Skip to main content
  • Research article
  • Open access
  • Published:

Effect of policosanol from insect wax on amyloid β-peptide-induced toxicity in a transgenic Caenorhabditis elegans model of Alzheimer’s disease

Abstract

Background

Alzheimer’s disease (AD), an age-related neurodegenerative disorder and a serious public health concern, is mainly caused by β-amyloid (Aβ)-induced toxicity. Currently, a limited number of drugs are effective against AD, and only a few are used for its treatment. According to traditional Chinese medicine, white wax is mainly composed of policosanol, hexacosanol, and octacosanol. Policosanol has been shown to reduce lipid levels in blood and alleviate the symptoms associated with diabetic complications and neurodegenerative disorders, such as Parkinson’s disease and AD. However, the efficacy of policosanol depends on the purity and composition of the preparation, and the therapeutic efficacy of policosanol derived from insect wax (PIW) in AD is unknown.

Methods

Here, we identified the main components of PIW and investigated the effects of PIW on Aβ-induced toxicity and life-span in a transgenic Caenorhabditis elegans model of AD, CL4176. Furthermore, we estimated the expression of amyloid precursor-like protein (apl-1) and the genes involved in various pathways associated with longevity and alleviation of AD-related symptoms in PIW-fed CL4176.

Results

PIW mainly consists of tetracosanol, hexacosanol, octacosanol, and triacontanol; it could decrease the Aβ-induced paralysis rate from 86.87 to 66.97% (P < 0.01) and extend the life-span from 6.2 d to 7.8 d (P < 0.001) in CL4176 worms. Furthermore, PIW downregulated apl-1, a gene known to be associated with the levels of Aβ deposits in C. elegans. Additionally, our results showed that PIW modulated the expression of genes associated with longevity-related pathways such as heat shock response, anti-oxidative stress, and glutamine cysteine synthetase.

Conclusion

Our findings suggest that PIW may be a potential therapeutic agent for the prevention and treatment of AD. However, its effects on murine models and patients with AD need to be explored further.

Peer Review reports

Background

Insect wax secreted by the male second-instar larva of the scale insect Ericerus pela Chavannes (Fig. 1a, Fig. 1b, Fig. 1c), also known as white wax (Fig. 1d) [1], has been used in China for over a thousand years for various medicinal and industrial purposes [2, 3]. It is extensively used in the textile and batik, food packaging, precision instrument lubrication and sealing, medicine, and cosmetic industries [1, 4, 5]. E. pela is found in the north subtropical, middle subtropical, and temperate regions, including China, Vietnam, Japan, and North Korea. In China, it is mainly found in the Yunnan, Guizhou, Sichuan, Zhejiang, Shaanxi, Guangxi, and Hunan provinces. Owing to the various applications of white wax, breeding of the scale insect is vital to the white wax bio-industry in these regions. Chinese white wax has been a featured bio-industry product, and hundreds of tons of the insect wax is produced every year in China [1]. The primary component of insect wax is high molecular weight wax esters, consisting of monobasic saturated fatty alcohol and monobasic saturated fatty acids, accounting for about 93–95% of its constituents [6]. Hexacosanoic acid hexacosyl ester and octocosoic acid dioctadecyl ester are the major wax esters in insect wax [7]. As described in Li Shizhen’s “Ben Cao Gan Mu” (Compendium of Materia Medica), insect wax is nontoxic and can be used as a hemostatic agent for bones and muscles, to relieve pain and reinforce deficiencies, to promote concrescence of fractures, and to treat alopecia of the scalp [8]. As demonstrated by recent studies, Chinese white wax scale modulates humoral and cellular immunity, has antioxidant activities, improves immune response in vivo, and attenuates atopic dermatitis [9,10,11,12,13]. In China, research on white wax and the white wax scale insect has been continually carried out to promote the growth of its insect wax industry.

Fig. 1
figure 1

Morphology of insect wax and the extracted policosanol. a The second-instar male nymphs of Ericerus pela Chavannes. b The second-instar male nymphs of Ericerus pela begin to secrete wax. c Secreted wax layer. d White wax. e Powdery form of policosanol extracted from insect wax using the reduction method

Policosanol derived from insect wax (PIW) is a mixture of saturated monohydric alcohols. It is a white powdery substance (Fig. 1e) obtained by reducing white wax at yields of 95–98% [14]. Policosanols mainly exist as fatty acid esters in the natural sources, such as bees wax, sugar cane wax, rice bran wax, white wax, corn bran wax, wheat germ, epidermis of some plants, rhizome, and grains [15,16,17,18]. Researchers from Cuba have isolated and produced the first policosanol supplement from sugarcane wax [19]. The beneficial physiological effects of policosanols include reducing platelet aggregation, endothelial damage, and foam cell formation [20], increasing muscle endurance [21], improving performance of coronary heart disease patients during exercise [22], and anti-arthritic and antioxidant properties [23]. Furthermore, studies exploring the physiological activities of policosanol have focused on its role in decreasing the levels of low-density lipoprotein (LDL) and increasing the levels of high-density lipoprotein (HDL) in the blood [24,25,26,27,28,29,30,31]. In addition, PIW promotes skin wound healing in mice; exhibits antibacterial, anti-inflammatory, and analgesic properties; and is skin-safe [32]. Furthermore, it has been reported to cause proliferation of human follicle dermal papilla cells [33] and promote hair growth in androgen-induced alopecia mice [34]. However, the efficacy of policosanols depends on the purity and composition of the preparation [19]; hence, policosanols obtained from different sources may have varied effects. PIW is composed of hexacosanol, octacosanol, tetradecanol, triacontanol, pentadecanol, and heptanol [14]. Hexacosanol and octacosanol have been reported to reduce the levels of lipids in the blood and alleviate the symptoms associated with diabetic complications and neurodegenerative disorders, such as Parkinson’s and Alzheimer’s disease (AD) [35,36,37].

