
Mechanisms of Action of Cassiae Semen for Weight Management: A Computational Molecular Docking Study of Serotonin Receptor 5-HT2C
Previous Article in Special Issue
Healthy Effects of Plant Polyphenols: Molecular Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer’s Disease

1
Department of Bioinnovation Center, Buddhist Tzu Chi Medical foundation, Hualien 97002, Taiwan
2
Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan
3
Department of Neurosurgery, Hualien Tzu Chi General Hospital, Hualien 97002, Taiwan
4
Department of Pathology, Hualien Tzu Chi General Hospital, Hualien 97002, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to the work.
Int. J. Mol. Sci. 2020, 21(4), 1327; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21041327
Submission received: 14 December 2019 / Revised: 13 February 2020 / Accepted: 14 February 2020 / Published: 16 February 2020
(This article belongs to the Special Issue Natural Inhibitors of Amyloid Aggregation)

Abstract

:
Alzheimer’s disease (AD) is characterized by extracellular amyloid plaques composed of the β-amyloid peptides and intracellular neurofibrillary tangles and associates with progressive declines in memory and cognition. Several genes play important roles and regulate enzymes that produce a pathological accumulation of β-amyloid in the brain, such as gamma secretase (γ-secretase). Induced pluripotent stem cells from patients with Alzheimer’s disease with different underlying genetic mechanisms may help model different phenotypes of Alzheimer’s disease and facilitate personalized drug screening platforms for the identification of small molecules. We also discuss recent developments by γ-secretase inhibitors and modulators in the treatment of AD. In addition, small-molecule drugs isolated from Chinese herbal medicines have been shown effective in treating Alzheimer’s disease. We propose a mechanism of small-molecule drugs in treating Alzheimer’s disease. Combining therapy with different small-molecule drugs may increase the chance of symptomatic treatment. A customized strategy tailored to individuals and in combination with therapy may be a more suitable treatment option for Alzheimer’s disease in the future.

1. Introduction

Alzheimer’s disease is a prevalent neurodegenerative disease that worsens over time and causes brain lesions that can result in behavioral changes. It accounts for the largest number of dementia cases [1]. In the United States, the Alzheimer’s Association estimates approximately two new cases per year per 1000 people aged 65 to 74, 11 new cases per year per 1000 people aged 75 to 84, and 37 new cases per year per 1000 people aged 85 and older [2]. Alzheimer’s disease is characterizing by short-term memory loss accompanied by depression. Impairment of language ability leads patients to avoid difficult or complex words; the ability to recognize written characters is greatly reduced as well [3]. In the older age group (>65 years), Alzheimer’s disease is one of several dreaded neurodegenerative diseases that cause dementia. Others include frontotemporal dementia, dementia with Lewy bodies, vascular dementia, progressive supranuclear palsy, alcoholic dementia, Creutzfeldt–Jakob disease, and corticobasal degeneration (Figure 1) [4]. Alzheimer’s disease has two forms: early-onset and late-onset. Early-onset Alzheimer’s disease, also known as familial Alzheimer’s disease, occurs mainly before the age of 65 in association with hereditary genetic mutations. Late-onset Alzheimer’s disease, also known as sporadic Alzheimer’s disease, occurs after the age of 65, usually in association with environmental factors and age, and no inherited genetic mutation is known to be involved [5]. The early-onset form accounts for only about 5% of all patients with Alzheimer’s disease. However, gene expression is most directly related to early-onset Alzheimer’s disease [6]. Several gene mutations lead to increased accumulation of amyloid in the brain. The genes include amyloid precursor protein (APP), presenilin-1 (PSEN-1), and presenilin-2 (PSEN-2). The APP gene sequence is located on human chromosome 21. APP protein was found in many tissues but is concentrated mainly in neuronal synapses. The functions of APP include the formation of regulatory synapses and the expulsion of iron atoms from cells [7]. APP protein exists in many different species and is highly conserved [8]. In humans, the PSEN-1 gene sequence is located on chromosome 14, and the PSEN-2 gene sequence is located on chromosome 1. PSEN-1 and PSEN-2 are highly homologous [9]. The function of PSENs is to form a complex with APP in cells and participate in the translocation and post-synthetic processing of APP [10].
Pathologically, AD is characterized by extracellular amyloid plaques composed of the β-amyloid peptides and intracellular neurofibrillary tangles that comprise the microtubule-associated protein tau [11]. Understanding the two pathologies that lead to neuronal death has been a focus of AD research. However, the generally used cell models of AD-PC12 and SH-SY5Y require the addition of Aβ1–42 peptides with induced cytotoxicity to mimic the symptoms of the disease [12,13,14,15,16,17]. However, the amyloid plaques are not the only symptoms of AD; hence, these in vitro research models are not comprehensive. The study of AD mechanisms in a human genetic context and natural display of AD symptoms in a cell model allow the understanding of AD pathology. Induced pluripotent stem cells (iPSCs) are a pathologically relevant in vitro model for the mechanistic studies and preclinical drug discovery of AD [18].
iPSCs are pluripotent stem cells formed by the lentiviral vector transfer of four transcription factors (i.e., Oct4, Sox2, c-Myc, and Klf4) into adult cells [19]. The benefits of iPSCs are that they differentiate into different types of neuronal cells from the stem cell stage and can be used to produce forebrain acetylcholine neurons, dopaminergic progenitor cells, and Purkinje cells [20,21,22,23]. Neuronal cells derived from iPSCs exhibit electrophysiological responses. Different types of iPSC-derived neurons have been used in the study of different diseases [24,25,26]. Basal forebrain cholinergic neurons degenerate in AD, Down syndrome, and dementia. Injectable acetylcholine agonists improve memory degradation in patients with AD [27]. Therefore, acetylcholine cells play a potentially regulatory role in memory and learning in the central nervous system [28]. The platelet-derived growth factor promoter driven amyloid precursor protein (PDAPP) transgenic mice exhibited amyloid deposition in the brain and memory dysfunction.
The cholinergic neurons derived from human iPSCs are transplanted to the hippocampus of PDAPP transgenic mice. Compared with vehicle-injected PDAPP mice, the transplantation of iPSC-derived differentiated neuronal precursor cells to PDAPP mice significantly improved memory dysfunction [29]. Purkinje cells are the only neurons in the cerebellar cortex that automatically produce two types of action potentials [30]. Damage to Purkinje cells is related to cognitive impairment and motor deficits in patients with AD [31]. The iPSC-induced increase in the number of Purkinje cell bodies is a potential novel strategy for AD treatment [22,32].
The AD cell model is useful for testing potential therapeutic drugs. Its use will increase the possibility of identifying an effective AD treatment through the collection of cells from patients with AD or Down syndrome and their reversal to iPSCs. The cells that have differentiated into neuronal cells and produce AD symptoms render them useful for exploring whether drugs are effective in the inhibition of these symptoms [33]. The use of iPSCs from patients with AD who have familial gene mutations will increase the possibility of developing effective therapeutic drugs, particularly those targeting the mechanisms of Aβ accumulation [34]. For example, a recent study using an iPSC platform exclusively derived from patients with AD was effective in the identification of small molecules and compounds with synergistic anti-Aβ effects [35]. Moreover, the collection of different early-onset AD gene mutations can produce different iPSCs based on AD symptoms. These approaches are helpful for understanding the mechanisms of AD and for developing drugs for this disease (Table 1).

2. Pluripotent Stem Cells Induced by AD Gene Mutations

2.1. APP Gene Mutations

Early-onset APP gene-mutated patient cells were used to produce a model of iPSCs overexpression, including a double mutation (KM670/671NL) of APP, which increases the total Aβ burden [36], followed by APP duplication to prepare iPSCs from patients with late-AD onset. The results revealed significantly higher levels of Aβ 1–40, phospho-tau (Thr 231), and active glycogen synthase kinase-3β (aGSK-3β) [37]. Interestingly, APP (A673T) mutations are protective against AD and cognitive decline [38].

2.2. Trisomy 21 Gene Mutation

Trisomy 21 is caused by the presence of an extra copy of the APP gene and is associated with increased Aβ peptide production. The accumulation of Aβ peptides is associated with a high risk of AD dementia in patients with Down syndrome [39]. iPSCs from individuals with trisomy 21 can induce Aβ aggregation, which causes increased expression of the tau protein and affects synapse formation and functionality [40].

2.3. PSEN1/2 Gene Mutation

Mutations of PSEN1 and PSEN2, which are catalytic components of γ-secretase, are causative factors of autosomal dominant early-onset familial AD. Patients with familial AD have mutations in PSEN1 (A246E) and PSEN2 (N141I). Differentiated neurons derived from iPSCs of patients with familial AD exhibit increased toxic Aβ42 secretion [41]. Differentiated neurons derived from iPSCs with PSEN1 (ΔE9) mutations can exhibit increased Aβ production, altered cytokine release, and deregulation of Ca2+ homeostasis [42].

2.4. Microtubule-Associated Protein Tau Gene Mutation

Microtubule-associated protein tau (MAPT) gene mutations are among the most common causes of familial frontotemporal dementia. MAPT (p.A152T) has been linked with frontotemporal dementia and Alzheimer’s disease. iPSCs from individuals carrying the tau-A152T variant of MAPT neurons showed accumulation, redistribution, and decreased solubility of tau. Upregulation of tau was coupled with enhanced stress-inducible markers and vulnerability of cells to proteotoxic, excitotoxic, and mitochondrial stressors [43]. iPSCs with MAPT mutations N279K, P301L, and E10 + 16 had neuritis outgrowth deficiencies [44]. The iPSCs were prepared separately, and their phenotypes manifested accumulations of tau protein, mitochondrial dysfunction, and defects of nerve growth and differentiation [45].

