AU2014100319A4 - A novel autophagy enhancer for treatment of neurodegenerative diseases - Google Patents

A novel autophagy enhancer for treatment of neurodegenerative diseases Download PDF

Info

Publication number
AU2014100319A4
AU2014100319A4 AU2014100319A AU2014100319A AU2014100319A4 AU 2014100319 A4 AU2014100319 A4 AU 2014100319A4 AU 2014100319 A AU2014100319 A AU 2014100319A AU 2014100319 A AU2014100319 A AU 2014100319A AU 2014100319 A4 AU2014100319 A4 AU 2014100319A4
Authority
AU
Australia
Prior art keywords
onjisaponin
autophagy
cells
neurodegenerative diseases
synuclein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2014100319A
Inventor
Yuen Kwan Law
Liang Liu
Kam Wai Wong
An Guo Wu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Macau University of Science and Technology
Original Assignee
Macau University of Science and Technology
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US14/231,671 external-priority patent/US9005677B2/en
Application filed by Macau University of Science and Technology filed Critical Macau University of Science and Technology
Application granted granted Critical
Publication of AU2014100319A4 publication Critical patent/AU2014100319A4/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Landscapes

  • Medicines Containing Plant Substances (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A Novel Autophagy Enhancer for Treatment of Neurodegenerative Diseases Use and application of Onjisaponin B derived and isolated from Radix Polygalae as novel autophagy enhancer are provided. A method of preventing, treating and/or delaying the onset of neurodegenerative diseases comprising administering an effective amount of Onjisaponin B is also provided. <DocRef#00 119901-RM > M006.023.UMDAU 20 Spec Draft As Filed