AD is a progressive neurodegenerative disorder and is the most common cause of dementia in the aged population [38]. The major constituent of senile plaques, the 39–43 amino acid-long amyloid-beta peptide (Aβ), is thought to be central to the pathogenesis of AD [39, 40]. Currently, the number of drugs effective against AD is limited and only a few drugs, including acetylcholinesterase inhibitors and N-methyl-D-aspartate receptor antagonists, have been approved by the U.S. Food and Drug Administration for the treatment of AD [41, 42]. However, these drugs provide short-term relief to AD patients with mild to moderate symptoms and have obvious side effects [43]. Hence, efforts have been made to develop strategies that target Aβ for the prevention and treatment of AD [44, 45]. Nonetheless, it is particularly important to explore natural sources for drugs with fewer side effects and high efficacy.

In the current study, we investigated the effects of PIW on Aβ-induced toxicity using a transgenic Caenorhabditis elegans strain, CL4176. Additionally, we explored various pathways associated with the alleviation of paralysis, along with their underlying mechanisms, as well as discussed the potential of PIW as a treatment option for AD.

Methods

Preparation of policosanol from white wax and quantification of PIW using gas chromatography (GC)

White wax was purchased from a market in the Zhijiang county, Hunan province in the east of China. Policosanol was prepared by reduction of white wax using lithium aluminum hydride (LiAlH4), as described previously [14]. The ratio of wax and LiAlH4 was 100:5 ~ 7(w: w) and the reaction was carried out in a 250 mL flask. Briefly, white wax and LiAlH4 were allowed to react in a flask, after which tetrahydrofuran was added slowly at 60 °C with ultrasonication (40 KHz). This was followed by neutralization of tetrahydrofuran with 1 mol/L of chlorohydric acid and washes with hot water; the resultant product was then dried at 65 °C. Finally, the solid residue was recrystallized using a solution of chloroform and absolute ethanol (v/v, 3:1), followed by drying at 65 °C. The w:w ratio of the solid residue to the mixture of chloroform and absolute ethanol was 1:10, and the obtained yield of PIW was 95–98%.

Four standard components— 20.9 mg of tetracosanol, 20.1 mg of hexacosanol, 20.9 mg of octacosanol, and 20.6 mg of triacontanol (Sigma-Aldrich)—were dissolved in 100 mL chloroform, using ultrasonication, to prepare a standard solution. Then, 0.50, 1.00, 2.50, 5.00, and 10.00 mL of the standard solution was added into five 10 mL volumetric flasks, followed by the addition of chloroform, to prepare a gradient of standard working solutions for further use. PIW (16.0 mg) was dissolved in chloroform to prepare a sample solution with a concentration of 1.6 mg/ml. After filtration through a 0.45 μm organic film (PVDF, Millipore), 2 μL of each working solution and the sample solution was injected into the Agilent Gas Chromatograph instrument (7809B). The column used was HP-5 (30 m × 0.25 mm × 0.25 μm), with the following parameters: injector temperature; 320 °C, detector temperature: 340 °C, column oven temperature: 200 °C (1 min), increased to 320 °C (10 min) at a rate of 5 °C/min, carrier gas: nitrogen, purity ≥99.99%, fuel gas: hydrogen, purity ≥99.99%, column flow rate: 1.0 mL/min, hydrogen flow rate: 30 mL/min, air flow rate: 300 mL/min. The contents of PIW were calculated using the peak area of the four standard components and sample solution.

C. elegans strains and maintenance conditions

CL4176, the transgenic C. elegans strain, was provided by Dr. Ding Aijun, Kunming University, Yunnan, China. The strain was maintained at 16 °C on nematode growth media (NGM) plates seeded with E. coli OP50, and the temperature was increased to the permissive 25 °C to activate the expression of human Aβ1–42 peptide and induce paralysis. E. coli OP50 was grown in Luria–Bertani growth medium. The synchronization of nematode culture was achieved by sodium hypochlorite treatment, which kills adult worms, and the hatched L1 larvae were recovered on a NGM plate. The worms, either from the L1 stage (1 d of age) or egg stage, were fed the drugs.

Paralysis assay

The synchronized nematode hatchlings derived from the synchronized eggs (day 1) were cultured in M9 buffer at 16 °C. Worms at stage L1 were fed 0.5, 1, 2, 3, or 4 μg/mL of PIW, whereas unfed worms were considered as controls. The concentrations of PIW were decided based on the results of a preliminary study(data not shown). Meanwhile, the temperature was increased to 25 °C at start of the 36th hour, which represents the 3rd larval stage, and was maintained at 25 °C until the end of the paralysis assay. Paralysis rates of the control group and different treatments were scored at the 48th hour, which represents the late period of the 4th larval stage. Worms that did not move their whole body or only moved their head when gently touched with a platinum loop, were scored as paralyzed. Each group had at least 100 worms.

Life-span analysis

Worms were fed PIW (2 or 3 μg/mL) from stage L1, and late 4th stage larvae were synchronized. Untreated worms were considered as controls. Day 1 of the life-span assay started at 24 h after transferring the worms onto new plates. The number of worms was counted every day until the last worm died. Worms that did not respond to 3 to 5 mechanical touches were scored as dead and were removed from the plates. Furthermore, the worms that died from crawling off the agar were excluded from the analysis.

Measurement of superoxide dismutase (SOD) and reactive oxygen species (ROS) levels

Intracellular SOD levels were measured in transgenic CL4176 worms using a SOD kit (Nanjing Jiancheng Bioengineering Institute, Jiangsu, China). Age-synchronized, L1 stage C. elegans were transferred on to NGM plates containing 2 or 3 μg/mL of PIW and incubated for 48 h at 25 °C. The worms were then collected into a microfuge tube and washed twice with phosphate-buffered saline (PBS) to remove E. coli. Then, 200 μL of PBS was added to the homogenate on ice, centrifuged, and the resultant supernatant was used to measure SOD activity.

Intracellular ROS levels were measured using the 2′,7′-dichlorodihydro-fluorescein diacetate (H2DCF-DA) fluorescent probe (Nanjing Jiancheng Bioengineering Institute, Jiangsu, China). After 48 h of incubation, the worms were collected into a microfuge tube and washed thrice with M9 buffer, ground, and transferred to 96-well plates. Then, 50 μL of H2DCF-DA was added to each well and a final concentration of 50 μmol/L of H2DCF-DA was obtained. The fluorescence intensity of (dichloro) fluorescein was measured at 640 nm using the fluorescence enzyme method, and the concentration of ROS was determined.