3. γ-secretase Inhibitors and γ-Secretase Modulators

Mutations in the PSEN-1 gene account for approximately 50% of the pathogenesis of early onset of AD and are the most common gene mutations in this disease [47]. Although gene mutations are responsible for early onset of AD, 95% of AD cases are late-onset diseases and are not necessarily caused by genetic mutations [48]. Late-onset AD can be caused by mutations in the PSEN-1 gene; such mutations account for 25% of cases of late-onset AD [49]. The PSEN-1 gene is an important regulator of γ-secretase in early- and late-onset AD (Figure 2). γ-secretase cleaves >90 different substrates. Studies of the γ-secretase-mediated cleavage of APP and Notch have focused on AD and cancer [50]. Complete inhibition of γ-secretase, as achieved by nonselective γ-secretase inhibitors (GSIs), abolishes all γ-secretase activity, leading to decreased Aβ production and Notch signaling. A recent publication used cryo-electron microscopy to elucidate the structures of the γ-secretase basis of the APP-C83 fragment and the structures of the γ-secretase basis of the Notch 100 fragment [51]. This will help in the development of substrate-specific γ-secretase inhibitors or modulators. Therefore, we discuss the development of GSIs and γ-secretase modulators (GSMs) in clinical settings.

3.1. γ-Secretase Inhibitors

Most of the mutations in the PSEN-1 gene are single nucleotide substitution mutations, which are missense mutations. PSEN-1 mutations cause an increase in the production of amyloid-β protein (~90% 40-residue amyloid beta-protein and ~10% 42-residue amyloid beta-protein) [52,53,54]. PSEN-1 is one of the components of γ-secretase [55]. The drugs’ development strategies are through inhibition cleaved by γ-secretase and reducing the production of amyloid-β protein [56,57]. However, the mechanism to treat Alzheimer’s disease led to no improvement with cognitive function and accompanying adverse side effects led to failure [58]. Despite the initial failure of GSIs to treat Alzheimer’s disease successfully, these inhibitors are still considered to have potential. γ-secretase suppresses the expression of the intracellular domain of the Notch protein (NICD), which represents a failure of this strategy, because the Notch protein plays a key role in the function of adult neural stem cells [59], affecting the differentiation and proliferation of neurons [60]. Development of GSIs must account for the effect on the Notch signals (Figure 3), and GSIs can produce cell toxicity by increasing the levels of APP carboxy-terminal fragments (β-CTF) [61].

3.1.1. Semagacestat

Semagacestat (LY-450139) (Figure 4a) was the first GSI to enter a phase III trial for AD. The compounds showed beta-amyloid-lowering effects in the in vitro assays [62]. Semagacestat is a γ-secretase inhibitor for Aβ42, Aβ40, and Aβ38 with IC50 of 10.9, 12.1, and 12.0 nM, respectively. It also inhibits Notch signaling with an IC50 of 14.1 nM in H4 human glioma cells [61]. The Tg2576 mouse model is an in vivo AD model. This model carries the APP KM670/671NL mutations. Oral administration of semagacestat (1 mg/kg) to 5.5-month-old APP-transgenic Tg2576 mice significantly improved the memory deficits [63]. Recently, reports have suggested that semagacestat is a pseudo-inhibitor of γ-secretase [64]. A phase III trial showed that semagacestat did not improve cognitive status and those patients receiving the higher dose exhibited significant deterioration of their functional ability. Semagacestat has more adverse effects, including cancers and skin infections (clinical trial number NCT00594568) [65].

3.1.2. Avagacestat

Avagacestat (BMS-708163) (Figure 4b) was expected to enter a clinical phase II trial for AD. Avagacestat is a γ-secretase inhibitor of Aβ40 and Aβ42 with IC50 of 0.3 and 0.27 nM, respectively, and exhibits 193-fold selectivity against Notch in H4 human neuroglioma cells [66]. In vitro assays revealed that avagacestat reduced CSF Aβ levels without causing Notch-related toxicities [67]. The drug is administered orally to rats and dogs in the in vivo studies, and it significantly reduced Aβ40 levels for sustained periods in the brain, plasma, and cerebrospinal fluid [66]. A phase II clinical trial revealed that avagacestat was not effective (clinical trial number NCT00890890) [68].
γ-Secretase inhibitors (GSIs) block the γ-secretase-mediated cleavage of APP. However, these GSIs also block the γ-secretase-mediated cleavage of other substrates, including the critical signaling molecule Notch. For these reasons, drugs that do not inhibit the Notch signaling cleavage of γ-secretase modulators (GSMs) may be candidate treatments for AD [69]. Given the amyloid hypothesis for Alzheimer’s disease and ensuing cognitive decline, several treatments, including anti-beta-amyloid (Aβ) small organic molecules, antigens, and monoclonal antibodies, have been developed to interfere with Aβ production or increase clearance, including β-site beta-secretase (BACE) inhibitors, to block the first enzymatic step of the formation of Aβ or GSIs. However, these clinical trials targeting amyloid plaques of Alzheimer’s disease have failed in patients with mild-to-moderate Alzheimer’s disease [70]. GSIs have problems about leading to critical issues of Notch-related neuron toxicity [57,58]. Therefore, GSMs may be a better and safer strategy for drug development against AD. GSMs can act through an allosteric binding site to increase the concentrations of the shorter non-Aβ 1-42/Aβ 1-40 peptides.

3.2. γ-secretase modulators

GSMs differ from GSIs in that they do not alter total Aβ production, do not increase APP, and do not alter the generation of γ-secretase substrates but rather decrease Aβ1–42 peptides and increase Aβ1–38 peptides (Figure 5) [71]. Nonsteroidal anti-inflammatory drugs (NSAIDs), such as aspirin, is commonly used for pain and inflammation in adults. NSAIDs selectively reduce the formation of Aβ1-42 without inhibiting the proteolysis of Notch1. The first-generation GSMs include R-flurbiprofen and indomethacin (Figure 4c–g) [72].

3.2.1. R-flurbiprofen (Tarenflurbil, MPC-7869)

R-flurbiprofen was expected to enter a clinical phase III AD trial (clinical trial number NCT00105547). R-flurbiprofen has functioned based on downregulation by neuronal tryptophan 2, 3-dioxygenase (TDO2). TDO2 expression was increased by neuronal cyclooxygenase-2 (COX-2) activities. The overexpression of TDO2 in the hippocampus led to behavioral deficits. R-flurbiprofen has prevented behavioral deficits in APPSwe-PS1ΔE9 mice [73]. In clinical trials of patients with Alzheimer’s disease; R-flurbiprofen had low potency and brain penetration and was considered an unsuccessful treatment [74,75]. The shortcomings of the drug were not improvement by changes in the drug delivery route or the development of nanoparticle carriers [76].

3.2.2. Indomethacin

Indomethacin was expected to enter a clinical phase III AD trial (clinical trial number NCT00432081). Indomethacin showed more inhibitory effects against COX-1 than against COX-2 [77]. Indomethacin attenuates neuronal pathological changes and cognitive impairments in APP/PS1 transgenic mice [78]. However, a clinical trial revealed that indomethacin did not improve the slow cognitive decline in patients with AD [79].
First-generation GSMs have low potency and inefficient blood–brain barrier penetrance, which affects the treatment of AD [80]. In second-generation GSMs, which include EVP-0015962, BIIB042 exhibited improved brain penetration ability [81]. GSM treatment of Alzheimer’s disease is considered feasible. BPN-15606 is a highly potent GSM that is a candidate for human clinical trials [82].

3.2.3. EVP-0015962

EVP-0015962 (Figure 4e) decreased Aβ1–42 peptides and increased the shorter Aβ1–38 peptides, thus lowering Aβ42 in the H4 cell model. The total levels of Aβ and the amyloid precursor protein were not changed. In vivo, EVP-0015962 decreased memory deficits and reduced Aβ plaque formation in Tg2576 transgenic animals [83]. EVP-0015962 will enter a clinical phase II trial for the treatment of early AD onset (clinical trial number NCT01661673).

3.2.4. BIIB042

BIIB042 (Figure 4f) is a potent γ-secretase modulator that lowered Aβ42, increased Aβ38, and did not affect Notch signaling in an in vitro model [84]. Tg2576 mice overexpress the human amyloid precursor protein and serve as a research model for AD. BIIB042 reduced Aβ42 levels and Aβ plaque burden in Tg2576 mice [85]. BIIB042 is a candidate drug for AD.

3.2.5. BPN-15606

BPN-15606 (Figure 4g) inhibited the production of the Aβ1–42 and Aβ1–40 peptides, while facilitating the concomitant increase in Aβ1–38 and Aβ1–37 peptides production without inhibiting the γ-secretase-mediated proteolysis of Notch. BPN-15606 can lower Aβ42 levels significantly in the central nervous system of rats and mice [82]. Recent reports indicate that BPN-15606 is effective when administered prior to the severe pathological manifestations of AD [86].

4. Small-Molecular Combination GSM Treatments of Alzheimer’s Disease

GSMs have the opportunity to become candidate drugs for the treatment of AD, but a combined treatment with drugs involving different mechanisms might provide additional efficacy for ameliorating cognitive impairment in Alzheimer’s disease. Many Chinese herbal medicines are considered to have the effect of treating dementia. We list small-molecule drugs extracted from commonly used Chinese herbal medicines and their mechanisms of treating Alzheimer’s disease (Table 2). The GSM structure is shown in Figure 6.

4.1. Icariside

Icariside is an extract from Herba epimedii, an ancient herbal medicine. Aggregates of Aβ are the agents responsible for disease progression in Alzheimer’s disease, and the accumulation of these aggregates is toxic to the brain. The major regulatory alpha-secretase in neurons, A disintegrin and metalloproteinase 10 (ADAM10), are responsible for cleaving APP. This cleavage generates a neuroprotective APP-derived short fragment (C83) and attenuates the production of Aβ peptides [87]. The mechanism of icariside in the treatment of Alzheimer’s disease involves increased ADAM10 expression and decreased expression of both APP and BACE1, resulting in reduced production of Aβ [88].