Description

A Novel Autophagy Enhancer for Treatment of Neurodegenerative Diseases FIELD OF INVENTION [0001] This invention relates to a novel autophagy enhancer and the use thereof in treating neurodegenerative diseases. BACKGROUND OF INVENTION [0002]Autophagy is a cellular process that involves the formation of an isolated membrane to form double membrane vesicles (autophagosomes) that sequesters the cytoplasmic materials. Followed by fusion of the autophagosome with lysosome to form an autolysosome, all the engulfed materials are degraded by lysosomal hydrolases to recycle intracellular nutrients and energy [1, 2]. Autophagy-lysosome and ubiquitin proteasome pathways are the two major protein degradation pathways in cells [3]. While short-lived nuclear and cytosolic proteins are degraded by proteasomes, large membrane proteins, oligomers and aggregates which cannot pass through the narrow pore of the proteasome barrel are degraded by autophagy [4]. [0003]A variety of neurodegenerative diseases are caused by toxic, aggregate-prone or oligomeric proteins [5-9]. For example, Huntington's disease (HD) is caused by an over 35 CAG trinucleotide repeat expansion, which results in a long mutant polyQ tract in the huntingtin protein. These polyQ expansions are highly associated with the aggregate formation and toxicity [10, 11]. Autophagy, however, can reduce mutant huntingtin protein levels and its toxicity in cell and mouse models [6, 7]. Parkinson disease (PD) is caused by A53T or A30P a-synuclein mutants, which are identified as substrates of autophagy, and the clearance of these mutant proteins are also highly dependent on autophagy for removal [6-8, 11-14]. Pharmacological activation of autophagy reduces the levels and toxicity of mutant huntingtin, mutant proteins in spinocerebellar ataxia, mutant a-synuclein and mutant tau in either mouse or drosophila models [5, 15]. Furthermore, autophagy-related gene (ATG) knockdown leads to aggregate formation and toxicity in C.elegans [16, 17]. In addition to the formation of protein aggregates, the accumulation M006.023.UMDAU 1 Spec Draft As Filed of abnormal mitochondria or endoplasmic reticulum, and an increase in the size and number of lipid droplets were observed in ATG gene knockout animal models [14, 18 20]. SUMMARY OF INVENTION [0004] In the light of the foregoing background, it is an object of the present invention to provide a novel autophagy enhancer with potential therapeutic application in neurodegenerative diseases by enhancing the clearance of mutant huntingtin and a synuclein, and reduces a-synuclein oligomerization. As a result, the autophagy enhancer is able to recover from mutant huntingtin or a-synuclein-induced neuronal cell death. [0005]In the first aspect, the present invention relates to a method of preventing, treating and/or delaying the onset of neurodegenerative diseases comprising administering an effective amount of Onjisaponin B to a subject in need thereof. [0006]In one embodiment, Onjisaponin B is isolated from Chinese medicinal herbs, Radix Polygalae. [0007]In another embodiment, the neurodegenerative diseases are caused by the buildup of oligomeric proteins comprising mutant huntingtin and mutant a-synuclein. [0008]In yet another embodiment, the neurodegenerative diseases are selected from a group consisting of Parkinson's disease, and Huntington's disease. [0009]In another aspect of this invention, a use of Onjisaponin B as an autophagy enhancer for treating neurodegenerative diseases is provided. In one embodiment, Onjisaponin B has been described in the foregoing paragraph(s); in another embodiment, the neurodegenerative diseases have also been described in the foregoing paragraph(s). [0010]In yet another aspect, the present invention provides a pharmaceutical composition comprising Onjisaponin B admixed with a pharmaceutical carrier for treating neurodegenerative diseases. In one embodiment, Onjisaponin B has been described in the foregoing paragraph(s); in another embodiment, the neurodegenerative diseases have also been described in the foregoing paragraph(s). M006.023.UMDAU 2 Spec Draft As Filed [0011]In another aspect, the present invention provides a dietary supplement comprising Onjisaponin B admixed with a pharmaceutical carrier for treating neurodegenerative diseases. In one embodiment, Onjisaponin B has been described in the foregoing paragraph(s); in another embodiment, the neurodegenerative diseases have also been described in the foregoing paragraph(s). [0012]The present invention is based on the identification of novel autophagy enhancer, Onjisaponin B as mentioned above. The invention is also based on the neuroprotective effect of Onjisaponin B on neuronal cells via enhancing the clearance of mutant huntingtin and a-synuclein, as well as preventing the oligomerization of a-synuclein. [0013]Onjisaponin B exhibits no significant cytotoxic effect on a rat adrenal pheochromocytoma cells (PC 12) up to 100 gM. It is also relatively non-toxic to mouse, rat and human neuronal cells and brain tissues. [0014]Onjisaponin B is a novel autophagy enhancer isolated from Radix Polygalae and has never been reported before. Also, Onjisaponin B is capable of inducing autophagy in cells and animals. [0015]Onjisaponin B-induced autophagy is dependent on autophagy-related gene 7 (Atg7); further, Onjisaponin B is capable of inducing autophagy in Atg7-dependent manner. [0016]Besides, Onjisaponin B induces autophagy via activation of AMP-activated protein kinase (AMPK) and inhibition of mammalian target of rapamycin (mTOR) signaling. Onjisaponin B is capable of inducing autophagy via modulation of AMPK mTOR signaling pathway. [0017]Further, Onjisaponin B enhances the clearance of mutant huntingtin and mutant A53T a-synuclein; further, Onjisaponin B is capable of enhancing the clearance of mutant huntingtin and mutant A53T a-synuclein. M006.023.UMDAU 3 Spec Draft As Filed [0018] Onjisaponin B enhances the clearance of mutant huntingtin in autophagy-wild type cells, but not in autophagy-deficient cells; further, Onjisaponin B is capable of enhancing the clearance of mutant huntingtin through autophagy induction. [0019]Onjisaponin B reduces the oligomerization of a-synuclein; further, Onjisaponin B is capable of reducing the oligomerization of a-synuclein. [0020] Onjisaponin B reduces toxicity in PC-12 cells expressing either mutant huntingtin or mutant A53T u-synuclein; further, Onjisaponin B is capable of recovering from the mutant huntingtin or mutant A53T a-synuclein-mediated toxicity in neuronal cells or tissues. [0021]Onj isaponin B can be developed as novel neuroprotective agents for patients with neurodegenerative diseases. BRIEF DESCRIPTION OF FIGURES [0022] Fig. 1 shows the chemical structure of Onjisaponin B. [0023] Fig. 2a and Fig. 2b show the results of cytotoxicity study of Onjisaponin B in PC12 cells. [0024] Fig. 3a, Fig. 3b and Fig. 3c show that Onjisaponin B induces autophagic GFP LC3 puncta formation, LC3-II conversion and autophagic flux in PC12 cells. [0025] Fig. 4 shows that Onjisaponin B-induced autophagy is dependent on the presence of autophagy-related gene7 (Atg7). [0026] Fig. 5a and Fig. 5b show that