Quantitative reverse transcription (qRT)-PCR analysis

Worms were synchronized on NGM plates with 2 or 3 μg/mL of PIW after culturing for 2 d. Untreated nematodes were used as controls. Total RNA was isolated using the TRIzol reagent (Invitrogen, USA) as per manufacturer’s protocol. Equal amounts of RNA were reverse transcribed into cDNA with the PrimeScript™ RT reagent kit (Takara, Japan). Primers used for amplification, along with their corresponding gene names, are listed in Table 1. qRT PCR was performed on the Applied Biosystems QuantStudio 3 (ABI, USA) using 20 μL of reaction mixture containing primers, cDNA, and PowerUp™ SYBR Green Master Mix (ABI, USA). The PCR amplification of 40 cycles was carried out at two stages; the hold stage involving UDG activation at 50 °C for 2 min and Dual-Lock TM DNA at 95 °C for 2 min, and the PCR stage involving denaturation at 95 °C for 15 s and annealing and extension at 60 °C for 1 min. Relative quantification of the genes was performed using the 2-ΔΔCT method, and act-1 was used as the internal standard.

Table 1 Primer sequences used for amplification of genes in policosanol derived from insect wax (PIW)-fed transgenic Caenorhabditis elegans

Statistical analysis

Statistical analysis was performed using the GraphPad Prism 8.0 software. All values were presented as means ± standard deviation. One-way analysis of variance was used for determining the statistically significant differences between groups, and a P value < 0.05 was considered significant. Three independent experiments were conducted, and all the experiments were performed in triplicates.

Results

Analysis of the main components of PIW

GC analysis showed that PIW mainly consists of four compounds, namely tetracosanol, hexacosanol, octacosanol, and triacontanol (Fig. 2a, Fig. 2b). Their concentration in the obtained PIW was 170.6 mg/L of tetracosanol, 644.4 mg/L of hexacosanol, 515.5 mg/L of octacosanol, and 170.9 mg/L of triacontanol (Fig. 2c, Fig. 2d).

Fig. 2
figure 2

Composition analysis of policosanol derived from insect wax (PIW). a Chemical structures of tetracosanol, hexacosanol, octacosanol and triacontanol. b Chromatogram of standard compounds (200 mg/L) obtained using gas chromatography (GC). c Chromatogram of PIW obtained using GC. d Concentrations of the different components of PIW

Paralysis assay of PIW-fed transgenic C. elegans

We investigated the protective role played by PIW against Aβ-induced toxicity using the transgenic C. elegans strain, CL4176. Our results indicated that the CL4176 worms fed PIW showed a reduced rate of paralysis compared to that of control worms. PIW treatment also rescued paralysis in worms (Fig. 3a) and allowed complete sinusoidal motion (Fig. 3b). However, this restoration by PIW was not dose-dependent. Furthermore, PIW at 2 μg/mL concentration was found to be the most effective dose, wherein the rate of paralysis was reduced from 86.87% (control worms) to 66.97% at 48 h after temperature elevation (Fig. 3c). Based on these results, we used concentrations of 2 and 3 μg/mL of PIW in the subsequent experiments.

Fig. 3
figure 3

Paralysis assay of the Caenorhabditis elegans strain CL4176 fed varying concentrations of policosanol derived from insect wax (PIW). a Representative image of a paralyzed CL4176 worm. b Representative image of a non-paralyzed CL4176 worm. c Paralysis rate of the CL4176 worms fed different concentrations of PIW, and control worms at 48 h after temperature elevation. *P < 0.05 and **P < 0.01 compared to those in the untreated control

Life-span analysis of PIW-fed transgenic C. elegans

Life-span analysis showed that PIW treatment significantly increased the survival rate of the transgenic worms (Fig. 4a). Furthermore, a significant (P value < 0.001) increase in the mean life-span of PIW-fed worms, compared to that of the control group, was observed (Fig. 4b). The average life-span of the control group was 6.2 d, whereas that of the PIW-fed (3 μg/mL) group was 7.8 d.

Fig. 4
figure 4

Life-span assay of the transgenic Caenorhabditis elegans strain CL4176 fed 2 or 3 μg/mL of policosanol derived from insect wax (PIW). a Survival curves of worms from the two treatment groups and the control group. b Average survival rate of worms from the two treatment groups and the control group. **P < 0.01 and ***P < 0.001 compared to those in the untreated control group

SOD and ROS levels in PIW-fed transgenic C. elegans strain CL4176

Our results demonstrated that PIW treatment decreased and increased the SOD levels at 2 and 3 μg/mL concentrations, respectively, compared to that in the control group; however, these changes were statistically insignificant (Fig. 5a). Similarly, PIW treatment in the C. elegans strain CL4176 had no effect on ROS levels (Fig. 5b).

Fig. 5
figure 5

Levels of superoxide dismutase (SOD) and reactive oxygen species (ROS) in policosanol derived from insect wax (PIW)-fed (2 or 3 μg/mL) Caenorhabditis elegans strain CL4176. a SOD and in PIW-fed (2 or 3 μg/mL) the C. elegans strain CL4176, 48 h after temperature elevation. b ROS in PIW-fed (2 or 3 μg/mL) the C. elegans strain CL4176, 48 h after temperature elevation

Relative mRNA expression levels in PIW-fed transgenic C. elegans strain CL4176

We estimated the expression levels of amyloid precursor-like protein (apl-1) and the genes involved in various pathways associated with longevity and alleviation of AD-related symptoms, in the PIW-fed transgenic C. elegans strain CL4176. The expression of genes involved in redox homeostasis (daf-16 and sod-3), heat shock response (hsf-1 and hsp-16.2), anti-oxidative stress (skn-1 and gcs-1), lipid metabolism (lips-17 and lipl-4), and branched-chain amino acids (BCAAs) metabolism (bcat-1) was estimated. The results showed that PIW downregulated the expression of apl-1 (0.87 ± 0.05 fold; P < 0.05) (Fig. 6a), which is the only ortholog of human amyloid precursor protein (APP), and bcat-1(0.73 ± 0.12 fold; P < 0.05) (Fig. 6b). However, PIW treatment had no significant effect on the expression of daf-16, sod-3, lipl-4, and lips-17 (Fig. 6c-f), whereas, it significantly upregulated hsf-1, hsp-16.2, skn-1, and gcs-1 (1.18 ± 0.12 fold; 3.18 ± 1.64 fold; 1.44 ± 0.31 fold; 1.60 ± 0.37 fold; P < 0.05) (Fig. 6g-j).