4.2. Onjisaponin

Onjisaponin B (OB) decreases Aβ by degradation of APP without direct inhibition of BACE1 and γ-secretase activities. In vivo, oral administration of OB mitigates Aβ pathology and behavioral defects in APP/PS1 mice [89,90]. OB has effects of reduced IL-1β, IL-6, TNF-α, and malondialdehyde in the serum and hippocampus of lipopolysaccharide-induced cognitive-deficit rats. OB have beneficial effects by the antioxidant, anti-inflammatory, and anti-apoptosis in the lipopolysaccharide-induced PC12 cells [91].

4.3. Bajijiasu

Bajijiasu can inhibit neuronal inflammation, increase the levels of insulin degradation enzymes and neprilysin, and decrease BACE1 expression [92]. Bajijiasu have a function of neuron-protectiveness against Aβ-induced neurotoxicity in PC12 cells [93]. Animal studies also found the same function. In APP/PS1 mice models, oral of bajijiasu improved learning and memory ability. The effects of bajijiasu have protected neurons against apoptosis, elevated the expression of neurotrophic factors, and alleviated endoplasmic reticulum stress [94].

4.4. Asarones

Asarones, the active constituents of Acori Tatarinowii Rhizoma, can improve the proliferation and self-renewal of neural progenitor cells. A mechanistic study showed that asarones activated extracellular signal-regulated kinase (ERK), part of a critical kinase cascade for neurogenesis [95]. Asarone have decreased Aβ deposition in the cortex and hippocampus of APP/PS1 mice brains [96].

4.5. Catalpol

There is a significant positive correlation between brain-derived neurotropic factor (BDNF) content and memory. Catalpol can improve memory and reduce neurodegeneration by increasing BDNF expression [97]. In vitro, catalpol has been able to reduce the levels of soluble Aβ40 and Aβ42 in the cerebral cortex, and thus, inhibit the formation of senile plaques [98].

4.6. Liquiritin

Liquiritin have been extracted from Glycyrrhiza uralensis. Oxidative stress plays an important role in the pathogenesis of Alzheimer’s disease. Histopathological hallmarks of Alzheimer’s disease include formation of senile plaques composed of the Aβ peptides. Aβ can aggregate in formation with metal ions to catalyze production of reactive oxygen species [99]. Excessive oxidative stresses are generally regarded as a pathological condition. Malondialdehyde (MDA) and 8-hydroxy-2′deoxyguanosine (8-OHdG) are products of lipid peroxidation and are the most commonly detected biomarkers of oxidative stress [100]. Liquiritin reduces MDA and 8-OHdG in the hippocampus of rats with Alzheimer’s disease and, through related mechanisms, can significantly improve spatial learning and memory [101].

4.7. Glabridin

Deterioration of cholinergic innervation in the human cerebral cortex leads to significant cognitive and behavioral disorders in Alzheimer’s disease [102]. Glabridin, a small-molecule drug studied as a potential treatment for Alzheimer’s disease, has anticholinesterase activity. Glabridin reduced the brain cholinesterase activity and memory improvement in amnesia mice. The U.S. Food and Drug Administration approved cholinesterase inhibitors for the treatment of Alzheimer’s disease [103].

4.8. Tanshinone IIA and Cryptotanshinone

Tanshinone IIA (TIIA) and cryptotanshinone (CT) are extracted from Salviae miltiorrhizae as major lipophilic compounds. In vitro, TIIA and CT significantly reduced intracellular NO and Ca2+ levels in H9c2 cells [104]. The gliosis-related and neuro-inflammatory markers in the hippocampal tissues revealed a remarkable reduction in the expression of glial fibrillary acidic protein (GFAP), S100β protein, COX-2, inducible nitric oxide synthase (iNOS), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kBp65) after CRY treatment. TIIA and CT display anti-inflammatory and neuroprotective effects in a nongenetic mouse model of AD [105].

4.9. Ginsenoside Rg1

Ginsenoside Rg1 have been extracted from Panax ginseng. Ginsenoside Rg1 repaired hippocampal long-term potentiation and memory in an Alzheimer’s disease mouse model. Through a mechanism that includes increased activity of the antioxidant enzymes glutathione peroxidase and superoxide dismutase and reduces astrocyte activation, it can increase the proliferation of hippocampal cells [106,107].

4.10. Astragaloside IV

Peroxisome proliferator-activated receptor (PPAR) agonists inhibit the expression of interleukin 6, tumor necrosis factor α, and cyclooxygenase 2 in microglial cells and monocytes stimulated by Aβ and reduce the proinflammatory and neurotoxic substances that these cells produce [108]. Astragaloside IV (AS-IV), derived from Astragalus membranaceus, is a natural PPARγ agonist that suppresses the activity of BACE1 and reduces the generation of Aβ. Activation of PPARγ in microglia by PPAR may be of value in the treatment of Alzheimer’s disease and associated inflammatory diseases [109].

4.11. n-Butylidenephthalide

n-Butylidenephthalide (n-BP) is derived from Angelica sinensis. It has been applied to the treatment of neurodegenerative diseases, including Parkinson’s disease [110], amyotrophic lateral sclerosis [111], and Alzheimer’s disease [40]. n-BP was shown to reduce Aβ, total tau protein and its hyperphosphorylated form, and cellular toxicity in iPSCs-derived neurons induced by Down syndrome [40].

5. Conclusions

Recent studies report tau pathology and neurodegeneration are enhanced, but amyloid pathology is generally unaffected by concurrent tau pathology [113,114,115]. Through iPSCs, models get verified [116]. Using iPSCs that naturally produce Alzheimer’s disease symptoms to explore the mechanisms of small-molecule drugs and combining this approach with GSM or other therapies is a strategy for developing Alzheimer’s drugs. At the same time, using an iPSC platform for drug screening may be a good way to develop personalized medicine. Individual differences are perhaps the main cause of setbacks to the development of Alzheimer’s drugs. Establishing the patient’s iPSC to confirm the symptoms first and then accurately selecting the appropriate therapeutic drug may be a good way to provide effective treatment to patients while reducing the rate of failure of drug development. Through cryo-electron microscopy (cryo-EM) analysis, the structures for γ-secretase acting on the APP-C83 fragment and Notch-100 fragment will help develop substrate-specificity secretase inhibitors or modulators. Many small molecules extracted from Chinese herbal medicines, such as Curcumin, Asarones, and Astragaloside, have been used for the treatment of dementia [117,118,119]. However, these treatments have not achieved the expected results; thus, combinations of drugs with different mechanisms may be necessary. An increase in the number of choices may increase the chances of success in fighting Alzheimer’s disease (Figure 7).

Funding

Buddhist Tzu Chi Medical Foundation TCMF-SP 108-03(108).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

ADAlzheimer’s disease
ADAMA disintegrin and metalloproteinase
APPAmyloid precursor protein
AS-IVAstragaloside IV
BACEβ-site beta-secretase
BDNFBrain-derived neurotropic factor
cryo-EMCryo-electron microscopy
COX-2Cyclooxygenase-2
CTCryptotanshinone
DLBDementia with Lewy bodies
ERKExtracellular signal-regulated kinase
FTLDFrontotemporal dementia
GSIγ-secretase inhibitors
GSMsγ-secretase modulators
GFAPGlial fibrillary acidic protein
iNOSInducible nitric oxide synthase
MAPTMicrotubule-associated protein tau
MCIMild cognitive impairment
MDAMalondialdehyde
NICDNotch intracellular domains
NSAIDsNonsteroidal anti-inflammatory drugs
NF-kBp65Nuclear factor kappa-light-chain-enhancer of activated B cells
n-BPn-Butylidenephthalide
OBOnjisaponin B
PSEN-1Presenilin-1
PSEN-2Presenilin-2
PDAPPPDGF promoter driven amyloid precursor protein
PPARPeroxisome proliferator-activated receptor
SCDSubjective cognitive decline
TIIATanshinone IIA
VaDVascular dementia
8-OHdG8-hydroxy-2′deoxyguanosine