Onjisaponin B activates autophagy through an AMPK-mTOR signaling pathway. [0027] Fig. 6a and Fig. 6b show that Onjisaponin B enhances the clearance of mutant huntingtin and mutant A53T a-synuclein. [0028] Fig. 7a, Fig. 7b and Fig. 7c show that Onjisaponin B enhances the clearance of mutant huntingtin in autophagy-wild type cells, but not in autophagy-deficient cells. M006.023.UMDAU 4 Spec Draft As Filed [0029] Fig. 8 shows that Onjisaponin B inhibits oligomerization of a-synuclein. [0030] Fig. 9a and Fig. 9b show that Onjisaponin B reduces toxicity in PC-12 cells expressing either mutant huntingtin or mutant A53T u-synuclein. DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS [0031] As used herein and in the claims, "comprising" means including the following elements but not excluding others. [0032] In this study, novel autophagy enhancers with neuroprotective effects by evaluating their efficacy in enhancing the clearance of mutant huntingtin and a-synuclein, and in increasing of cells viability, are identified. Through screening of their library of natural product extracts from the Chinese medicinal herbs, inventors have demonstrated that both ethanol extract of Radix Polygalae and its single component, Onjisaponin B, are able to induce autophagy with potential therapeutic application in neurodegeneration. [0033] The formula, molecular weight and accurate mass of Onjisaponin B are summarized in Table 1 below. Table 1 Chemical Name Formula Molecular Weight Accurate MS Onjisaponin B C 75
H
12 0 35 1573.689 1572.698425 [0034] The reference cited throughout this application is identified in square bracket as "[xx]" with xx referring to the number of the corresponding reference on the "References" list. [0035] The following preparations and examples are given to enable those skilled in the art to more clearly understand and to practice the present invention. They should not be considered as limiting the scope of the invention, but merely as being illustrative and representative thereof M006.023.UMDAU 5 Spec Draft As Filed [0036] Example 1 [0037] This example describes in vitro cytotoxicity of Onjisaponin B in a rat adrenal pheochromocytoma cells (PC12). [0038] Cell culture and cytotoxicity assay: The test compound of Onjisaponin B was dissolved in DMSO at a final concentration of 100 mmol/L and stored at -20 'C. Cytotoxicity was assessed using the 3-(4,5-dimethylthiazol-2-yl) -2,5 diphenyltetrazolium bromide assay as described previously [28]. PC-12, 4000 cells were seeded on 96-well plates per well. After overnight pre-incubation, the cells were exposed to different concentrations of Onjisaponin B (0.039 - 100ptmol/L) for 2 days. Subsequently, 10 pL of MTT reagents was added to each well and incubated at 37'C for 4 hours followed by the addition of 100 pL solubilization buffer (10% SDS in 0.01 mol/L HCl) and overnight incubation. Absorbance at 585 nm was determined from each well the next day. For cell viability assay measured by crystal violet staining, PC-12 cells were incubated in 35 mm disc followed by the addition of Onjisaponin B at the indicated concentrations for 24 hours. The cells were then incubated with crystal violet for 10 minutes followed by a ddH 2 0 wash. The stained cells image was captured by CCD digital camera Spot RT3TM under the Nikon ECLIPSE 80i microscope with 4X magnification. Cell viability was quantified by dissolving the stained cells in 10% acetic acid (200 ptL/well). The colorimetric reading of the solute mixture was then determined by spectrophotometer at OD 560 nm. The percentage of cell viability was calculated using the following formula: Cell viability (%) = Cells number treated / Cells number DMSO control x 100. Data was obtained from three independent experiments. [0039]Results: There was no toxicity observed overtly in PC-12 cells treated with Onjisaponin B for 48 hours as revealed by MTT assay as shown in Fig. 2a. Besides, no significant morphological damage was found in PC12 cells treated with Onjisaponin B for 24 hours as revealed by the crystal violet assay as shown in Fig. 2b. [0040] Conclusion: The result shows that Onjisaponin B is non-toxic in rat adrenal pheochromocytoma cells (PC-12). M006.023.UMDAU 6 Spec Draft As Filed [0041] Example 2 [0042] This example describes an in vitro study to demonstrate the autophagic effect of Onjisaponin B. [0043]Quantification of autophagy GFP-LC3 Puncta. GFP-LC3 puncta formation was quantified as described previously [2]. In brief, cells grown on coverslips in a 6-well plate were fixed in 4% paraformaldehyde for 20 minutes at room temperature and then rinsed with PBS. Slides were mounted with FluorSaveTM mounting media (Calbiochem, San Diego, California) and examined by fluorescence microscopy. The number of GFP positive cells with GFP-LC3 puncta formation was examined under the Nikon ECLIPSE 80i microscope. Representative images were captured with CCD digital camera Spot RT3TM (Diagnostic Instruments, Inc., Melville, NY). To quantify autophagy, the percentage of cells with punctate GFP-LC3 fluorescence was calculated by counting the number of the cells with punctate GFP-LC3 fluorescence in GFP-positive cells. A minimum of 150 cells from 3 randomly selected fields was scored. [0044]Detection of autophagic marker protein LC3 conversion. After drug treatments with or without autophagy inhibitor (10 g/mL E64D and pepstain A), cells were harvested and lysed in RIPA buffer (Cell Signaling Technologies Inc., Beverly, MA). The cell lysates were then resolved by SDS-PAGE. After electrophoresis, the proteins from SDS-PAGE were transferred to nitrocellulose membrane which was then blocked with 5% non-fat dried milk for 60 minutes. The membrane was then incubated with LC3 primary antibodies (1:1000) in TBST overnight at 4'C. After that, the membrane was further incubated with HRP-conjugated secondary antibodies for 60 minutes. Finally, protein bands were visualized by using the ECL Western Blotting Detection Reagents (Invitrogen, Paisley, Scotland, UK). [0045]Results: The positive control drug, rapamycin, demonstrated a markedly increase in GFP-LC3 puncta formation. While the other active components of Radix Polygalae including polygalacic acid and senegenin showed no autophagy activity in cells, Onjisaponin B increased the formation of GFP-LC3 puncta formation in a dose dependent manner as revealed by fluorescent microscopy as shown in Fig. 3a. In addition, M006.023.UMDAU 7 Spec Draft As Filed western blot analysis of autophagic marker LC3 also demonstrated the increase of LC3-II conversion upon Onjisaponin B treatment as shown in Fig. 3b. Furthermore, Onjisaponin B increased the rate of LC3-II formation in the presence of protease inhibitors as shown in Fig. 3c, suggesting that Onjisaponin B induces autophagic flux, rather than promotes the blockage of fusion between autophagosome and lysosome. [0046] Conclusion: These data proved that Onjisaponin B has the autophagic effect and suggested that Onjisaponin B is a novel autophagy enhancer in Radix Polygalae. [0047] Example 3 [0048]This example describes an in vitro study to demonstrate the autophagic effect of Onjisaponin B is dependent on the presence of autophagy-related