Fig. 6
figure 6

Effect of policosanol derived from insect wax (PIW) on the relative expression of genes in CL4176. Relative expression of the genes involved in various pathways associated with longevity in the transgenic Caenorhabditis elegans strain CL4176. Expression levels of (a) apl-1 (b) bcat-1 (c) daf-16 (d) sod-3 (e) lipl-4 (f) lips-17 (g) hsf-1 (h) hsp-16.2 (i) skn-1, and (j) gcs-1. *P < 0.05 compared to that in the control group

Discussion

In this study, we employed a transgenic C. elegans AD model (CL4176) to evaluate the pharmacological effects of PIW on counteracting Aβ-induced toxicity (Fig. 7). Our results indicate that PIW alleviates paralysis by decreasing the levels of amyloid formation and extending the lifespan of the transgenic C. elegans by modulating the pathways associated with heat shock response, anti-oxidative stress, and glutamine cysteine synthetase.

Fig. 7
figure 7

Flow-chart summary of the article

CL4176 is a commonly used in vivo model for the experimental evaluation of drugs for the treatment of AD [46, 47]. It has been used to study the effects of Ginkgo biloba extract EGb761 [48], traditional Chinese medicine Liu Wei Di Huang Wan [49], tetracycline [50], and lignans from the roots of Acorus tatarinowii [51], against Aβ-induced toxicity. The studies exploring the longevity and alleviation of paralysis in CL4176 have mainly focused on pathways related to redox homeostasis, heat shock response, oxidative stress, and insulin signaling [49, 52,53,54,55,56]. Previous studies demonstrated that the active substances mainly function by targeting the redox homeostasis and anti-oxidative stress pathways in the transgenic C. elegans AD model, and alleviate paralysis by increasing the activity of antioxidant enzymes, such as SOD and catalase, and upregulating the expression of daf-16, ctl-1, hsp-16.2, sod-3, and sir-2.1 [49, 54, 55, 57,58,59]. Our study showed that PIW treatment led to the downregulation of apl-1, indicating a reduction in the AD-associated aggregation of APP [60,61,62]. However, PIW treatment had no significant effect on SOD and the expression of related genes in transgenic C. elegans; hence, the reduction in Aβ protein aggregation may be attributed to the induced expression of the genes related to heat shock response, anti-oxidative stress, and glutamine cysteine synthetase pathways. The heat shock factor HSF-1 is known to regulate protein folding and gene expression in response to heat stress and is associated with longevity [63]. Furthermore, it acts as a transcription factor for the genes associated with longevity and Aβ-induced toxicity in transgenic C. elegans [52]. Small heat shock proteins are low molecular weight polypeptides with chaperone-like activity that increase the survival of C. elegans under stress, and are induced by Aβ expression [64]. Since the heat shock response of C. elegans is controlled by neurons, hsp-16.2, which is induced by Aβ protein, protects against the abnormal accumulation of toxic proteins [65]. Furthermore, the overexpression of hsp-16.2 is known to inhibit Aβ-induced toxicity [66]. In our study, PIW enhanced the mRNA levels of hsf-1 and its classical target gene hsp-16.2, indicating that PIW can regulate the transcriptional activity of hsf-1, which may explain the counteracting effect of PIW against Aβ-induced cytotoxicity. Oxidative stress is one of the key factors in the process of ageing and has been reported to play a crucial role in the pathophysiology of AD [67, 68]. Additionally, it has been shown that oxidative stress-induced damages often occur near amyloid plaques in the brain tissues of AD patients [69]. The upregulation of skn-1 and gcs-1 in our study suggested that the elevation of antioxidant stress in transgenic C. elegans may be one of the mechanisms that protect it from Aβ-induced toxicity. In the brain, the glutamate level is tightly regulated through metabolite exchange in neuronal, astrocytic, and endothelial cells. In the brain of an AD patient, Aβ can interrupt the effective uptake of glutamate by astrocytes, in turn evoking a cascade of events that leads to neuronal swelling, destruction of membrane integrity, and ultimately cell death [70]. The branched chain aminotransferase (BCAT) enzyme plays an integral role in regulating the brain glutamate levels [71]. In AD patients, an increase in the level of BCAT has been reported in the hippocampus [72]. Furthermore, the altered expression of bcat-1 in C. elegans leads to increase in the levels of BCAAs, which has been reported to promote longevity in C. elegans [73]. Therefore, downregulation of bcat-1 by PIW treatment in our study may have played a role in the remission of AD-related symptoms. Hence, a detailed study on the BCAA metabolism pathway may provide a novel approach for the treatment of AD. For decades, treatment strategies related to AD have focused on the amelioration of Aβ toxicity and promotion of longevity-related pathways. However, these treatment strategies are challenging and have failed to demonstrate efficacy. Although a few studies have indicated the importance of glutamate pathway in the brain of an AD patient [70], it has been not been explored as a treatment option for AD. We found that PIW can reduce the expression of bcat-1, which has been reported to be linked to various pathological states, including cell proliferation [74], decreased survival of septic mice [75], and increased accumulation of liver fat [76]. Although our study demonstrates the effect of PIW on the glutamate metabolism pathway in the transgenic C. elegans strain CL4176 and may help in drug development, further studies are required to identify the detailed underlying mechanisms.

Conclusion

In conclusion, the efficacy of policosanol depends on the source, purity, and composition of the preparation. We explored the effect of PIW treatment on alleviation of AD-related symptoms and associated preliminary mechanisms. Additionally, for the first time, we investigated the role of PIW in reducing Aβ-induced toxicity in the transgenic C. elegans strain CL4176. We showed that policosanol, which is generally thought to reduce LDL and increase HDL cholesterol levels in the blood, had no significant effect on the expression of lipid metabolism-related genes (lips-17 and lipl-4) in the transgenic AD model. Furthermore, PIW reduced the Aβ-induced toxicity in transgenic C. elegans by upregulating the expression of genes related to heat shock response and oxidative stress, and downregulating the expression of bcat-1, which is involved in the glutamine cysteine synthetase pathway and should be explored further as a drug target in AD. In the future, we will conduct the pharmacodynamic study of each component of PIW, including tetracosanol, hexacosanol, octacosanol, and triacontanol. Nevertheless, the effectiveness of PIW needs to be explored in murine models and humans.