References

  1. Van der Flier, W.M.; Scheltens, P. Amsterdam Dementia Cohort: Performing Research to Optimize Care. J. Alzheimers Dis. 2018, 62, 1091–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Association, A.S. 2019 Alzheimer’s disease facts and figures. Alzheimers Dement. 2019, 15, 321–387. [Google Scholar] [CrossRef]
  3. Forstl, H.; Kurz, A. Clinical features of Alzheimer’s disease. Eur. Arch. Psychiatry Clin. Neurosci. 1999, 249, 288–290. [Google Scholar] [CrossRef] [PubMed]
  4. Mahalingam, S.; Chen, M.K. Neuroimaging in Dementias. Semin Neurol. 2019, 39, 188–199. [Google Scholar] [CrossRef]
  5. Koedam, E.L.; Lauffer, V.; van der Vlies, A.E.; van der Flier, W.M.; Scheltens, P.; Pijnenburg, Y.A. Early-versus late-onset Alzheimer’s disease: more than age alone. J. Alzheimers Dis. 2010, 19, 1401–1408. [Google Scholar] [CrossRef] [Green Version]
  6. Mendez, M.F. Early-onset Alzheimer’s disease: Nonamnestic subtypes and type 2 AD. Arch. Med. Res. 2012, 43, 677–685. [Google Scholar] [CrossRef] [Green Version]
  7. Duce, J.A.; Tsatsanis, A.; Cater, M.A.; James, S.A.; Robb, E.; Wikhe, K.; Leong, S.L.; Perez, K.; Johanssen, T.; Greenough, M.A.; et al. Iron-export ferroxidase activity of beta-amyloid precursor protein is inhibited by zinc in Alzheimer’s disease. Cell 2010, 142, 857–867. [Google Scholar] [CrossRef] [Green Version]
  8. De Strooper, B. Loss-of-function presenilin mutations in Alzheimer disease. Talking Point on the role of presenilin mutations in Alzheimer disease. EMBO Rep. 2007, 8, 141–146. [Google Scholar] [CrossRef] [Green Version]
  9. Bekris, L.M.; Yu, C.E.; Bird, T.D.; Tsuang, D.W. Genetics of Alzheimer disease. J. Geriatr Psychiatry Neurol. 2010, 23, 213–227. [Google Scholar] [CrossRef] [Green Version]
  10. Chow, V.W.; Mattson, M.P.; Wong, P.C.; Gleichmann, M. An overview of APP processing enzymes and products. Neuromolecular Med. 2010, 12, 1–12. [Google Scholar] [CrossRef] [Green Version]
  11. Bloom, G.S. Amyloid-beta and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol 2014, 71, 505–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Westerink, R.H.; Ewing, A.G. The PC12 cell as model for neurosecretion. Acta Physiol (Oxf) 2008, 192, 273–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Jiang, X.; Kumar, M.; Zhu, Y. Protective Effect of Hyperforin on beta Amyloid Protein Induced Apoptosis in PC12 Cells and Colchicine Induced Alzheimer’s Disease: An Anti-oxidant and Anti-inflammatory Therapy. J. Oleo Sci. 2018, 67, 1443–1453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Biedler, J.L.; Helson, L.; Spengler, B.A. Morphology and growth, tumorigenicity, and cytogenetics of human neuroblastoma cells in continuous culture. Cancer Res. 1973, 33, 2643–2652. [Google Scholar]
  15. Pascual-Caro, C.; Berrocal, M.; Lopez-Guerrero, A.M.; Alvarez-Barrientos, A.; Pozo-Guisado, E.; Gutierrez-Merino, C.; Mata, A.M.; Martin-Romero, F.J. STIM1 deficiency is linked to Alzheimer’s disease and triggers cell death in SH-SY5Y cells by upregulation of L-type voltage-operated Ca(2+) entry. J. Mol. Med. (Berl) 2018, 96, 1061–1079. [Google Scholar] [CrossRef] [Green Version]
  16. Ng, J.; Kaur, H.; Collier, T.; Chang, K.; Brooks, A.E.S.; Allison, J.R.; Brimble, M.A.; Hickey, A.; Birch, N.P. Site-specific glycation of Abeta1-42 affects fibril formation and is neurotoxic. J. Biol. Chem. 2019, 294, 8806–8818. [Google Scholar] [CrossRef]
  17. Galante, D.; Corsaro, A.; Florio, T.; Vella, S.; Pagano, A.; Sbrana, F.; Vassalli, M.; Perico, A.; D’Arrigo, C. Differential toxicity, conformation and morphology of typical initial aggregation states of Abeta1-42 and Abetapy3-42 beta-amyloids. Int. J. Biochem. Cell Biol. 2012, 44, 2085–2093. [Google Scholar] [CrossRef]
  18. Arber, C.; Lovejoy, C.; Wray, S. Stem cell models of Alzheimer’s disease: Progress and challenges. Alzheimers Res. Ther. 2017, 9, 42. [Google Scholar] [CrossRef]
  19. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [Green Version]
  20. Shimojo, D.; Onodera, K.; Doi-Torii, Y.; Ishihara, Y.; Hattori, C.; Miwa, Y.; Tanaka, S.; Okada, R.; Ohyama, M.; Shoji, M.; et al. Rapid, efficient, and simple motor neuron differentiation from human pluripotent stem cells. Mol. Brain 2015, 8, 79. [Google Scholar] [CrossRef] [Green Version]
  21. Kikuchi, T.; Morizane, A.; Doi, D.; Magotani, H.; Onoe, H.; Hayashi, T.; Mizuma, H.; Takara, S.; Takahashi, R.; Inoue, H.; et al. Human iPS cell-derived dopaminergic neurons function in a primate Parkinson’s disease model. Nature 2017, 548, 592–596. [Google Scholar] [CrossRef] [PubMed]
  22. Watson, L.M.; Wong, M.M.K.; Vowles, J.; Cowley, S.A.; Becker, E.B.E. A Simplified Method for Generating Purkinje Cells from Human-Induced Pluripotent Stem Cells. Cerebellum 2018, 17, 419–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Crompton, L.A.; Byrne, M.L.; Taylor, H.; Kerrigan, T.L.; Bru-Mercier, G.; Badger, J.L.; Barbuti, P.A.; Jo, J.; Tyler, S.J.; Allen, S.J.; et al. Stepwise, non-adherent differentiation of human pluripotent stem cells to generate basal forebrain cholinergic neurons via hedgehog signaling. Stem Cell Res. 2013, 11, 1206–1221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Muratore, C.R.; Srikanth, P.; Callahan, D.G.; Young-Pearse, T.L. Comparison and optimization of hiPSC forebrain cortical differentiation protocols. PLoS ONE 2014, 9, e105807. [Google Scholar] [CrossRef] [PubMed]
  25. Stanslowsky, N.; Haase, A.; Martin, U.; Naujock, M.; Leffler, A.; Dengler, R.; Wegner, F. Functional differentiation of midbrain neurons from human cord blood-derived induced pluripotent stem cells. Stem Cell Res. Ther. 2014, 5, 35. [Google Scholar] [CrossRef] [Green Version]
  26. Wang, S.; Wang, B.; Pan, N.; Fu, L.; Wang, C.; Song, G.; An, J.; Liu, Z.; Zhu, W.; Guan, Y.; et al. Differentiation of human induced pluripotent stem cells to mature functional Purkinje neurons. Sci. Rep. 2015, 5, 9232. [Google Scholar] [CrossRef] [Green Version]
  27. Callahan, P.M.; Bertrand, D.; Bertrand, S.; Plagenhoef, M.R.; Terry, A.V., Jr. Tropisetron sensitizes alpha7 containing nicotinic receptors to low levels of acetylcholine in vitro and improves memory-related task performance in young and aged animals. Neuropharmacology 2017, 117, 422–433. [Google Scholar] [CrossRef]
  28. Chien, M.Y.; Chuang, C.H.; Chern, C.M.; Liou, K.T.; Liu, D.Z.; Hou, Y.C.; Shen, Y.C. Salvianolic acid A alleviates ischemic brain injury through the inhibition of inflammation and apoptosis and the promotion of neurogenesis in mice. Free Radic. Biol. Med. 2016, 99, 508–519. [Google Scholar] [CrossRef]
  29. Fujiwara, N.; Shimizu, J.; Takai, K.; Arimitsu, N.; Saito, A.; Kono, T.; Umehara, T.; Ueda, Y.; Wakisaka, S.; Suzuki, T.; et al. Restoration of spatial memory dysfunction of human APP transgenic mice by transplantation of neuronal precursors derived from human iPS cells. Neurosci Lett. 2013, 557 Pt B, 129–134. [Google Scholar] [CrossRef]
  30. Tang, T.; Xiao, J.; Suh, C.Y.; Burroughs, A.; Cerminara, N.L.; Jia, L.; Marshall, S.P.; Wise, A.K.; Apps, R.; Sugihara, I.; et al. Heterogeneity of Purkinje cell simple spike-complex spike interactions: zebrin- and non-zebrin-related variations. J. Physiol. 2017, 595, 5341–5357. [Google Scholar] [CrossRef] [Green Version]
  31. Mavroudis, I.A.; Manani, M.G.; Petrides, F.; Petsoglou, K.; Njau, S.D.; Costa, V.G.; Baloyannis, S.J. Dendritic and spinal pathology of the Purkinje cells from the human cerebellar vermis in Alzheimer’s disease. Psychiatr Danub 2013, 25, 221–226. [Google Scholar] [PubMed]
  32. Jiang, Y.Q.; Wang, X.L.; Cao, X.H.; Ye, Z.Y.; Li, L.; Cai, W.Q. Increased heat shock transcription factor 1 in the cerebellum reverses the deficiency of Purkinje cells in Alzheimer’s disease. Brain Res. 2013, 1519, 105–111. [Google Scholar] [CrossRef] [PubMed]
  33. Tcw, J. Human iPSC application in Alzheimer’s disease and Tau-related neurodegenerative diseases. Neurosci Lett. 2019, 699, 31–40. [Google Scholar] [CrossRef] [PubMed]
  34. Mungenast, A.E.; Siegert, S.; Tsai, L.H. Modeling Alzheimer’s disease with human induced pluripotent stem (iPS) cells. Mol. Cell Neurosci. 2016, 73, 13–31. [Google Scholar] [CrossRef] [Green Version]