gene 7 (Atg7). [0049] Quantification of autophagy GFP-LC3 Puncta in Atg7 wild type and deficient MEFs. GFP-LC3 puncta formation was quantified as described previously [2]. In brief, both Atg7 wild-type and deficient mouse embryonic fibroblasts (MEFs) grown on coverslips in a 6-well plate were treated with indicated concentrations of Onjisaponin B. The cells were then fixed in 4% paraformaldehyde for 20 minutes at room temperature and then rinsed with PBS. Slides were mounted with FluorSaveTM mounting media (Calbiochem, San Diego, California) and examined by fluorescence microscopy. The number of GFP-positive cells with GFP-LC3 puncta formation was examined under the Nikon ECLIPSE 80i microscope. Representative images were captured with CCD digital camera Spot RT3TM (Diagnostic Instruments, Inc., Melville, NY). To quantify for autophagy, the percentage of cells with punctate GFP-LC3 fluorescence was calculated by counting the number of the cells with punctate GFP-LC3 fluorescence in GFP-positive cells. A minimum of 150 cells from 3 randomly selected fields was scored. [0050]Results: Onjisaponin B was found to induce GFP-LC3 puncta formation in wild type Atg7 cells but not in Atg7-knockout mouse embryonic fibroblasts as shown in Fig.4. [0051] Conclusion: The results show that Onjisaponin B works as a novel autophagy enhancer which depends on autophagy related gene, Atg7, for the induction of autophagy. M006.023.UMDAU 8 Spec Draft As Filed [0052] Example 4 [0053] This example describes an in vitro study to demonstrate the molecular mechanism of Onjisaponin B in autophagy induction. [0054]Detection of mTOR signaling marker proteins. PC12 cells treated with indicated time and concentrations of Onjisaponin B were harvested and lysed in RIPA buffer (Cell Signaling). The cell lysates were then resolved by SDS-PAGE. After electrophoresis, the proteins from SDS-PAGE were transferred to nitrocellulose membrane which was then blocked with 5% non-fat dried milk for 60 minutes. The membrane was then incubated with P-p70S6K, p70S6K, P-AMPK, AMPK primary antibodies (1:1000) in TBST overnight at 4'C respectively. After that, the membrane was further incubated with HRP-conjugated secondary antibodies for 60 minutes. Finally, protein bands were visualized by using the ECL Western Blotting Detection Reagents (Invitrogen). [0055] Quantification of Onjisaponin B-mediated autophagy in the presence of specific inhibitors. GFP-LC3 puncta formation was quantified as described previously [2]. In brief, PC12 cells expressing GFP-LC3 were treated with Onjisaponin B (Onji, 25 gM) in the presence of AMPK inhibitor (compound C) (CC, 5 M) or 3-Methyladenine (3-MA, 5 mM) for 12 hours. The cells were then fixed in 4% paraformaldehyde for 20 minutes at room temperature and then rinsed with PBS. Slides were mounted with FluorSaveTM mounting media (Calbiochem) and examined by fluorescence microscopy. To quantify for autophagy, the percentage of cells with punctate GFP-LC3 fluorescence was calculated by counting the number of the cells with punctate GFP-LC3 fluorescence in GFP-positive cells. A minimum of 150 cells from 3 randomly selected fields was scored. [0056]Results: Onjisaponin B was found to activate the phosphorylation of AMPK in a time dependent manner (as observed from columns 2-4 of Fig. 5a that show the results of the study in which protein bands of 25ptM Onjisaponin B were visualized at the 8 th, 1 6 h and 24h hour) and dose dependent manner (as observed from columns 5-8 of Fig. 5a that show the results of the study in which protein bands of Onjisaponin B at concentrations M006.023.UMDAU 9 Spec Draft As Filed of 3piM, 6.25 ptM, 12.5piM and 25piM were visualized at the 24h hour), and this activation was also accompanied by a concomitant reduction in its downstream p70S6K phosphorylation. In addition, there was a significant reduction in Onjisaponin B-induced GFP-LC3 puncta formation in PC12 cells treated with the presence of AMPK inhibitor (compound C) or the autophagy inhibitor (3-Methyladenine,3-MA), which is a specific inhibitor of the class III P13K responsible for autophagy induction as shown in Fig. 5b. [0057] Conclusion: The results show that Onjisaponin B activates autophagy through an AMPK-mTOR signaling pathway. [0058] Example 5 [0059] This example describes an in vitro study to demonstrate the clearance of mutant huntingtin and mutant A53T a-synuclein by Onjisaponin B. [0060]Removal of mutant huntingtin and mutant a-synuclein. PC 12 cells were transfected transiently with EGFP-HDQ74 plasmids for 24 hours using Lipofectamine Plus LTX reagent (Invitrogen) according to the manufacturer's protocol. The transfected cells were then treated with Onjisaponin B for 24 hours. The removal of mutant huntingtin, EGFP-HDQ74, was then quantitated by immunoblotting with antibody against EGFP. To measure the clearance of a-synuclein in cellular model, the overexpression of mutant A53T a-synuclein was first induced by the addition of doxycycline (1 gg/ml ) (Sigma) for 24 hours [11], by using the doxycycline inducible PC 12 cell line transfected with A53T a-synuclein plasmid. The expression of mutant A53T a-synuclein was then switched off by removing doxycycline from medium. Cells were then incubated with Onjisaponin B for a further 24 hours. The clearance of mutant a synuclein was then measured by immunoblotting with antibody against myc tag. [0061]Results: Both ethanol extract of Radix Polygalae and Onjisaponin B enhanced the clearance of overexpressed EGFP-tagged mutant huntingtin with 74 CAG repeats as measured by immunoblotting against EGFP antibody as shown in Fig. 6a. On the other hand, the expression of A53T a-synuclein can be switched on by adding the chemical agent, doxycycline. Upon the induction by doxycycline, whether specific compounds M006.023.UMDAU 10 Spec Draft As Filed enhance the clearance of mutant proteins [11, 29, 30] can be evaluated. As shown in Fig. 6b, both Radix Polygalae ethanol extract and Onjisaponin B accelerate the clearance of myc-tagged mutant A53T a-synuclein, whereas cells without Onjisaponin B or ethanol extract incubation showed no removal of mutant protein after doxycycline induction. The fold change in both Figs. 6a and 6b indicated the fold change of EGFP-HDQ 74 level. [0062] Conclusion: The results show that Onjisaponin B works as a useful neuroprotective agent through accelerating the clearance of mutant huntingtin and a synuclein in vitro. [0063] Example 6 [0064] This example describes an in vitro study to demonstrate that the clearance of mutant huntingtin by Onj isaponin B requires autophagy induction. [0065] Quantification of mutant huntingtin aggregates in PC12, Atg7 wild type and deficient MEFs. In brief, both EGFP-HDQ74 transfected PC12, Atg7 wild-type and deficient mouse embryonic fibroblasts (MEFs) grown on coverslips in a 6-well plate were treated with indicated concentrations of Onjisaponin B. The cells were then fixed in 4% paraformaldehyde for 20 minutes at room temperature and then rinsed with PBS. Slides were mounted with FluorSaveTM mounting media (Calbiochem, San Diego, California) and examined by fluorescence microscopy. The number of cells with GFP-aggregates formation