Availability of data and materials

The data is available and will be provided upon request to the corresponding author.

Abbreviations

AD:

Alzheimer’s disease

PIW:

Policosanol derived from insect wax

Aβ:

β-amyloid

PVDF:

Poly (1,1-difluoroethylene)

LDL:

Low-density lipoprotein

HDL:

High-density lipoprotein

NGM:

Nematode growth medium

SOD:

Superoxide dismutase

ROS:

Reactive oxygen species

PBS:

Phosphate-buffered saline

H2DCF-DA:

2′,7′-dichlorodihydro-fluorescein diacetate

BCAT:

Branched chain aminotransferase

BCAAs:

Branched-chain amino acids

References

  1. Chen XM. Natural population ecology of Ericerus Pela. Beijing: Science Press; 2011. p. 1–8.

    Google Scholar 

  2. Zou SW. A history of Chinese entomology. Beijing: Science Press; 1982. p. 112–4.

    Google Scholar 

  3. Zhou Y. The history of entomology in China. Shanxi: Xian Entomotaxonomia; 1980. p. 41–2.

    Google Scholar 

  4. Yang P, Chen XM. Protein profiles of Chinese white wax scale, Ericerus pela, at the male pupal stage by high-throughput proteomics. Arch Insect Biochem Physiol. 2014;87(4):214–33. https://doi.org/10.1002/arch.21191.

    Article  CAS  PubMed  Google Scholar 

  5. Yang P, Chen XM, Liu WW, Feng Y, Sun T. Transcriptome analysis of sexually dimorphic Chinese white wax scale insects reveals key differences in developmental programs and transcription factor expression. Sci Rep. 2015;5(1):1–8. https://doi.org/10.1038/srep08141.

    Article  CAS  Google Scholar 

  6. Hou XY, Cao MH, Gong J, Li N, Gao A, Jia X, et al. Overview of pharmacological research of insect wax. J Anhui Agric Sci. 2011;39(5):2817–8.

    Google Scholar 

  7. Wang ZD, Feng Y, Ma LY, Li X, Ding WF, Chen XM. Hair growth promoting effect of white wax and policosanol from white wax on the mouse model of testosterone-induced hair loss. Biomed Pharmacother. 2017;89:438–46. https://doi.org/10.1016/j.biopha.2017.02.036.

    Article  CAS  PubMed  Google Scholar 

  8. Li SZ. The Compendium of Materia Medica (Ming Dynasty, 1578 C.E.). Beijing: Commercial Press; 1926.

  9. Feng Y, Chen XM, Ma Y, He Z. Experimental study on immunomodulation of white wax scale (Ericerus pela Chavannes). For Res. 2006;19(2):221–4.

    Google Scholar 

  10. Feng Y, He Z, Li X, Cheng ZY, Sun L. Immunomodulatory and antitumor activities of polysaccharide from Chinese white wax scale. For Res. 2014;27(3):388–92.

    Google Scholar 

  11. He Z, Li X, Sun L, Chen ZY, Feng Y. Antioxidant activities of five insect polysaccharides in vitro. J Environ Entomol. 2015;37(1):61–7.

    Google Scholar 

  12. He Z, Sun L, Feng Y, Cheng XM. The extraction of polysaccharide from white wax scale and analysis of monosaccharide compositions. For Res. 2008;21(6):792–6. https://doi.org/10.3901/JME.2008.09.177.

    Article  Google Scholar 

  13. Lin L, Zhou Y, Li H, Pang D, Zhang L, Lu X, et al. Polysaccharide extracted from Chinese white wax scale ameliorates 2,4-dinitrochlorobenzene-induced atopic dermatitis-like symptoms in BALB/c mice. Saudi Pharm J. 2017;25(4):625–32. https://doi.org/10.1016/j.jsps.2017.04.035.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Ma LY, Wang YQ, Zhang ZQ, Gan J, Zheng H, Guo YH, et al. Preparation of Policosanol from insect wax by reduction method. Chem Industry Forest Prod. 2009;29(5):6–10. https://doi.org/10.1360/972009-1551.

    Article  Google Scholar 

  15. Lin Y, Rudrum M, van der Wielen RP, Trautwein EA, McNeill G, Sierksma A, et al. Wheat germ policosanol failed to lower plasma cholesterol in subjects with normal to mildly elevated cholesterol concentrations. Metabolism. 2004;53(10):1309–14. https://doi.org/10.1016/j.metabol.2004.05.006.

    Article  CAS  PubMed  Google Scholar 

  16. Hwang KT, Weller CL, Cuppett SL, Hanna MA. Policosanol contents and composition of grain Sorghum kernels and dried distillers grains. Cereal Chem. 2004;81(3):345–9. https://doi.org/10.1094/CCHEM.2004.81.3.345.

    Article  CAS  Google Scholar 

  17. Harrabia S, Boukhchinaa S, Mayerb PM, Kallela H. Policosanol distribution and accumulation in developing corn kernels. Food Chem. 2009;115(3):918–23. https://doi.org/10.1016/j.foodchem.2008.12.098.

    Article  CAS  Google Scholar 

  18. Adhikari P, Hwang KT, Park JN, Kim CK. Policosanol content and composition in Perilla seeds. J Agric Food Chem. 2006;54(15):5359–62. https://doi.org/10.1021/jf060688k.

    Article  CAS  PubMed  Google Scholar 

  19. Marinangeli CPF, Jones PJH, Kassis AN, Eskin MNA. Policosanols as Nutraceuticals: fact or fiction. Criti Rev Food Sci Nutr. 2010;50(3):259–67. https://doi.org/10.1080/10408391003626249.

    Article  CAS  Google Scholar 

  20. Carbajal D, Arruzazabala ML, Valdes S, Mas R. Effect of policosanol on platelet aggregation and serum levels of arachidonic acid metabolites in healthy volunteers. Prostaglandins Leukot Essent Fatty Acids. 1998;58(1):61–4. https://doi.org/10.1016/s0952-3278(98)90130-2.

    Article  CAS  PubMed  Google Scholar 

  21. Kabir Y, Kimura S. Tissue distribution of (8-14C)-octacosanol in liver and muscle of rats after serial administration. Ann Nutr Metabol. 1995;39(5):279–84. https://doi.org/10.1159/000177873.

    Article  CAS  Google Scholar 

  22. Stüsser R, Batista J, Padrón R, Sosa F, Pereztol O. Longterm therapy with octacosanol improves treadmill exercise-ECG testing performance of coronary heart disease patients. Int J Clin Pharmacol Ther. 1998;36(9):469–73. https://doi.org/10.1002/zaac.200700297.

    Article  CAS  PubMed  Google Scholar 

  23. Harrabi S, Ferchichi A, Bacheli A, Fellah H. Policosanol composition, antioxidant and anti-arthritic activities of milk thistle (Silybium marianum L.) oil at different seed maturity stages. Lipids Health Dis. 2018;17(1):1–7. https://doi.org/10.1186/s12944-018-0682-z.

    Article  CAS  Google Scholar 

  24. Varady KA, Wang Y, Jones PJH. Role of Policosanols in the prevention and treatment of cardiovascular disease. Nutr Rev. 2010;61(11):376–83. https://doi.org/10.1301/nr.2003.nov.376-383.

    Article  Google Scholar 

  25. Arruzazabala ML, Molina V, Mas R, Fernández L, Carbajal D, Valdés S, et al. Antiplatelet effects of policosanol (20 and 40 mg/day) in healthy volunteers and dyslipidaemic patients. Clin Exp Pharmacol Physiol. 2002;29(10):891–7. https://doi.org/10.1046/j.1440-1681.2002.03746.x.

    Article  CAS  PubMed  Google Scholar 

  26. Dullens SP, Mensink RP, Bragt MC, Kies AK, Plat J. Effects of emulsified policosanols with different chain lengths on cholesterol metabolism in heterozygous LDL receptor-deficient mice. J Lipid Res. 2008;49(4):790–6. https://doi.org/10.1194/jlr.M700497-JLR200.

    Article  CAS  PubMed  Google Scholar 

  27. Affuso F, Ruvolo A, Micillo F, Sacca L, Fazio S. Effects of a nutraceutical combination (berberine, red yeast rice and policosanols) on lipid levels and endothelial function randomized, double-blind, placebo-controlled study. Nutr Metab Cardiovasc Dis. 2010;20(9):656–61. https://doi.org/10.1016/j.numecd.2009.05.017.

    Article  CAS  PubMed  Google Scholar 

  28. Agostoni C, Bresson JL, Fairweather-Tait S, Flynn A, Golly I, Korhonen H. Scientific opinion on the substantiation of health claims related to policosanols from sugar cane wax and maintenance of normal blood LDL-cholesterol concentrations (ID 1747, 1748, 1864, 1951, 1954, 4693) and maintenance of normal blood HDL-cholesterol co. Eur Food Safety Authority. 2011;9(6):2255. https://doi.org/10.2903/j.efsa.2011.2255.

    Article  CAS  Google Scholar 

  29. Elseweidy MM, Zein N, Aldhamy SE, Elsawy MM, Saeid SA. Policosanol as a new inhibitor candidate for vascular calcification in diabetic hyperlipidemic rats. Exp Biol Med. 2016;241(17):1943–9. https://doi.org/10.1177/1535370216659943.

    Article  CAS  Google Scholar 

  30. Lee JY, Choi HY, Kang YR, Chang HB, Chun HS, Lee MS, et al. Effects of long-term supplementation of policosanol on blood cholesterol/glucose levels and 3-hydroxy-3-methylglutaryl coenzyme a reductase activity in a rat model fed high cholesterol diets. Food Sci Biotechnol. 2016;25(3):899–904. https://doi.org/10.1007/s10068-016-0147-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sánchez-López J, Illnait-Ferrer J, Mas-Ferreiro R, Mendoza-Castaño S, Fernández-Dorta L, Mesa-Angarica M, et al. Long-term effect of policosanol on the functional recovery of non-cardioembolic ischemic stroke patients: a one year study. Rev Neurol. 2017;64(4):153–61.

    Google Scholar 

  32. Ma JJ, Ma LY, Zhang H, Zhang ZQ, Wang YQ. The preparation and evaluation of efficacy on skin wound healing in mice of insect wax compound ointment. J Environ Entomol. 2018;40(6):1238–47.

    Google Scholar 

  33. Wang ZD, Feng Y, Li X, Ding WF, Chen XM. Effect of white wax and Policosanol from white wax tween aqueous solution on HFDPCS. For Res. 2017;30(1):41–5. https://doi.org/10.13275/j.cnki.lykxyj.2017.01.006.

    Article  Google Scholar 

  34. Wang ZD, LI X, Ding WF, Sun L, Feng Y. Mechanism of white wax on treating Seborrheic alopecia. Chin J Ethnomed Ethnopharm. 2019;28(10):17–21.

    Google Scholar 

  35. Wang T, Liu YY, Wang X, Yang N, Zhu HB, Zuo PP. Protective effects of octacosanol on 6-hydroxydopamine-induced parkinsonism in rats via regulation of ProNGF and NGF signaling. Acta Pharmacol Sin. 2010;31(7):765–74. https://doi.org/10.1038/aps.2010.69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Renoudet VV, Costa-Mallen P, Hopkins E. A diet low in animal fat and rich in N-hexacosanol and fisetin is effective in reducing symptoms of Parkinson’s disease. J Med Food. 2012;15(8):758–61. https://doi.org/10.1089/jmf.2012.0060.

    Article  CAS  PubMed  Google Scholar 

  37. Moosbrugger I, Bischoff P, Beck JP, Borg J. Studies on the immunological effects of fatty alcohols: I. Effects of n-hexacosanol on murine macrophages in culture. Int J Immunopharmacol. 1992;14(2):293–302. https://doi.org/10.1016/0192-0561(92)90042-J.

    Article  CAS  PubMed  Google Scholar 

  38. Goedert M, Spillantini MG. A century of Alzheimer’s disease. Science. 2006;314(5800):777–81. https://doi.org/10.1126/science.1132814.

    Article  CAS  PubMed  Google Scholar 

  39. Selkoe DJ. Alzheimer’s disease genes, proteins, and therapy. Physiol Rev. 2001;8(2):741–66. https://doi.org/10.1152/physrev.2001.81.2.741.

    Article  Google Scholar 

  40. Snyder SW, Ladror US, Wade WS, Wang GT, Barrett LW, Matayoshi ED, et al. Amyloid beta aggregation selective inhibition of aggregation in mixtures of amyloid with different chain lengths. Biophys J. 1994;67(3):1216–28. https://doi.org/10.1016/s0006-3495(94)80591-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Shinagawa S, Shigeta M. Acetylcholinesterase inhibitors for treatment of Alzheimer’s disease. Brain Nerve. 2014;66(5):507–16. https://doi.org/10.1155/2012/728983.

    Article  CAS  PubMed  Google Scholar 

  42. Folch J, Busquets O, Ettcheto M, Sanchez-Lopez E, Castro-Torresa RD, Verdaguer E, et al. Memantine for the treatment of dementia: a review on its current and future applications. J Alzheimers Dis. 2018;62(3):1223–40. https://doi.org/10.3233/JAD-170672.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Anand R, Gill KD, Mahdi AA. Therapeutics of Alzheimer's disease: past, present and future. Neuropharmacology. 2014;76:27–50. https://doi.org/10.1016/j.neuropharm.2013.07.004.

    Article  CAS  PubMed  Google Scholar 

  44. Misra S, Medhi B. Drug development status for Alzheimer's disease: present scenario. Neurol Sci. 2013;34(6):831–9. https://doi.org/10.1080/10408391003626249.

    Article  CAS  PubMed  Google Scholar 

  45. Lemere CA, Masliah E. Can Alzheimer disease be prevented by amyloid-β immunotherapy? Nat Rev Neurol. 2010;6(2):108–19. https://doi.org/10.1038/nrneurol.2009.21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Link CD. Expression of human beta-amyloid peptide in transgenic Caenorhabditis elegans. Proc Natl Acad Sci U S A. 1995;92(27):9368–72. https://doi.org/10.1073/pnas.92.20.9368.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Link CD, Johnson CJ, Fonte V, Paupard M-C, Hall DH, Styren S, et al. Visualization of fibrillar amyloid deposits in living, transgenic Caenorhabditis elegans animals using the sensitive amyloid dye, X-34. Neurobiol Aging. 2001;22(2):217–26. https://doi.org/10.1016/s0197-4580(00)00237-2.

    Article  CAS  PubMed  Google Scholar 

  48. Wu Y, Wu Z, Butko P, Christen Y, Lambert MP, Klein WL, et al. Amyloid-beta-induced pathological behaviors are suppressed by Ginkgo biloba extract EGb 761 and ginkgolides in transgenic Caenorhabditis elegans. J Neurosci. 2006;26(50):13102–13. https://doi.org/10.1523/JNEUROSCI.3448-06.2006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sangha JS, Sun X, Wally OS, Zhang K, Ji X, Wang Z, et al. Liuwei Dihuang (LWDH), a traditional Chinese medicinal formula, protects against beta-amyloid toxicity in transgenic Caenorhabditis elegans. PLoS One. 2012;7(8):1–10. https://doi.org/10.1371/journal.pone.0043990.

    Article  CAS  Google Scholar 

  50. Diomede L, Cassata G, Fiordaliso F, Salio M, Ami D, Natalello A, et al. Tetracycline and its analogues protect Caenorhabditis elegans from β amyloid-induced toxicity by targeting oligomers. Neurobiol Dis. 2010;40(2):424–31. https://doi.org/10.1016/j.nbd.2010.07.002.

    Article  CAS  PubMed  Google Scholar 

  51. Luo XH, Zhang YY, Chen XY, Sun ML, Li S, Wang HB. Lignans from the roots of Acorus tatarinowii Schott ameliorate beta amyloid-induced toxicity in transgenic Caenorhabditis elegans. Fitoterapia. 2016;108:5–8. https://doi.org/10.1016/j.fitote.2015.11.010.

    Article  CAS  PubMed  Google Scholar 

  52. Tullet JMA, Hertweck M, An JH, Baker J, Ji YH, Shu L, et al. Direct inhibition of the longevity-promoting factor SKN-1 by insulin-like signaling in C. elegans. Cell. 2008;132(6):1025–38. https://doi.org/10.1016/j.cell.2008.01.030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Abbas S, Wink M. Epigallocatechin gallate inhibits beta amyloid oligomerization in Caenorhabditis elegans and affects the daf-2/insulin-like signaling pathway. Phytomedicine. 2010;17(11):902–9. https://doi.org/10.1016/j.phymed.2010.03.008.

    Article  CAS  PubMed  Google Scholar 

  54. Zhang XG, Wang X, Zhou TT, Wu XF, Peng Y, Zhang WQ, et al. Scorpion venom heat-resistant peptide protects transgenic Caenorhabditis elegans from β-amyloid toxicity. Front Pharmacol. 2016;7:1–9. https://doi.org/10.3389/fphar.2016.00227.

    Article  CAS  Google Scholar 

  55. Li F, Cui XD, Ma XL, Li J, Wang ZH. Glutaredoxin delays the toxicity induced by β-amyloid in AD Transgenic C.elegans in AD Transgenic C.elegans. Chin J Biochem Mol Biol. 2018;34(8):844–53.

    Google Scholar 

  56. Kenyon CJ. The genetics of ageing. Nature. 2010;464(7288):504–12. https://doi.org/10.1038/nature08980.

    Article  CAS  PubMed  Google Scholar 

  57. Gutierrez-Zepeda A, Santell R, Wu Z, Brown M, Wu Y, Khan I, et al. Soy isoflavone glycitein protects against beta amyloid-induced toxicity and oxidative stress in transgenic Caenorhabditis elegans. BMC Neurosci. 2005;6(54):1–9. https://doi.org/10.1186/1471-2202-6-54.

    Article  CAS  Google Scholar 

  58. Chen W, Lin HR, Wei CM, Luo XH, Sun ML, Yang ZZ, et al. Echinacoside, a phenylethanoid glycoside from Cistanche deserticola, extends lifespan of Caenorhabditis elegans and protects from Aβ-induced toxicity. Biogerontology. 2018;19(1):47–65. https://doi.org/10.1007/s10522-017-9738-0.

    Article  CAS  PubMed  Google Scholar 

  59. An JH, Blackwell TK. SKN-1 links C. elegans mesendodermal specification to a conserved oxidative stress response. Genes Dev. 2003;17(15):1882–93. https://doi.org/10.1101/gad.1107803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wiese M, Antebi A, Zheng H. Intracellular trafficking and synaptic function of APL-1 in Caenorhabditis elegans. PLoS One. 2010;5(9):1–12. https://doi.org/10.1371/journal.pone.0012790.

    Article  CAS  Google Scholar 

  61. Ewald CY, Raps DA, Li C. APL-1, the Alzheimer's amyloid precursor protein in Caenorhabditis elegans, modulates multiple metabolic pathways throughout development. Genetics. 2012;191(2):493–507. https://doi.org/10.1534/genetics.112.138768.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lorenzo N, Altruda F, Silengo L, Del Carmen Dominguez M. APL-1, an altered peptide ligand derived from heat-shock protein, alone or combined with methotrexate attenuates murine collagen-induced arthritis. Clin Exp Med. 2017;17(2):209–16. https://doi.org/10.1007/s10238-016-0412-7.

    Article  CAS  PubMed  Google Scholar 

  63. Douglas PM, Baird NA, Simic MS, Uhlein S, McCormick MA, Wolff SC, et al. Heterotypic Signals from Neural HSF-1 Separate Thermotolerance from Longevity. Cell Rep. 2015;12(7):1196–204. https://doi.org/10.1016/j.celrep.2015.07.026.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Link CD, Cypser JR, Johnson CJ, Johnson TE. Direct observation of stress response in Caenorhabditis elegans using a reporter transgene. Cell Stress Chaperones. 1999;4(4):235–42. https://doi.org/10.1379/1466-1268(1999)004<0235:doosri>2.3.co;2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Prahlad V, Cornelius T, Morimoto RI. Regulation of the cellular heat shock response in Caenorhabditis elegans by thermosensory neurons. Science. 2008;320(5877):811–4. https://doi.org/10.1126/science.1156093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Fonte V, Kipp DR, Yerg J, Merin D, Link CD. Suppression of in vivo Beta-amyloid peptide toxicity by overexpression of the HSP-16.2 small chaperone protein. J Biol Chem. 2008;283(2):784–91. https://doi.org/10.1074/jbc.M703339200.

    Article  CAS  PubMed  Google Scholar 

  67. Gerstbrein B, Stamatas G, Kollias N, Driscoll M. In vivo spectrofluorimetry reveals endogenous biomarkers that report healthspan and dietary restriction in Caenorhabditis elegans. Aging Cell. 2005;4(3):127–37. https://doi.org/10.1111/j.1474-9726.2005.00153.x.

    Article  CAS  PubMed  Google Scholar 

  68. Zhao Y, Zhao BL. Oxidative stress and the pathogenesis of Alzheimer's disease. Oxid Med Cell Longev. 2013;2013:1–10. https://doi.org/10.1155/2013/316523.

    Article  CAS  Google Scholar 

  69. Lovell MA, Ehmann WD, Butler SM, Markesbery WR. Elevated thiobarbituric acid-reactive substances and antioxidant enzyme activity in the brain in Alzheimer's disease. Neurology. 1995;45(8):1594–601. https://doi.org/10.1212/wnl.45.8.1594.

    Article  CAS  PubMed  Google Scholar 

  70. Conway ME. Alzheimer’s disease: targeting the glutamatergic system. Biogerontology. 2020;21(3):257–74. https://doi.org/10.1007/s10522-020-09860-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Butterfield DA, Pocernich CB. The Glutamatergic system and Alzheimer's disease. CNS Drugs. 2003;17(9):641–52. https://doi.org/10.2165/00023210-200317090-00004.

    Article  CAS  PubMed  Google Scholar 

  72. Hull J, Patel V, El Hindy M, Lee C, Odeleye E, Hezwani M, et al. Regional increase in the expression of the BCAT proteins in Alzheimer's disease brain: implications in glutamate toxicity. J Alzheimers Dis. 2015;45(3):891–905. https://doi.org/10.3233/JAD-142970.

    Article  CAS  PubMed  Google Scholar 

  73. Mansfeld J, Urban N, Priebe S, Groth M, Frahm C, Hartmann N, et al. Branched-chain amino acid catabolism is a conserved regulator of physiological ageing. Nat Commun. 2015;6(1):1–12. https://doi.org/10.1038/ncomms10043.

    Article  CAS  Google Scholar 

  74. Tönjes M, Barbus S, Park YJ, Wang W, Schlotter M, Lindroth AM, et al. BCAT1 promotes cell proliferation through amino acid catabolism in gliomas carrying wild-type IDH1. Nat Med. 2013;19(7):1–11. https://doi.org/10.1016/S0959-8049(14)50309-7.

    Article  Google Scholar 

  75. Lang CH, Lynch CJ, Vary TC. BCATm deficiency ameliorates endotoxin-induced decrease in muscle protein synthesis and improves survival in septic mice. Am J Physiol Regul Integr Comp Physiol. 2010;299(3):935–44. https://doi.org/10.1152/ajpregu.00297.2010.

    Article  CAS  Google Scholar 

  76. Greco D, Kotronen A, Westerbacka J, Puig O, Arkkila P, Kiviluoto T. Gene expression in human NAFLD. Am J Physiol Gastrointest Liver Physiol. 2008;294(5):1281–7. https://doi.org/10.1152/ajpgi.00074.2008.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the Chinese Academy of Forestry (No. CAFYBB2018ZB007).

Author information

Authors and Affiliations

Authors

Contributions

YF and XC conceived the research theme and supervised the implementation. XZ, CM, LS, ZH, and XL designed the method and performed the experiments. XZ wrote the manuscript. YF and JG contributed to the revision of the manuscript. All authors have reviewed and approved the final manuscript.

Corresponding author

Correspondence to Ying Feng.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1.

Results of preliminary experiments

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, X., Ma, C., Sun, L. et al. Effect of policosanol from insect wax on amyloid β-peptide-induced toxicity in a transgenic Caenorhabditis elegans model of Alzheimer’s disease. BMC Complement Med Ther 21, 103 (2021). https://doi.org/10.1186/s12906-021-03278-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-021-03278-2

Keywords