  35. Kondo, T.; Imamura, K.; Funayama, M.; Tsukita, K.; Miyake, M.; Ohta, A.; Woltjen, K.; Nakagawa, M.; Asada, T.; Arai, T.; et al. iPSC-Based Compound Screening and In Vitro Trials Identify a Synergistic Anti-amyloid beta Combination for Alzheimer’s Disease. Cell Rep. 2017, 21, 2304–2312. [Google Scholar] [CrossRef] [Green Version]
  36. Oksanen, M.; Hyotylainen, I.; Voutilainen, J.; Puttonen, K.A.; Hamalainen, R.H.; Graff, C.; Lehtonen, S.; Koistinaho, J. Generation of a human induced pluripotent stem cell line (LL008 1.4) from a familial Alzheimer’s disease patient carrying a double KM670/671NL (Swedish) mutation in APP gene. Stem Cell Res. 2018, 31, 181–185. [Google Scholar] [CrossRef]
  37. Israel, M.A.; Yuan, S.H.; Bardy, C.; Reyna, S.M.; Mu, Y.; Herrera, C.; Hefferan, M.P.; Van Gorp, S.; Nazor, K.L.; Boscolo, F.S.; et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 2012, 482, 216–220. [Google Scholar] [CrossRef]
  38. Lehtonen, S.; Hoytylainen, I.; Voutilainen, J.; Sonninen, T.M.; Kuusisto, J.; Laakso, M.; Hamalainen, R.H.; Oksanen, M.; Koistinaho, J. Generation of a human induced pluripotent stem cell line from a patient with a rare A673T variant in amyloid precursor protein gene that reduces the risk for Alzheimer’s disease. Stem Cell Res. 2018, 30, 96–99. [Google Scholar] [CrossRef]
  39. Potter, H.; Granic, A.; Caneus, J. Role of Trisomy 21 Mosaicism in Sporadic and Familial Alzheimer’s Disease. Curr. Alzheimer Res. 2016, 13, 7–17. [Google Scholar] [CrossRef]
  40. Chang, C.Y.; Chen, S.M.; Lu, H.E.; Lai, S.M.; Lai, P.S.; Shen, P.W.; Chen, P.Y.; Shen, C.I.; Harn, H.J.; Lin, S.Z.; et al. N-butylidenephthalide attenuates Alzheimer’s disease-like cytopathy in Down syndrome induced pluripotent stem cell-derived neurons. Sci. Rep. 2015, 5, 8744. [Google Scholar] [CrossRef] [Green Version]
  41. Yagi, T.; Ito, D.; Okada, Y.; Akamatsu, W.; Nihei, Y.; Okano, H.; Suzuki, N. [Modeling familial Alzheimer’s disease with induced pluripotent stem cells]. Rinsho Shinkeigaku 2012, 52, 1134–1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Oksanen, M.; Petersen, A.J.; Naumenko, N.; Puttonen, K.; Lehtonen, S.; Gubert Olive, M.; Shakirzyanova, A.; Leskela, S.; Sarajarvi, T.; Viitanen, M.; et al. PSEN1 Mutant iPSC-Derived Model Reveals Severe Astrocyte Pathology in Alzheimer’s Disease. Stem Cell Rep. 2017, 9, 1885–1897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Silva, M.C.; Cheng, C.; Mair, W.; Almeida, S.; Fong, H.; Biswas, M.H.U.; Zhang, Z.; Huang, Y.; Temple, S.; Coppola, G.; et al. Human iPSC-Derived Neuronal Model of Tau-A152T Frontotemporal Dementia Reveals Tau-Mediated Mechanisms of Neuronal Vulnerability. Stem Cell Rep. 2016, 7, 325–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Garcia-Leon, J.A.; Cabrera-Socorro, A.; Eggermont, K.; Swijsen, A.; Terryn, J.; Fazal, R.; Nami, F.; Ordovas, L.; Quiles, A.; Lluis, F.; et al. Generation of a human induced pluripotent stem cell-based model for tauopathies combining three microtubule-associated protein TAU mutations which displays several phenotypes linked to neurodegeneration. Alzheimers Dement. 2018, 14, 1261–1280. [Google Scholar] [CrossRef]
  45. Iovino, M.; Agathou, S.; Gonzalez-Rueda, A.; Del Castillo Velasco-Herrera, M.; Borroni, B.; Alberici, A.; Lynch, T.; O’Dowd, S.; Geti, I.; Gaffney, D.; et al. Early maturation and distinct tau pathology in induced pluripotent stem cell-derived neurons from patients with MAPT mutations. Brain 2015, 138, 3345–3359. [Google Scholar] [CrossRef] [Green Version]
  46. Maclean, G.A.; Menne, T.F.; Guo, G.; Sanchez, D.J.; Park, I.H.; Daley, G.Q.; Orkin, S.H. Altered hematopoiesis in trisomy 21 as revealed through in vitro differentiation of isogenic human pluripotent cells. Proc. Natl. Acad. Sci. USA 2012, 109, 17567–17572. [Google Scholar] [CrossRef] [Green Version]
  47. Clarimon, J.; Djaldetti, R.; Lleo, A.; Guerreiro, R.J.; Molinuevo, J.L.; Paisan-Ruiz, C.; Gomez-Isla, T.; Blesa, R.; Singleton, A.; Hardy, J. Whole genome analysis in a consanguineous family with early onset Alzheimer’s disease. Neurobiol. Aging 2009, 30, 1986–1991. [Google Scholar] [CrossRef]
  48. Awada, A.A. Early and late-onset Alzheimer’s disease: What are the differences? J. Neurosci. Rural Pract. 2015, 6, 455–456. [Google Scholar] [CrossRef] [Green Version]
  49. Kehoe, P.; Williams, J.; Lovestone, S.; Wilcock, G.; Owen, M.J. Presenilin-1 polymorphism and Alzheimer’s disease. The UK Alzheimer’s Disease Collaborative Group. Lancet 1996, 347, 1185. [Google Scholar] [CrossRef]
  50. Haapasalo, A.; Kovacs, D.M. The many substrates of presenilin/gamma-secretase. J. Alzheimers Dis. 2011, 25, 3–28. [Google Scholar] [CrossRef]
  51. Yang, G.; Zhou, R.; Zhou, Q.; Guo, X.; Yan, C.; Ke, M.; Lei, J.; Shi, Y. Structural basis of Notch recognition by human gamma-secretase. Nature 2019, 565, 192–197. [Google Scholar] [CrossRef] [PubMed]
  52. Citron, M.; Westaway, D.; Xia, W.; Carlson, G.; Diehl, T.; Levesque, G.; Johnson-Wood, K.; Lee, M.; Seubert, P.; Davis, A.; et al. Mutant presenilins of Alzheimer’s disease increase production of 42-residue amyloid beta-protein in both transfected cells and transgenic mice. Nat. Med. 1997, 3, 67–72. [Google Scholar] [CrossRef]
  53. Haass, C.; De Strooper, B. The presenilins in Alzheimer’s disease--proteolysis holds the key. Science 1999, 286, 916–919. [Google Scholar] [CrossRef] [PubMed]
  54. Duff, K.; Eckman, C.; Zehr, C.; Yu, X.; Prada, C.M.; Perez-tur, J.; Hutton, M.; Buee, L.; Harigaya, Y.; Yager, D.; et al. Increased amyloid-beta42(43) in brains of mice expressing mutant presenilin 1. Nature 1996, 383, 710–713. [Google Scholar] [CrossRef] [PubMed]
  55. Kaether, C.; Haass, C.; Steiner, H. Assembly, trafficking and function of gamma-secretase. Neurodegener Dis. 2006, 3, 275–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Coric, V.; van Dyck, C.H.; Salloway, S.; Andreasen, N.; Brody, M.; Richter, R.W.; Soininen, H.; Thein, S.; Shiovitz, T.; Pilcher, G.; et al. Safety and tolerability of the gamma-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Arch. Neurol. 2012, 69, 1430–1440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kumar, D.; Ganeshpurkar, A.; Kumar, D.; Modi, G.; Gupta, S.K.; Singh, S.K. Secretase inhibitors for the treatment of Alzheimer’s disease: Long road ahead. Eur. J. Med. Chem. 2018, 148, 436–452. [Google Scholar] [CrossRef]
  58. Mehta, D.; Jackson, R.; Paul, G.; Shi, J.; Sabbagh, M. Why do trials for Alzheimer’s disease drugs keep failing? A discontinued drug perspective for 2010–2015. Expert Opin. Investig. Drugs 2017, 26, 735–739. [Google Scholar] [CrossRef]
  59. Ables, J.L.; Breunig, J.J.; Eisch, A.J.; Rakic, P. Not(ch) just development: Notch signalling in the adult brain. Nat. Rev. Neurosci. 2011, 12, 269–283. [Google Scholar] [CrossRef] [Green Version]
  60. Mumm, J.S.; Kopan, R. Notch signaling: from the outside in. Dev. Biol. 2000, 228, 151–165. [Google Scholar] [CrossRef] [Green Version]
  61. Mitani, Y.; Yarimizu, J.; Saita, K.; Uchino, H.; Akashiba, H.; Shitaka, Y.; Ni, K.; Matsuoka, N. Differential effects between gamma-secretase inhibitors and modulators on cognitive function in amyloid precursor protein-transgenic and nontransgenic mice. J. Neurosci. 2012, 32, 2037–2050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Elvang, A.B.; Volbracht, C.; Pedersen, L.O.; Jensen, K.G.; Karlsson, J.J.; Larsen, S.A.; Mork, A.; Stensbol, T.B.; Bastlund, J.F. Differential effects of gamma-secretase and BACE1 inhibition on brain Abeta levels in vitro and in vivo. J. Neurochem. 2009, 110, 1377–1387. [Google Scholar] [CrossRef] [PubMed]
  63. Barten, D.M.; Meredith, J.E., Jr.; Zaczek, R.; Houston, J.G.; Albright, C.F. Gamma-secretase inhibitors for Alzheimer’s disease: balancing efficacy and toxicity. Drugs R D 2006, 7, 87–97. [Google Scholar] [CrossRef] [PubMed]
  64. Tagami, S.; Yanagida, K.; Kodama, T.S.; Takami, M.; Mizuta, N.; Oyama, H.; Nishitomi, K.; Chiu, Y.W.; Okamoto, T.; Ikeuchi, T.; et al. Semagacestat Is a Pseudo-Inhibitor of gamma-Secretase. Cell Rep. 2017, 21, 259–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Doody, R.S.; Raman, R.; Farlow, M.; Iwatsubo, T.; Vellas, B.; Joffe, S.; Kieburtz, K.; He, F.; Sun, X.; Thomas, R.G.; et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N. Engl. J. Med. 2013, 369, 341–350. [Google Scholar] [CrossRef]