was examined under the Nikon ECLIPSE 80i microscope. Representative images were captured with CCD digital camera Spot RT3TM (Diagnostic Instruments, Inc., Melville, NY). To quantify the clearance of mutant huntingtin, the percentage of cells with GFP-aggregates was calculated by counting the number of the cells with punctate GFP-LC3 fluorescence in GFP-positive cells. A minimum of 150 cells from 3 randomly selected fields was scored. [0066] Detection of autophagic marker protein LC3 conversion and HTT 74 mutant aggregates. After drug treatments for Atg7 wild type and deficient MEFs, cells were harvested and lysed in RIPA buffer (Cell Signaling Technologies Inc., Beverly, MA). The cell lysates were then resolved by SDS-PAGE. After electrophoresis, the proteins M006.023.UMDAU 11 Spec Draft As Filed from SDS-PAGE were transferred to nitrocellulose membrane which were then blocked with 5% non-fat dried milk for 60 minutes. The membrane was then incubated with LC3 primary antibodies (1:1000) in TBST overnight at 4'C. After that, the membrane was further incubated with HRP-conjugated secondary antibodies for 60 minutes. Finally, protein bands were visualized by using the ECL Western Blotting Detection Reagents (Invitrogen, Paisley, Scotland, UK). [0067] Results: Immunocytochemistry analysis further confirmed that Onjisaponin B enhanced the clearance of inclusions formed by EGFP-HDQ74 as shown in Fig. 7a. To further confirm that the protective effect of Onjisaponin B was due to an Atg7 dependent autophagic effect, wild type Atg7 and Atg7-knockout mouse embryonic fibroblasts were transfected with EGFP-HDQ74 for fluorescent inclusions formation. Results showed that Onjisaponin B enhanced the clearance of EGFP-HDQ74 inclusions as illustrated in Fig. 7b; further, as shown in Fig. 7c, the rate of LC3II formation in wild type Atg7 cells but not in Atg7-knockout cells suggested the compound-mediated neuroprotective effect was autophagy dependent. [0068] Conclusion: The enhanced clearance of mutant huntingtin by Onjisaponin B requires the induction of autophagy in cells. [0069] Example 7 [0070] This example describes an in vitro study to demonstrate the reduced oligomerization of a-synuclein mediated by Onjisaponin B. [0071]Bimolecular fluorescence complementation (BiFC) assay. In brief, HeLa cells transfected with both GFP-N terminal- a-synuclein (GNS) and a-synuclein-GFP-C terminal (SGC) plasmids were incubated at 37C for 4 hours. Then, the transfected cells were incubated with different concentrations of Radix Polygalae ethanol extract or Onjisaponin B for a further 24 hours at 30 'C [31]. Fluorescent signals upon complete GFP fluorophore reconstitution in cells were then detected by flow analysis. [0072]Results: By using the bimolecular fluorescence complementation (BiFC) assay, the oligomerization of a-synuclein in living cells [31] can be directly quantified. In brief, M006.023.UMDAU 12 Spec Draft As Filed a-synuclein proteins fused with two different non-fluorescent GFP terminal fragments, (i) GFP-N terminal-a-synuclein (GNS) or (ii) a-synuclein-GFP-C terminal (SGC) respectively were used [31, 32]. Upon oligomerization of a-synuclein, the two non fluorescent fractions of GFP will therefore reconstitute the complete GFP fluorophore and transmit GFP fluorescent signal within cells. This signal in turn can be quantified by flow cytometry analysis. As measured by flow analysis as shown in Fig. 8, both Radix Polygalae crude extract and Onjisaponin B inhibit the oligomerization of a-synuclein in HeLa cells transfected with GNS and SGC. [0073] Conclusion: From the result, the decrease of GFP fluorescent signal suggested that both Radix Polygalae ethanol extract and its active component Onjisaponin B may play a protective role in Parkinson's disease through inhibiting a-synuclein oligomerization, which is a crucial step for a-synuclein aggregation. Taken together, these novel findings provide evidence to further support the neuroprotective function of Radix Polygalae ethanol extract and Onj isaponin B in cellular model. [0074] Example 8 [0075] This example describes an in vitro study to demonstrate that Onjisaponin B reduces toxicity in PC-12 cells expressing either mutant huntingtin or mutant A53T u synuclein. [0076] Cell cytotoxicity assay: Cytotoxicity was assessed using the 3-(4,5 dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay as described previously [28]. PC-12 cells were transfected with or without mutant huntingtin EGFP-HDQ74 for 24 h. The PC-12 cells were then seeded on 96-well plates with 4000 cells per well and then exposed to the indicated concentrations of Onjisaponin B (25 ptmol/L) for 2 days. Subsequently, 10 pL of MTT reagents was added to each well and incubated at 37 0 C for 4 hours, followed by the addition of 100 pL solubilization buffer (10% SDS in 0.01 mol/L HCl) and overnight incubation. Absorbance at 585 nm was determined from each well on the next day. The percentage of cell viability was calculated using the following formula: Cell viability (%) = Cells number treated / Cells number DMSO control x 100. Data was obtained from three independent experiments. M006.023.UMDAU 13 Spec Draft As Filed [0077] Flow cytometry analysis: Doxycycline-inducible PC-12 cell lines transfected with mutant A53T u-synuclein were induced with doxycycline (1 pig/mL) for 24 h and the expression of transgene was then switched off by the removal of doxycycline. Cells were then treated with Onjisaponin B (50 pM) for a further 24 h after doxycycline removal. The control group (Ctrl) contained cells without the addition of doxycycline, while the Dox group contained cells with the removal of doxycycline (1 pig/mL) after 24 h of induction but without treatment of Onjisaponin B. Cell death was then assessed by flow cytometry using Annexin V staining kit and Propidium iodide staining. Data from the bar chart represented the means ± S.D. of three independent experiments. ** p < 0.01; * p < 0.05. Flow cytometry was carried out using a FACSCalibur flow cytometer. Data acquisition and analysis were performed with CellQuest. [0078] Results: With the result that Onjisaponin B treatment enhanced the clearance of mutant aggregate-prone proteins, Onjisaponin B was studied to see if it plays a protective role in reducing cell death induced by mutant huntingtin or mutant A53T u-synuclein expression. To study the toxicity of mutant huntingtin or mutant A53T u-synuclein in cells, PC-12 cells transfected transiently with EGFP-HDQ 74 or myc-tagged mutant A53T u-synuclein were used to examine the effect of Onjisaponin B on cell viability. As shown in Figures 9a and 9b, while transient expression of mutant huntingtin or mutant A53T u-synuclein lead to a decrease in cell viability, the addition of Onjisaponin B reduced toxicity in PC-12 cells expressing either mutant huntingtin (as shown in Fig. 9a) or mutant A53T u-synuclein (as shown in Fig. 9b), respectively. [0079] Conclusion: In consistent with the results from the above Examples that Onjisaponin B increases the clearance of mutant huntingtin and mutant A53T u-synuclein and reduces oligomerization of u-synuclein, the result of this Example further demonstrated the potential therapeutic role of onjisaponin B working as a neuroprotective agent, through lowering of mutant huntingtin and a-synuclein toxicity in cells. [0080] Brief summary of studies described in Examples 1 to 8 [0081] In summary, throughout these studies, Onjisaponin B is proven to show no in vitro toxicity and possess autophagic effect, which is further demonstrated to be Atg7 M006.023.UMDAU 14 Spec Draft As Filed dependent. Also, the activation of autophagy by Onjisaponin B is demonstrated to be through an AMPK-mTOR signaling pathway. Onjisaponin B is further shown to be a useful neuroprotective agent through accelerating the clearance of mutant huntingtin and a-synuclein in vitro, in which such clearance requires the induction of autophagy in cells. In addition, Onjisaponin B is proven to inhibit the oligomerization of a-synuclein and reduce toxicity in PC-12 cells expressing either mutant huntingtin or mutant A53T a synuclein. [0082] Taken together, these findings provide evidence to further support the neuroprotective function of Onjisaponin B and its medical application in treating neurodegenerative disease. [0083] The exemplary embodiments of the present invention are thus fully described. Although the description referred to particular embodiments, it will be clear to one skilled in the art that the present invention may be practiced with variation of these specific details. Hence this invention should not be construed as limited to the embodiments set forth herein. [0084] References: 1. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008 Jan 11; 132 (1): 27-42. 2. Law BY, Wang M, Ma DL, Al-Mousa F, Michelangeli F, Cheng SH, et al. Alisol B, a novel inhibitor of the sarcoplasmic/endoplasmic reticulum Ca(2+) ATPase pump, induces autophagy, endoplasmic reticulum stress, and apoptosis. Mol Cancer Ther Mar; 9 (3): 718-730. 3.Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 2006 Oct 19; 443 (7113): 780-786. 4.Rubinsztein DC, DiFiglia M, Heintz N, Nixon RA, Qin ZH, Ravikumar B, et al. Autophagy and its possible roles in nervous system diseases, damage and repair. Autophagy 2005 Apr; 1 (1): 11-22. 5.Rubinsztein DC, Gestwicki JE, Murphy LO, Klionsky DJ. Potential therapeutic applications of autophagy. Nat Rev Drug Discov 2007 Apr; 6 (4): 304-312. M006.023.UMDAU 15 Spec Draft As Filed 6. Ravikumar B, Duden R, Rubinsztein DC. Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 2002 May 1; 11 (9): 1107-1117. 7.Ravikumar B, Vacher C, Berger Z, Davies JE, Luo S, Oroz LG, et al. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat Genet 2004 Jun; 36 (6): 585-595. 8.Webb JL, Ravikumar B, Atkins J, Skepper IN, Rubinsztein DC. Alpha-Synuclein is degraded by both autophagy and the proteasome. J Biol Chem 2003 Jul 4; 278 (27): 25009-25013. 9.Rubinsztein DC, Codogno P, Levine B. Autophagy modulation as a potential therapeutic target for diverse diseases. Nat Rev Drug Discov Sep; 11 (9): 709-730. 10.Rubinsztein DC. Lessons from animal models of Huntington's disease. Trends Genet 2002 Apr; 18 (4): 202-209. 11. Sarkar S, Davies JE, Huang Z, Tunnacliffe A, Rubinsztein DC. Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein. J Biol Chem 2007 Feb 23; 282 (8): 5641-5652. 12.Sarkar S, Floto RA, Berger Z, Imarisio S, Cordenier A, Pasco M, et al. Lithium induces autophagy by inhibiting inositol monophosphatase. J Cell Biol 2005 Sep 26; 170 (7): 1101-1111. 13.Berger Z, Ravikumar B, Menzies FM, Oroz LG, Underwood BR, Pangalos MN, et al. Rapamycin alleviates toxicity of different aggregate-prone proteins. Hum Mol Genet 2006 Feb 1; 15 (3): 433-442. 14.Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative diseases. Nat Neurosci Jul; 13 (7): 805-811. 15.Renna M, Jimenez-Sanchez M, Sarkar S, Rubinsztein DC. Chemical inducers of autophagy that enhance the clearance of mutant proteins in neurodegenerative diseases. J Biol Chem Apr 9; 285 (15): 11061-11067. 16.Jia K, Levine B. Autophagy is required for dietary restriction-mediated life span extension in C. elegans. Autophagy 2007 Nov-Dec; 3 (6): 597-599. 17.Jia K, Hart AC, Levine B. Autophagy genes protect against disease caused by polyglutamine expansion proteins in Caenorhabditis elegans. Autophagy 2007 Jan-Feb; 3 M006.023.UMDAU 16 Spec Draft As Filed (1): 21-25. 18.Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 2006 Jun 15; 441 (7095): 880-884. 19.Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 2006 Jun 15; 441 (7095): 885-889. 20.Komatsu M, Wang QJ, Holstein GR, Friedrich VL, Jr., Iwata J, Kominami E, et al. Essential role for autophagy protein Atg7 in the maintenance of axonal homeostasis and the prevention of axonal degeneration. Proc Natl Acad Sci U S A 2007 Sep 4; 104 (36): 14489-14494. 21.Yang F, Lim GP, Begum AN, Ubeda OJ, Simmons MR, Ambegaokar SS, et al. Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J Biol Chem 2005 Feb 18; 280 (7): 5892-5901. 22.Zhao Z, Fang M, Xiao D, Liu M, Fefelova N, Huang C, et al. Potential antiarrhythmic effect of methyl 3,4,5-trimethoxycinnamate, a bioactive substance from roots of polygalae radix: suppression of triggered activities in rabbit myocytes. Biol Pharm Bull; 36 (2): 238-244. 23.Zhang H, Han T, Zhang L, Yu CH, Wan DG, Rahman K, et al. Effects of tenuifolin extracted from radix polygalae on learning and memory: a behavioral and biochemical study on aged and amnesic mice. Phytomedicine 2008 Aug; 15 (8): 587-594. 24.Lin Z, Gu J, Xiu J, Mi T, Dong J, Tiwari JK. Traditional chinese medicine for senile dementia. Evid Based Complement Alternat Med; 2012: 692621. 25.Xie W, Yang Y, Gu X, Zheng Y, Sun YE, Liang Y, et al. Senegenin attenuates hepatic ischemia-reperfusion induced cognitive dysfunction by increasing hippocampal NR2B expression in rats. PLoS One; 7 (9): e45575. 26.Choi JG, Kim HG, Kim MC, Yang WM, Huh Y, Kim SY, et al. Polygalae radix inhibits toxin-induced neuronal death in the Parkinson's disease models. J Ethnopharmacol Mar 24; 134 (2): 414-421. 27.Karakida F, Ikeya Y, Tsunakawa M, Yamaguchi T, Ikarashi Y, Takeda S, et al. Cerebral protective and cognition-improving effects of sinapic acid in rodents. Biol M006.023.UMDAU 17 Spec Draft As Filed Pharm Bull 2007 Mar; 30 (3): 514-519. 28.Wong VK, Zhou H, Cheung SS, Li T, Liu L. Mechanistic study of saikosaponin-d (Ssd) on suppression of murine T lymphocyte activation. J Cell Biochem 2009 May 15; 107 (2): 303-315. 29. Sarkar S, Perlstein EO, Imarisio S, Pineau S, Cordenier A, Maglathlin RL, et al. Small molecules enhance autophagy and reduce toxicity in Huntington's disease models. Nat Chem Biol 2007 Jun; 3 (6): 331-338. 30.Talloczy Z, Virgin HWt, Levine B. PKR-dependent autophagic degradation of herpes simplex virus type 1. Autophagy 2006 Jan-Mar; 2 (1): 24-29. 31. Outeiro TF, Putcha P, Tetzlaff JE, Spoelgen R, Koker M, Carvalho F, et al. Formation of toxic oligomeric alpha-synuclein species in living cells. PLoS One 2008; 3 (4): e1867. 32.Lu JH, Ardah MT, Durairajan SS, Liu LF, Xie LX, Fong WF, et al. Baicalein inhibits formation of alpha-synuclein oligomers within living cells and prevents Abeta peptide fibrillation and oligomerisation. Chembiochem Mar 7; 12 (4): 615-624. <DocRef#001 19901-RM > M006.023.UMDAU 18 Spec Draft As Filed

Claims (11)

1. A method of preventing, treating and/or delaying the onset of neurodegenerative diseases comprising administering an effective amount of Onjisaponin B to a subject in need thereof
2. The method according to claim 1 wherein said neurodegenerative diseases are selected from a group consisting of Parkinson's disease and Huntington's disease.
3. Use of Onj isaponin B as an autophagy enhancer for treating neurodegenerative diseases.
4. The use according to claim 3 wherein said neurodegenerative diseases are selected from a group consisting of Parkinson's disease and Huntington's disease.
5. The use according to claim 3 wherein said the autophagic effect of Onjisaponin B is dependent on the presence of autophagy-related gene 7.
6. The use according to claim 3 wherein said Onjisaponin B activates autophagy through an AMPK-mTOR signaling pathway.
7. The use according to claim 3 wherein said Onj isaponin B exhibits the neuroprotective function through accelerating the clearance of mutant huntingtin and mutant A53T alpha synuclein.
8. A pharmaceutical composition comprising Onjisaponin B admixed with a pharmaceutical carrier for treating neurodegenerative diseases.
9. The pharmaceutical composition according to claim 8 wherein said neurodegenerative diseases are selected from a group consisting of Parkinson's disease and Huntington's disease.
10. A dietary supplement comprising Onjisaponin B admixed with a pharmaceutical carrier for treating neurodegenerative diseases.
11. The dietary supplement according to claim 10 wherein said neurodegenerative diseases are selected from a group consisting of Parkinson's disease and Huntington's disease. M006.023.UMDAU 19 Spec Draft As Filed
AU2014100319A 2013-08-15 2014-04-02 A novel autophagy enhancer for treatment of neurodegenerative diseases Ceased AU2014100319A4 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361866045P 2013-08-15 2013-08-15
US61/866,045 2013-08-15
US14/231,671 2014-03-31
US14/231,671 US9005677B2 (en) 2013-08-15 2014-03-31 Autophagy enhancer for treatment of neurodegenerative diseases

Publications (1)

Publication Number Publication Date
AU2014100319A4 true AU2014100319A4 (en) 2014-05-01

Family

ID=50548878

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2014100319A Ceased AU2014100319A4 (en) 2013-08-15 2014-04-02 A novel autophagy enhancer for treatment of neurodegenerative diseases

Country Status (1)

Country Link
AU (1) AU2014100319A4 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111358801A (en) * 2020-03-21 2020-07-03 复旦大学 Application of tenuifolin F in preparing medicine for preventing and treating Huntington disease

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111358801A (en) * 2020-03-21 2020-07-03 复旦大学 Application of tenuifolin F in preparing medicine for preventing and treating Huntington disease

Similar Documents

Publication Publication Date Title
Wang et al. Protective effects of hesperidin against amyloid-β (Aβ) induced neurotoxicity through the voltage dependent anion channel 1 (VDAC1)-mediated mitochondrial apoptotic pathway in PC12 cells
Xu et al. Curcumin analogues attenuate Aβ25-35-induced oxidative stress in PC12 cells via Keap1/Nrf2/HO-1 signaling pathways
Wang et al. Echinacoside protects against 6‐hydroxydopamine‐induced mitochondrial dysfunction and inflammatory responses in PC12 cells via reducing ROS production
Yan et al. Overexpression of human E46K mutant α-synuclein impairs macroautophagy via inactivation of JNK1-Bcl-2 pathway
US9561219B2 (en) Group of alkaloids, the novel autophagic enhancers for treatment of cancers and neurodegenerative conditions thereof
Meng et al. Anthocyanins extracted from Aronia melanocarpa protect SH-SY5Y cells against amyloid-beta (1–42)-induced apoptosis by regulating Ca2+ homeostasis and inhibiting mitochondrial dysfunction
Lee et al. Novel synthetic chalcone‐coumarin hybrid for Aβ aggregation reduction, antioxidation, and neuroprotection
Hahm et al. Autophagy fails to alter withaferin A-mediated lethality in human breast cancer cells
Wong et al. Bioassay-guided isolation of neuroprotective compounds from Loranthus parasiticus against H2O2-induced oxidative damage in NG108-15 cells
Qi et al. Trans-cinnamaldehyde protected PC12 cells against oxygen and glucose deprivation/reperfusion (OGD/R)-induced injury via anti-apoptosis and anti-oxidative stress
Song et al. Schizandrol A protects against Aβ 1–42-induced autophagy via activation of PI3K/AKT/mTOR pathway in SH-SY5Y cells and primary hippocampal neurons
Henrı́quez et al. Neuroprotective effect of brazilin on amyloid β (25–35)-induced pathology in a human neuroblastoma model
Yao et al. Pseudoginsenoside‐F11 alleviates oligomeric β‐amyloid‐induced endosome‐lysosome defects in microglia
US9005677B2 (en) Autophagy enhancer for treatment of neurodegenerative diseases
Zhong et al. Protective effect of ethyl vanillin against Aβ‑induced neurotoxicity in PC12 cells via the reduction of oxidative stress and apoptosis
Eboji et al. Burkea africana Hook (Caesalpiniaceae) ethanolic extract causes cell cycle arrest at M phase and induces caspase dependent apoptosis
Luo et al. The protective effects of mogroside V against neuronal damages by attenuating mitochondrial dysfunction via upregulating Sirtuin3
Du et al. Catalpol ameliorates neurotoxicity in N2a/APP695swe cells and APP/PS1 transgenic mice
Cheung et al. Chronic exposure to U18666A induces apoptosis in cultured murine cortical neurons
He et al. Folium Hibisci Mutabilis extract, a potent autophagy enhancer, exhibits neuroprotective properties in multiple models of neurodegenerative diseases
Zhou et al. Dopamine-related and caspase-independent apoptosis in dopaminergic neurons induced by overexpression of human wild type or mutant α-synuclein
AU2014100319A4 (en) A novel autophagy enhancer for treatment of neurodegenerative diseases
Ravi et al. Cassia tora extract alleviates Aβ1–42 aggregation processes in vitro and protects against aluminium-induced neurodegeneration in rats
Bhattacharyya et al. Black tea induces tumor cell apoptosis by Bax translocation, loss in mitochondrial transmembrane potential, cytochrome c release and caspase activation
Huang et al. Rosavin thwarts amyloid-β-induced macromolecular damages and neurotoxicity, exhibiting anti-Alzheimer’s disease activity in Wister rat model

Legal Events

Date Code Title Description
FGI Letters patent sealed or granted (innovation patent)
MK22 Patent ceased section 143a(d), or expired - non payment of renewal fee or expiry