  66. Gillman, K.W.; Starrett, J.E., Jr.; Parker, M.F.; Xie, K.; Bronson, J.J.; Marcin, L.R.; McElhone, K.E.; Bergstrom, C.P.; Mate, R.A.; Williams, R.; et al. Discovery and Evaluation of BMS-708163, a Potent, Selective and Orally Bioavailable gamma-Secretase Inhibitor. ACS Med. Chem. Lett. 2010, 1, 120–124. [Google Scholar] [CrossRef] [Green Version]
  67. Albright, C.F.; Dockens, R.C.; Meredith, J.E., Jr.; Olson, R.E.; Slemmon, R.; Lentz, K.A.; Wang, J.S.; Denton, R.R.; Pilcher, G.; Rhyne, P.W.; et al. Pharmacodynamics of selective inhibition of gamma-secretase by avagacestat. J. Pharmacol. Exp. Ther. 2013, 344, 686–695. [Google Scholar] [CrossRef] [Green Version]
  68. Coric, V.; Salloway, S.; van Dyck, C.H.; Dubois, B.; Andreasen, N.; Brody, M.; Curtis, C.; Soininen, H.; Thein, S.; Shiovitz, T.; et al. Targeting Prodromal Alzheimer Disease With Avagacestat: A Randomized Clinical Trial. JAMA Neurol. 2015, 72, 1324–1333. [Google Scholar] [CrossRef]
  69. Ran, Y.; Cruz, P.E.; Ladd, T.B.; Fauq, A.H.; Jung, J.I.; Matthews, J.; Felsenstein, K.M.; Golde, T.E. gamma-Secretase processing and effects of gamma-secretase inhibitors and modulators on long Abeta peptides in cells. J. Biol. Chem. 2014, 289, 3276–3287. [Google Scholar] [CrossRef] [Green Version]
  70. Graham, W.V.; Bonito-Oliva, A.; Sakmar, T.P. Update on Alzheimer’s Disease Therapy and Prevention Strategies. Annu. Rev. Med. 2017, 68, 413–430. [Google Scholar] [CrossRef] [Green Version]
  71. Golde, T.E.; Koo, E.H.; Felsenstein, K.M.; Osborne, B.A.; Miele, L. gamma-Secretase inhibitors and modulators. Biochim. Biophys. Acta 2013, 1828, 2898–2907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Sanz-Blasco, S.; Valero, R.A.; Rodriguez-Crespo, I.; Villalobos, C.; Nunez, L. Mitochondrial Ca2+ overload underlies Abeta oligomers neurotoxicity providing an unexpected mechanism of neuroprotection by NSAIDs. PLoS ONE 2008, 3, e2718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Woodling, N.S.; Colas, D.; Wang, Q.; Minhas, P.; Panchal, M.; Liang, X.; Mhatre, S.D.; Brown, H.; Ko, N.; Zagol-Ikapitte, I.; et al. Cyclooxygenase inhibition targets neurons to prevent early behavioural decline in Alzheimer’s disease model mice. Brain 2016, 139, 2063–2081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Imbimbo, B.P.; Giardina, G.A. gamma-secretase inhibitors and modulators for the treatment of Alzheimer’s disease: disappointments and hopes. Curr. Top. Med. Chem. 2011, 11, 1555–1570. [Google Scholar] [CrossRef]
  75. Xia, W.; Wong, S.T.; Hanlon, E.; Morin, P. gamma-Secretase modulator in Alzheimer’s disease: shifting the end. J. Alzheimers Dis. 2012, 31, 685–696. [Google Scholar] [CrossRef]
  76. Wong, L.R.; Ho, P.C. Role of serum albumin as a nanoparticulate carrier for nose-to-brain delivery of R-flurbiprofen: implications for the treatment of Alzheimer’s disease. J. Pharm. Pharmacol. 2018, 70, 59–69. [Google Scholar] [CrossRef]
  77. Mitchell, J.A.; Akarasereenont, P.; Thiemermann, C.; Flower, R.J.; Vane, J.R. Selectivity of nonsteroidal antiinflammatory drugs as inhibitors of constitutive and inducible cyclooxygenase. Proc. Natl. Acad. Sci. USA 1993, 90, 11693–11697. [Google Scholar] [CrossRef] [Green Version]
  78. Wang, J.; Cheng, X.; Zhang, X.; Liu, G.; Wang, Y.; Zhou, W.; Zhang, Y. A combination of indomethacin and atorvastatin ameliorates cognitive and pathological deterioration in PrP-hAbetaPPswe/PS1(DeltaE9) transgenic mice. J. Neuroimmunol. 2019, 330, 108–115. [Google Scholar] [CrossRef]
  79. De Jong, D.; Jansen, R.; Hoefnagels, W.; Jellesma-Eggenkamp, M.; Verbeek, M.; Borm, G.; Kremer, B. No effect of one-year treatment with indomethacin on Alzheimer’s disease progression: A randomized controlled trial. PLoS ONE 2008, 3, e1475. [Google Scholar] [CrossRef]
  80. Hahn, S.; Bruning, T.; Ness, J.; Czirr, E.; Baches, S.; Gijsen, H.; Korth, C.; Pietrzik, C.U.; Bulic, B.; Weggen, S. Presenilin-1 but not amyloid precursor protein mutations present in mouse models of Alzheimer’s disease attenuate the response of cultured cells to gamma-secretase modulators regardless of their potency and structure. J. Neurochem. 2011, 116, 385–395. [Google Scholar] [CrossRef]
  81. Borgegard, T.; Jureus, A.; Olsson, F.; Rosqvist, S.; Sabirsh, A.; Rotticci, D.; Paulsen, K.; Klintenberg, R.; Yan, H.; Waldman, M.; et al. First and second generation gamma-secretase modulators (GSMs) modulate amyloid-beta (Abeta) peptide production through different mechanisms. J. Biol. Chem. 2012, 287, 11810–11819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Wagner, S.L.; Rynearson, K.D.; Duddy, S.K.; Zhang, C.; Nguyen, P.D.; Becker, A.; Vo, U.; Masliah, D.; Monte, L.; Klee, J.B.; et al. Pharmacological and Toxicological Properties of the Potent Oral gamma-Secretase Modulator BPN-15606. J. Pharmacol. Exp. Ther. 2017, 362, 31–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Rogers, K.; Felsenstein, K.M.; Hrdlicka, L.; Tu, Z.; Albayya, F.; Lee, W.; Hopp, S.; Miller, M.J.; Spaulding, D.; Yang, Z.; et al. Modulation of gamma-secretase by EVP-0015962 reduces amyloid deposition and behavioral deficits in Tg2576 mice. Mol. Neurodegener 2012, 7, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Peng, H.; Talreja, T.; Xin, Z.; Cuervo, J.H.; Kumaravel, G.; Humora, M.J.; Xu, L.; Rohde, E.; Gan, L.; Jung, M.Y.; et al. Discovery of BIIB042, a Potent, Selective, and Orally Bioavailable gamma-Secretase Modulator. ACS Med. Chem. Lett. 2011, 2, 786–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Scannevin, R.H.; Chollate, S.; Brennan, M.S.; Snodgrass-Belt, P.A.; Peng, H.; Xu, L.; Jung, M.Y.; Bussiere, T.; Arastu, M.F.; Talreja, T.; et al. BIIB042, a novel gamma-secretase modulator, reduces amyloidogenic Abeta isoforms in primates and rodents and plaque pathology in a mouse model of Alzheimer’s disease. Neuropharmacology 2016, 103, 57–68. [Google Scholar] [CrossRef] [PubMed]
  86. Prikhodko, O.; Rynearson, K.D.; Sekhon, T.; Mante, M.M.; Nguyen, P.D.; Rissman, R.A.; Tanzi, R.E.; Wagner, S.L. The GSM BPN-15606 as a Potential Candidate for Preventative Therapy in Alzheimer’s Disease. J. Alzheimers Dis. 2020. [Google Scholar] [CrossRef]
  87. Endres, K.; Deller, T. Regulation of Alpha-Secretase ADAM10 In vitro and In vivo: Genetic, Epigenetic, and Protein-Based Mechanisms. Front. Mol. Neurosci. 2017, 10, 56. [Google Scholar] [CrossRef]
  88. Yan, L.; Deng, Y.; Gao, J.; Liu, Y.; Li, F.; Shi, J.; Gong, Q. Icariside II Effectively Reduces Spatial Learning and Memory Impairments in Alzheimer’s Disease Model Mice Targeting Beta-Amyloid Production. Front. Pharmacol. 2017, 8, 106. [Google Scholar] [CrossRef] [Green Version]
  89. Li, X.; Cui, J.; Yu, Y.; Li, W.; Hou, Y.; Wang, X.; Qin, D.; Zhao, C.; Yao, X.; Zhao, J.; et al. Traditional Chinese Nootropic Medicine Radix Polygalae and Its Active Constituent Onjisaponin B Reduce beta-Amyloid Production and Improve Cognitive Impairments. PLoS ONE 2016, 11, e0151147. [Google Scholar] [CrossRef]
  90. Singh, J.C.; Kakalij, R.M.; Kshirsagar, R.P.; Kumar, B.H.; Komakula, S.S.; Diwan, P.V. Cognitive effects of vanillic acid against streptozotocin-induced neurodegeneration in mice. Pharm. Biol. 2015, 53, 630–636. [Google Scholar] [CrossRef] [Green Version]
  91. Li, X.; Sun, Y.; Wei, Y.; Zhou, L.; Liu, L.; Yin, P.; Liu, Y.; Wu, S.; Li, J.; Lu, C. Onjisaponin B (OB) is Neuroprotective During Cognitive Loss Through Immune-mediated and SIRT1 Pathways. Curr. Neurovasc Res. 2018, 15, 94–102. [Google Scholar] [CrossRef] [PubMed]
  92. Cai, H.; Wang, Y.; He, J.; Cai, T.; Wu, J.; Fang, J.; Zhang, R.; Guo, Z.; Guan, L.; Zhan, Q.; et al. Neuroprotective effects of bajijiasu against cognitive impairment induced by amyloid-beta in APP/PS1 mice. Oncotarget 2017, 8, 92621–92634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Chen, D.L.; Zhang, P.; Lin, L.; Shuai, O.; Zhang, H.M.; Liu, S.H.; Wang, J.Y. Protective effect of Bajijiasu against beta-amyloid-induced neurotoxicity in PC12 cells. Cell Mol. Neurobiol. 2013, 33, 837–850. [Google Scholar] [CrossRef] [PubMed]
  94. Xu, T.T.; Zhang, Y.; He, J.Y.; Luo, D.; Luo, Y.; Wang, Y.J.; Liu, W.; Wu, J.; Zhao, W.; Fang, J.; et al. Bajijiasu Ameliorates beta-Amyloid-Triggered Endoplasmic Reticulum Stress and Related Pathologies in an Alzheimer’s Disease Model. Cell Physiol. Biochem. 2018, 46, 107–117. [Google Scholar] [CrossRef]
  95. Mao, J.; Huang, S.; Liu, S.; Feng, X.L.; Yu, M.; Liu, J.; Sun, Y.E.; Chen, G.; Yu, Y.; Zhao, J.; et al. A herbal medicine for Alzheimer’s disease and its active constituents promote neural progenitor proliferation. Aging Cell 2015, 14, 784–796. [Google Scholar] [CrossRef]
  96. Yang, C.; Li, X.; Mo, Y.; Liu, S.; Zhao, L.; Ma, X.; Fang, Z.; Chen, J.; Chen, Y.; Yu, X.; et al. beta-Asarone Mitigates Amyloidosis and Downregulates RAGE in a Transgenic Mouse Model of Alzheimer’s Disease. Cell Mol. Neurobiol. 2016, 36, 121–130. [Google Scholar] [CrossRef]
  97. Wang, Z.; Liu, Q.; Zhang, R.; Liu, S.; Xia, Z.; Hu, Y. Catalpol ameliorates beta amyloid-induced degeneration of cholinergic neurons by elevating brain-derived neurotrophic factors. Neuroscience 2009, 163, 1363–1372. [Google Scholar] [CrossRef]
  98. Huang, J.Z.; Wu, J.; Xiang, S.; Sheng, S.; Jiang, Y.; Yang, Z.; Hua, F. Catalpol preserves neural function and attenuates the pathology of Alzheimer’s disease in mice. Mol. Med. Rep. 2016, 13, 491–496. [Google Scholar] [CrossRef]
  99. Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol. 2018, 14, 450–464. [Google Scholar] [CrossRef]
  100. Draper, H.H.; Hadley, M. Malondialdehyde determination as index of lipid peroxidation. Methods Enzymol. 1990, 186, 421–431. [Google Scholar] [CrossRef]
  101. Jia, S.L.; Wu, X.L.; Li, X.X.; Dai, X.L.; Gao, Z.L.; Lu, Z.; Zheng, Q.S.; Sun, Y.X. Neuroprotective effects of liquiritin on cognitive deficits induced by soluble amyloid-beta1-42 oligomers injected into the hippocampus. J. Asian Nat. Prod. Res. 2016, 18, 1186–1199. [Google Scholar] [CrossRef] [PubMed]
  102. Small, D.H.; Mok, S.S.; Bornstein, J.C. Alzheimer’s disease and Abeta toxicity: from top to bottom. Nat. Rev. Neurosci. 2001, 2, 595–598. [Google Scholar] [CrossRef] [PubMed]
  103. Cui, Y.M.; Ao, M.Z.; Li, W.; Yu, L.J. Effect of glabridin from Glycyrrhiza glabra on learning and memory in mice. Planta Med. 2008, 74, 377–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Jin, H.J.; Li, C.G. Tanshinone IIA and Cryptotanshinone Prevent Mitochondrial Dysfunction in Hypoxia-Induced H9c2 Cells: Association to Mitochondrial ROS, Intracellular Nitric Oxide, and Calcium Levels. Evid Based Complement. Alternat. Med. 2013, 2013, 610694. [Google Scholar] [CrossRef] [Green Version]
  105. Maione, F.; Piccolo, M.; De Vita, S.; Chini, M.G.; Cristiano, C.; De Caro, C.; Lippiello, P.; Miniaci, M.C.; Santamaria, R.; Irace, C.; et al. Down regulation of pro-inflammatory pathways by tanshinone IIA and cryptotanshinone in a non-genetic mouse model of Alzheimer’s disease. Pharmacol Res. 2018, 129, 482–490. [Google Scholar] [CrossRef]
  106. Li, F.; Wu, X.; Li, J.; Niu, Q. Ginsenoside Rg1 ameliorates hippocampal long-term potentiation and memory in an Alzheimer’s disease model. Mol. Med. Rep. 2016, 13, 4904–4910. [Google Scholar] [CrossRef] [Green Version]
  107. Zhu, J.; Mu, X.; Zeng, J.; Xu, C.; Liu, J.; Zhang, M.; Li, C.; Chen, J.; Li, T.; Wang, Y. Ginsenoside Rg1 prevents cognitive impairment and hippocampus senescence in a rat model of D-galactose-induced aging. PLoS ONE 2014, 9, e101291. [Google Scholar] [CrossRef] [Green Version]
  108. Combs, C.K.; Johnson, D.E.; Karlo, J.C.; Cannady, S.B.; Landreth, G.E. Inflammatory mechanisms in Alzheimer’s disease: inhibition of beta-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARgamma agonists. J. Neurosci. 2000, 20, 558–567. [Google Scholar] [CrossRef]
  109. Wang, X.; Wang, Y.; Hu, J.P.; Yu, S.; Li, B.K.; Cui, Y.; Ren, L.; Zhang, L.D. Astragaloside IV, a Natural PPARgamma Agonist, Reduces Abeta Production in Alzheimer’s Disease Through Inhibition of BACE1. Mol. Neurobiol. 2017, 54, 2939–2949. [Google Scholar] [CrossRef]
  110. Fu, R.H.; Harn, H.J.; Liu, S.P.; Chen, C.S.; Chang, W.L.; Chen, Y.M.; Huang, J.E.; Li, R.J.; Tsai, S.Y.; Hung, H.S.; et al. n-butylidenephthalide protects against dopaminergic neuron degeneration and alpha-synuclein accumulation in Caenorhabditis elegans models of Parkinson’s disease. PLoS ONE 2014, 9, e85305. [Google Scholar] [CrossRef] [Green Version]
  111. Hsueh, K.W.; Chiou, T.W.; Chiang, S.F.; Yamashita, T.; Abe, K.; Borlongan, C.V.; Sanberg, P.R.; Huang, A.Y.; Lin, S.Z.; Harn, H.J. Autophagic down-regulation in motor neurons remarkably prolongs the survival of ALS mice. Neuropharmacology 2016, 108, 152–160. [Google Scholar] [CrossRef] [PubMed]
  112. Hasanein, P. Glabridin as a major active isoflavan from Glycyrrhiza glabra (licorice) reverses learning and memory deficits in diabetic rats. Acta Physiol. Hung. 2011, 98, 221–230. [Google Scholar] [CrossRef] [PubMed]
  113. Bolmont, T.; Clavaguera, F.; Meyer-Luehmann, M.; Herzig, M.C.; Radde, R.; Staufenbiel, M.; Lewis, J.; Hutton, M.; Tolnay, M.; Jucker, M. Induction of tau pathology by intracerebral infusion of amyloid-beta -containing brain extract and by amyloid-beta deposition in APP x Tau transgenic mice. Am. J. Pathol. 2007, 171, 2012–2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Hurtado, D.E.; Molina-Porcel, L.; Iba, M.; Aboagye, A.K.; Paul, S.M.; Trojanowski, J.Q.; Lee, V.M. A{beta} accelerates the spatiotemporal progression of tau pathology and augments tau amyloidosis in an Alzheimer mouse model. Am. J. Pathol. 2010, 177, 1977–1988. [Google Scholar] [CrossRef] [PubMed]
  115. Lewis, J.; Dickson, D.W.; Lin, W.L.; Chisholm, L.; Corral, A.; Jones, G.; Yen, S.H.; Sahara, N.; Skipper, L.; Yager, D.; et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 2001, 293, 1487–1491. [Google Scholar] [CrossRef] [Green Version]
  116. Choi, S.H.; Kim, Y.H.; Hebisch, M.; Sliwinski, C.; Lee, S.; D’Avanzo, C.; Chen, H.; Hooli, B.; Asselin, C.; Muffat, J.; et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 2014, 515, 274–278. [Google Scholar] [CrossRef]
  117. Sreenivasmurthy, S.G.; Liu, J.Y.; Song, J.X.; Yang, C.B.; Malampati, S.; Wang, Z.Y.; Huang, Y.Y.; Li, M. Neurogenic Traditional Chinese Medicine as a Promising Strategy for the Treatment of Alzheimer’s Disease. Int. J. Mol. Sci. 2017, 18, 272. [Google Scholar] [CrossRef] [Green Version]
  118. Liu, S.J.; Yang, C.; Zhang, Y.; Su, R.Y.; Chen, J.L.; Jiao, M.M.; Chen, H.F.; Zheng, N.; Luo, S.; Chen, Y.B.; et al. Neuroprotective effect of beta-asarone against Alzheimer’s disease: regulation of synaptic plasticity by increased expression of SYP and GluR1. Drug Des. Devel Ther. 2016, 10, 1461–1469. [Google Scholar] [CrossRef] [Green Version]
  119. Chang, C.P.; Liu, Y.F.; Lin, H.J.; Hsu, C.C.; Cheng, B.C.; Liu, W.P.; Lin, M.T.; Hsu, S.F.; Chang, L.S.; Lin, K.C. Beneficial Effect of Astragaloside on Alzheimer’s Disease Condition Using Cultured Primary Cortical Cells Under beta-amyloid Exposure. Mol. Neurobiol. 2016, 53, 7329–7340. [Google Scholar] [CrossRef]
Figure 1. The proportion of dementia caused by various neurodegenerative diseases. AD = Alzheimer’s disease, MCI = mild cognitive impairment, SCD = subjective cognitive decline, FTLD = frontotemporal dementia, DLB = dementia with Lewy bodies, and VaD = vascular dementia.
Figure 1. The proportion of dementia caused by various neurodegenerative diseases. AD = Alzheimer’s disease, MCI = mild cognitive impairment, SCD = subjective cognitive decline, FTLD = frontotemporal dementia, DLB = dementia with Lewy bodies, and VaD = vascular dementia.
Ijms 21 01327 g001
Figure 2. Beta-amyloid accumulation. In general, cleavage of beta amyloid precursor protein (APP) by alpha-secretase and γ-secretase cuts APP into P3 peptides (1–24~1–26 amino acids). However, once the gene generates mutations, such as APP and GSIS1, PSEN1 gene expression increases, and the wrong cleavage is likely to occur and produce not easily metabolized peptide fragments (1–40~1–42 amino acids).
Figure 2. Beta-amyloid accumulation. In general, cleavage of beta amyloid precursor protein (APP) by alpha-secretase and γ-secretase cuts APP into P3 peptides (1–24~1–26 amino acids). However, once the gene generates mutations, such as APP and GSIS1, PSEN1 gene expression increases, and the wrong cleavage is likely to occur and produce not easily metabolized peptide fragments (1–40~1–42 amino acids).
Ijms 21 01327 g002
Figure 3. γ-secretase inhibitors. Notch and APP proteins are inactive when they are first synthesized and must have sections removed to become active. Furin and BACE first cleave these sections and activate the proteins. The second cleavage is by γ-secretase. GSIs can reduce γ-secretase but have a side effect of inhibiting Notch-signaling proteins. The function of this protein is related to nerve growth and differentiation. ADAM = A disintegrin and metalloproteinase, APP = amyloid precursor protein, BACE = beta-secretase, and GSI = γ-secretase inhibitors.
Figure 3. γ-secretase inhibitors. Notch and APP proteins are inactive when they are first synthesized and must have sections removed to become active. Furin and BACE first cleave these sections and activate the proteins. The second cleavage is by γ-secretase. GSIs can reduce γ-secretase but have a side effect of inhibiting Notch-signaling proteins. The function of this protein is related to nerve growth and differentiation. ADAM = A disintegrin and metalloproteinase, APP = amyloid precursor protein, BACE = beta-secretase, and GSI = γ-secretase inhibitors.
Ijms 21 01327 g003
Figure 4. GSI and GSM structures. The GSIs are (a) semagacestat and (b) avagacestat. The first-generation GSMs are (c) R-flurbiprofen and (d) indomethacin. The second-generation GSMs are (e) EVP-0015962, (f) BPN-15606, and (g) BIIB042.
Figure 4. GSI and GSM structures. The GSIs are (a) semagacestat and (b) avagacestat. The first-generation GSMs are (c) R-flurbiprofen and (d) indomethacin. The second-generation GSMs are (e) EVP-0015962, (f) BPN-15606, and (g) BIIB042.
Ijms 21 01327 g004
Figure 5. γ-secretase modulators. GSMs do not inhibit γ-secretase, but they can shift the profile of the secreted beta-amyloid peptides produced. GSMs can decrease the levels of Aβ1–42 peptides and increase the levels of Aβ1–38 peptides. Aβ = beta-amyloid and GSM = γ-secretase modulators.
Figure 5. γ-secretase modulators. GSMs do not inhibit γ-secretase, but they can shift the profile of the secreted beta-amyloid peptides produced. GSMs can decrease the levels of Aβ1–42 peptides and increase the levels of Aβ1–38 peptides. Aβ = beta-amyloid and GSM = γ-secretase modulators.
Ijms 21 01327 g005
Figure 6. Small-molecule drug structures.
Figure 6. Small-molecule drug structures.
Ijms 21 01327 g006
Figure 7. The iPSC screen drug strategy of Alzheimer’s disease. The AD-iPSC cell model can produces different AD symptoms, including β-amyloid accumulation, nerve entanglement, and inflammatory response. iPSCs have be customized according to personalization and, through various different treatment strategies, the treatment effect can be achieved more efficiently. AD = Alzheimer’s disease, GSI = γ-secretase inhibitors, GSM = γ -secretase modulators, and iPSC = induced pluripotent stem cell.
Figure 7. The iPSC screen drug strategy of Alzheimer’s disease. The AD-iPSC cell model can produces different AD symptoms, including β-amyloid accumulation, nerve entanglement, and inflammatory response. iPSCs have be customized according to personalization and, through various different treatment strategies, the treatment effect can be achieved more efficiently. AD = Alzheimer’s disease, GSI = γ-secretase inhibitors, GSM = γ -secretase modulators, and iPSC = induced pluripotent stem cell.
Ijms 21 01327 g007
Table 1. Alzheimer’s diseases modeled with induced pluripotent stem (iPS) cells.
Table 1. Alzheimer’s diseases modeled with induced pluripotent stem (iPS) cells.
Gene Mutation of iPSCsiPS Cells Produce EffectReferences
Presenilin-1 ΔE9 mutationImpair γ-secretase activity but do not disrupt γ-secretase-independent functions of PSEN1Minna Oksanen [42]
FAD patients with mutations in PSEN1 (A246E) and PSEN2 (N141I)Increased toxic Aβ1-42 secretionTakuya Yagi [41]
Amyloid precursor protein (APP) A673T mutationProtective against Alzheimer’s disease and cognitive declineŠárka Lehtonen [38]
APP double mutation (KM670/671NL)Increase the total Aβ burdenMinna Oksanen [36]
Duplication of the amyloid precursor protein gene (APP(Dp))Increased Aβ 1–40, p-tau (Thr 231), and active glycogen synthase kinase-3βMason A. Israel [37]
Trisomy of chromosome 21 (Ts21)Aβ aggregation and increased tau proteinGlenn A. Maclean [46]
A152T variant in MAPTA152T-iPS cells-derived neurons showed accumulation, redistribution, and decreased solubility of tau.M. Catarina Silva [43]
MAPT gene mutations (N279K, P301L, and E10+16)Deficiencies in neurite outgrowth and upregulation of neurodegenerative pathwaysJuan Antonio García-León [44]
Table 2. The role and mechanisms of small-molecule drugs treated in the Alzheimer’s disease model.
Table 2. The role and mechanisms of small-molecule drugs treated in the Alzheimer’s disease model.
ExtractsExtracted from/Chinese NameDrug Doses & Experimental ModelPossible Molecular Mechanism Effect Reference
Icariin IIHerba Epimedii/淫羊藿APP/PS1 transgenic mice were treated orally g icariside II 30 m/kg.Effectively ameliorated cognitive function deficits, but also inhibited neuronal degeneration and reduced the formation of plaque burden.[88]
Onjisaponin BRadix polygalae/遠志APP/PS1 transgenic mice were treated orally 200 μL of onjisaponin B 1 mg/mL.The Onjisaponin B suppresses Aβ production without direct inhibition of β-secretase and γ-secretase activities.[89]
BajijiasuMorinda officinalis/巴戟天APP/PS1 transgenic mice were treated orally 20 mg and 80 mg bajijiasu/kg.Reductions of Aβ deposition and senile plaques and have higher levels of neurotrophic factors and inhibitory function on neuroinflammation in the brains of APP/PS1 mice.[92]
AsaronesRhizoma Acori tatarinowii/石菖蒲Adult hippocampal neural progenitor cells (NPC) cultures treated with 1 μM asarones.The Asarones enhanced NPC proliferation and neurogenesis in the hippocampi of transgenic AD model mice.[95]
CatalpolRehmannia glutinosa/地黃APP/PS1 transgenic mice were treated orally catalpol 50 mg / kg.Improves memory and protects the forebrain neurons through increasing BDNF expression.[97]
LiquiritinGlycyrrhizae radix/甘草Soluble amyloid-β1-42 oligomers injected into the hippocampus induced cognitive-deficit rats. Treated by liquiritin was orally 50~100 mg/kg.The drugs improve Aβ1-42-induced spatial learning and memory impairment through inhibiting oxidative stress and neural apoptosis.[101]
GlabridinGlycyrrhiza glabra/洋甘草Oral glabridin 25 and 50 mg/kg treated for diabetic rats [112].It reversed learning and memory deficits of diabetic rats.[103]
Tanshinone IIASalviae miltiorrhizae/丹参Oral 10mg/kg treated for nongenetic mouse model of β-amyloid-induced AD.That TIIA and CT display anti-inflammatory and neuron-protective effects in a nongenetic mouse model of AD.[105].
Cryptotanshinone
Ginsenoside Rg1Radix ginseng/人參Oral 20 mg/kg D-galactose-induced aging rat model.It prevents cognitive impairment and hippocampus senescence of D-galactose-induced aging in a rat model.[107]
Astragaloside IVAstragalus membranaceus/黃耆APP/PS1 mice that were treated orally AS-IV 10 mg/kg.AS-IV treatment increased PPARγ and reduced Aβ plaque formation in the brain.[109]
n-ButylidenephthalideAngelica sinensis/當歸Down syndrome - iPSCs were treated n-BP 10 μM for 3 days.n-BP benefitted AD treatment by scavenging Aβ aggregates and neurofibrillary tangles.[40]

Share and Cite

MDPI and ACS Style

Wuli, W.; Tsai, S.-T.; Chiou, T.-W.; Harn, H.-J. Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 1327. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21041327

AMA Style

Wuli W, Tsai S-T, Chiou T-W, Harn H-J. Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer’s Disease. International Journal of Molecular Sciences. 2020; 21(4):1327. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21041327

Chicago/Turabian Style

Wuli, Wei, Sheng-Tzung Tsai, Tzyy-Wen Chiou, and Horng-Jyh Harn. 2020. "Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer’s Disease" International Journal of Molecular Sciences 21, no. 4: 1327. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21041327

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop