Skip to main content
Nanomedicine logoLink to Nanomedicine
. 2021 May 7;16(13):1081–1095. doi: 10.2217/nnm-2020-0477

A novel gene therapy for neurodegenerative Lafora disease via EPM2A-loaded DLinDMA lipoplexes

Hari Priya Vemana 1, Aishwarya Saraswat 1, Shraddha Bhutkar 1, Ketan Patel 1, Vikas V Dukhande 1,*
PMCID: PMC8162161  PMID: 33960213

Abstract

Aim:

To develop novel cationic liposomes as a nonviral gene delivery vector for the treatment of rare diseases, such as Lafora disease – a neurodegenerative epilepsy.

Materials & methods:

DLinDMA and DOTAP liposomes were formulated and characterized for the delivery of gene encoding laforin and expression of functional protein in HEK293 and neuroblastoma cells.

Results:

Liposomes with cationic lipids DLinDMA and DOTAP showed good physicochemical characteristics. Nanosized DLinDMA liposomes demonstrated desired transfection efficiency, negligible hemolysis and minimal cytotoxicity. Western blotting confirmed successful expression and glucan phosphatase assay demonstrated the biological activity of laforin.

Conclusion:

Our study is a novel preclinical effort in formulating cationic lipoplexes containing plasmid DNA for the therapy of rare genetic diseases such as Lafora disease.

Keywords: : DLinDMA liposomes, gene delivery, gene therapy, Lafora disease, laforin, neurodegenerative disorders, rare diseases

Graphical abstract

graphic file with name nnm-16-1081-GA.jpg


Lafora disease (LD) is a progressive myoclonic epilepsy; a rare genetic pediatric disorder of autosomal recessive inheritance that is characterized by progressive neurodegeneration, epileptic seizures, dementia and ultimately death [1]. LD results from mutations in either EPM2A gene encoding laforin or EPM2B/NHLRC1 gene encoding malin [2–4]. Laforin is a dual-specificity phosphatase, whereas malin is an E3 ubiquitin ligase [5,6]. Laforin is the only known phosphatase with a carbohydrate-binding module and was shown to act as a glycogen phosphatase [5,7,8]. Laforin binds to malin and thereby can also act as a scaffold protein [9,10]. The precise role of the LD proteins is not known but previous studies have pointed to their roles in regulating glycogen phosphorylation, glycogen structure and solubility, cellular autophagy and the dysfunction of these processes leads to insoluble glycogen structures in various organs including brain termed as Lafora bodies (LBs) [11–15].

LD is fatal and only symptomatic, palliative treatment is currently available [16]. Given the diversity of seizure types in LD, wide spectrum of antiepileptic drugs (AEDs) are prescribed such as valproic acid to manage the seizures [17,18]. Recently, perampanel, a new AMPA receptor antagonist and metformin, an activator of AMPK were shown to be somewhat beneficial for the treatment of LD [19,20]. In addition, vagal nerve stimulation serves as an adjunctive treatment option to treat LD [21,22]. Additionally, promising studies for LD with drugs such as antisense oligonucleotides (ASOs) and antibody–enzyme fusion are being developed. Research studies indicate that ASOs are an excellent therapy platform for neurodegenerative diseases [23]. ASO therapy reducing brain glycogen synthase (GYS) is currently being developed for LD [24]. Another drug which is being studied is antibody–enzyme fusion therapy called VAL-0417, which involves degradation of LBs by α-amylase [25,26]. Novel therapeutic strategies involving gene therapy have considerably advanced the treatments for genetic diseases and could be a promising therapeutic strategy to treat LD [27,28]. However, main challenge that must be overcome is the transfer of genetic material like the plasmid DNA to the nucleus of target cells considering the number of intra- and extra-cellular barriers that the vector/DNA complexes encounter [29,30]. Therefore, it is essential to design a safe and stable transport carrier for the effective targeted delivery of the plasmid DNA. Although viral vectors, including retroviruses, adenoviruses and lentiviruses, are more efficient than nonviral vectors, they may lead to severe off-target immunogenicity, inflammatory response and toxicity [31]. On the other hand, nonviral vectors such as cationic lipids or polymers are able to efficiently deliver nucleic acids and are easier to scale up and also much safer than the viral vectors in terms of generating immune responses and off-target toxicity [32,33].

Liposomes are generally formed by the self-assembly of dissolved phospholipid molecules composed of hydrophilic head groups attached to hydrophobic tails by a linker [34]. Cationic liposomes have numerous advantages as a nonviral delivery system for nucleic acids like plasmid DNA [34–36]. They have excellent biocompatibility following in vivo administration. The surface positive charge of cationic lipids allows a strong electrostatic interaction with the negatively charged nucleic acids to form lipoplexes. An endogenous lipid such as cholesterol is usually inserted between the lipid bilayers to increase the rigidity of the liposomal system; whereas a general approach to increase the in vivo stability of these carriers involves the insertion of polyethylene glycol (PEG)-conjugated neutral lipids (DSPE-PEG2000) for PEGylation of nanoparticles [35]. Neutral lipids like dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and dioleoyl-sn-glycero-3-phosphocholine (DOPC) help in transfection and endosomal escape due to their fusogenic property [37]. Thus, cationic liposomes could be a promising nanocarrier to deliver therapeutic plasmid DNA for the treatment of LD.

To our knowledge, there have been no previous studies that investigated the application of cationic liposomes as a gene delivery carrier for the gene therapy of LD. Therefore, aim of this research is to develop cationic lipid-mediated delivery of laforin-encoding plasmid DNA and test its preclinical safety and efficacy as a novel therapeutic approach for the treatment of LD.

Materials & methods

Plasmids & cloning

Plasmids pEGFP-N3, pcDNA3.1 N-terminal FLAG tagged (pCNF) laforin and catalytically inactive laforin-encoding pCNF laforin C266S were generous gifts from Dr. Matthew Gentry (University of Kentucky). Plasmids were amplified in Escherichia coli (DH5α strain) and purified using PureLink HiPure Plasmid Maxiprep kit (Invitrogen), according to the manufacturer’s protocol. The concentration and purity of DNA were determined using NanoDrop Onec (Thermo Fisher Scientific).

Cell culture

Human embryonic kidney HEK293 cells were grown in DMEM with high glucose 4.5 gm/l (Corning Cellgro), 5% v/v fetal bovine serum (FBS; Atlanta Biologicals) and 100 μg/ml penicillin/streptomycin/amphotericin B (MP Biomedicals). Human neuroblastoma SK-N-SH cells were cultured in DMEM containing glucose 1 gm/l (Corning Cellgro) and supplemented with 10% v/v FBS and 100 μg/ml penicillin/streptomycin/amphotericin B. All transfection experiments with liposomes were carried out 16 h after subculturing cells to about 70% confluency.

Preparation of DLinDMA liposomes

Cationic liposomes containing DLinDMA (MedChem Express) or DOTAP (Avanti Polar Lipids) at 3 mg/ml were prepared by the ethanol injection method followed by ultrasonication to produce small unilamellar vesicles. As mentioned in Table 1, the required amounts of neutral lipids (DOPE and DOPC), cholesterol and cationic lipid (DLinDMA or DOTAP) were dissolved in ethanol. The resulting organic phase was injected into the aqueous phase (Milli-Q water) to spontaneously form the liposomes. Liposomal suspension was then probe sonicated at 30 amplitudes for 120 s to obtain nanoliposomes. The liposomes were stirred at room temperature to remove the traces of ethanol.

Table 1. . Lipid composition of liposomal formulations.

Lipid Concentration (mM)
DLinDMA/DOTAP 5
DOPC 10
DOPE 2
Cholesterol 2

Preparation of liposome–DNA complex

The DNA/liposome complexes were prepared by mixing the plasmid DNA with different ratios of the liposomal formulation to identify the DNA-binding affinity of the cationic liposomes. Briefly, different volumes of the formulation were mixed with 1 μg of DNA and incubated for 30 min at room temperature (RT) to form the DNA/liposome complex.

Physicochemical characterization of cationic liposomes

Nanocarriers were diluted with Milli-Q water. Average particle size, size distribution and zeta potential analysis were performed by dynamic light scattering particle size analyzer (Malvern Zetasizer Nano ZS). Samples were analyzed using folded capillary cells at 25°C with a scattering angle of 173°.

Cell viability determination

Cytotoxicity of liposomes was measured in HEK293 cells and SK-N-SH cells by MTT cell viability assay. The cells were seeded in 96-well plates at density of 5000 cells per well. After 16 h incubation, cells were treated with DOTAP or DLinDMA liposomes at concentrations ranging from 1.56 to 200 μM. Transit LT1 (Mirus Bio) and PEI max were used as positive controls. After 48 h treatment, MTT dye (0.05% w/v final concentration) was added and cells were incubated at 37°C for 3 h. Formazan crystals formed from the reduction of MTT dye were dissolved in DMSO and the absorbance was measured at 570 nm using the SpectraMax M5e plate reader (Molecular Devices).

Cell proliferation assay

Cell proliferation was determined using the CyQuant Direct Proliferation Assay kit (Thermo Fisher Scientific) as per the manufacturer’s protocol. Briefly, at the end of DLinDMA liposome treatments (ranging from 1.56 to 200 μM) of HEK293 cells, medium was carefully aspirated from each well without disturbing the cells. 50 μl of 2X detection reagent was added to each well containing 50 μl media with cells and incubated for 30 min at 37°C. Fluorescence was measured for each well at wavelengths 480/535 nm with a Spark 10 M plate reader (Tecan Life Sciences).

Transfection efficiency

The transfection efficiency of DLinDMA liposomes was evaluated in HEK 293 and SK-N-SH cells. The cells were seeded in 96-well plates at density of 5000 cells per well and maintained in complete culture medium overnight prior to transfection. Lipoplexes were formed at 1:5, 1:10, 1:20, 1:40 and 1:60 mass ratio of pEGFP-N3 DNA:DLinDMA/DOTAP and incubated for 30 min. Cells were incubated with these lipoplexes at 37°C in 5% CO2 for 3 h in opti-MEM medium. The transfection medium was replaced with fresh complete culture medium and the cells were incubated for 48 h post-transfection. Commercial transfection reagents Transit LT1 and PEI max were used as positive controls at 1:3 ratio of pEGFP-N3 DNA:Transit LT1/PEI Max (3 μl reagent per 1 μg DNA). The transfection efficiency of pEGFP-N3 was monitored by fluorescence microscopy using EVOS microscope. Fluorescence images of transfected cells were taken after 48 h at 200X magnification.

Electrophoretic mobility shift assay

DLinDMA liposomes are capable of forming electrostatic complexes with DNA and this complex formation was measured by electrophoretic mobility shift assay (EMSA). The DLinDMA:DNA complexes at different charge ratios ranging from 1:5, 1:10, 1:20, 1:40, 1:60 mass ratio of DNA:DLinDMA were incubated at room temperature for 30 min and then analyzed by electrophoresis on a 1% (w/v) agarose gel in tris-acetate-EDTA (TAE) buffer containing SYBR green dye (Life Technologies). EDTA-free loading dye was used in order to curb the interaction of EDTA with the cationic lipids used in the liposomes. The gel was run for 1 h at 100 V and visualized by ultraviolet illumination using Azure biosystems c500 imager and electrophoretic mobility was investigated to evaluate the quantity of DNA entrapment. Naked plasmid DNA was used as control.

Fluorescence microscopy

SK-N-SH cells were grown on poly-D-lysine-coated glass coverslips in 24-well cell culture plates. Next day, cells were treated with DLinDMA lipoplexes containing pEGFP-N3 or FLAG-laforin. After 48 h of transfection, cells were washed with phosphate-buffered saline (PBS) and fixed using formalin (3.7% v/v in PBS). Cells were permeabilized using cold Triton X-100 (0.25% v/v in PBS) for 5 min, washed with PBS and incubated in FBS solution (10% v/v in PBS) for 1 h. Next, samples were incubated with anti-FLAG M2 antibody (Sigma-Aldrich) for 2 h followed by washes and incubation with Alexa Fluor 594-conjugated secondary antibody. Coverslips were mounted on slides using DAPI Fluoromount, and images were taken using EVOS FL Auto Imaging System.

Immunoprecipitation

SK-N-SH cells were transfected with FLAG laforin or FLAG laforin C266S using DLinDMA liposomes in 100-mm cell culture dishes. Cells were lysed using modified RIPA buffer (Tris pH 8.0 50 mM, NaCl 150 mM, NP40 1%, glycerol 10%, NaF 10 mM and EDTA 0.4 mM) with protease inhibitors. Lysates were incubated with M2 FLAG-agarose beads (Millipore Sigma) for 2 h at 4°C. The samples with the beads were washed with modified RIPA buffer three-times and resuspended in 100 μl of modified RIPA buffer. 5 μl of sample was used for each replicate of the phosphatase assay (n = 5). 2X SDS PAGE dye was added to the remaining sample and heated for 10 min. Heated samples were centrifuged at 4°C at 1100 g for 3 min and the supernatant was collected. Samples were analyzed by western blotting with 1:1000 laforin antibody.

Western blot analysis

SK-N-SH cells were lysed after transfection for 48 h using DLinDMA liposomes. The cell lysates were centrifuged at 4°C for 10 min at 10,000 g and the supernatant was collected. 4X SDS PAGE dye was added to the samples and were heated at 95°C for 12 min. The samples were loaded onto a 4–20% SDS-polyacrylamide gel and then transferred onto a polyvinylidene difluoride membrane. The blots were blocked with 5% milk in Tris-buffered saline (TBS) containing 0.1% Tween-20 for 1 h at RT and then incubated at 4°C overnight with primary antibodies for anti-FLAG M2 (Sigma-Aldrich) and β-actin (Proteintech). The membranes were washed three-times with TBST and incubated with secondary antibody (Bio-Rad) for 1 h at RT. The proteins were visualized by chemiluminescent substrate (Thermo Fisher Scientific) on Azure biosystems c500 imager.

Glucan phosphatase assay

Molybdate-based malachite green assay is widely used for quantifying picomolar quantities of inorganic-free phosphate and was employed to assess laforin’s glucan phosphtase activity [38,39]. Malachite green reagent was made by mixing 1 volume of 4.2% (w/v) ammonium molybdate tetrahydrate in 4 M HCl and three volumes of 0.045% (w/v) malachite green carbinol hydrochloride and filtered through 0.45-μm filter. Tween-20 was added at last to the required amount of malachite green reagent to make the final concentration of Tween-20 to 0.01% v/v. A mastermix of the assay components consisting of 5X buffer (0.5 M sodium acetate, 0.25 M Bis-Tris, 0.25 M Tris-HCl, pH 5), 100 mM dithiothreitol, 5 mg/ml amylopectin was prepared and immunoprecipitated (IP) laforin sample was added to this aliquot of mastermix in a microcentrifuge tube to initiate the reaction. All reactions were incubated for 30 min at 37°C. The enzymatic activity of laforin was stopped by adding 0.1 M NEM to the reaction mixture and the tube was vortexed for 10 s. Malachite green reagent with 0.01% Tween-20 was added to each of the reaction tubes and vortexed for 10 s. All reactions were incubated for 90 min at room temperature before measuring the absorbance at 620 nm using a spectrophotometer.

In vitro hemolysis study

Experimental protocol was approved by the St. John’s University Institutional Animal Care and Use Committee for collection of blood from mice for laboratory use. Red blood cells (RBCs) from mice were used to carry out the in vitro hemolysis study of cationic liposomes containing DLinDMA. C57BL/6 mice (5–6 weeks old) were received from Jackson laboratories (CT, USA). Briefly, mice were anesthetized by 2.5% isoflurane followed by a one-time blood collection using cardiac puncture technique. Then, the animals were immediately euthanized by carbon dioxide. Initially, the collected blood was centrifuged at 450 g for 10 min to separate the RBCs from plasma. The cell pellet was washed with PBS and redispersed into an appropriate volume of PBS to achieve the same hematocrit. Then, cationic liposomes were added to the RBC dispersion to achieve various cationic lipid (DLinDMA) concentrations. Following this, all the samples were incubated for 30 min at 37°C and then centrifuged at 450 g for 10 min. The supernatant was diluted with PBS and analyzed for hemoglobin release using a UV spectrophotometer at 550 nm. PBS was used as the negative control and 5% sodium lauryl sulfate (SLS) solution was used as the positive control (100% hemoglobin release). Percentage hemolysis was calculated by following formula:

% Hemolysis=(absorbance of test sampleabsorbance of negative control)(absorbance of positive controlabsorbance of negative control)×100%

Statistical analyses

The data presented are a mean of three independent experiments ± SEM unless noted otherwise. The statistical significance of difference was assessed with one-way analysis of variance, followed by Dunnett’s or Tukey’s post hoc analysis using GraphPad Prism to determine statistical significance as * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001.

Results

Development & characterization of cationic liposomes for gene delivery

Liposomes containing cationic lipids DLinDMA or DOTAP were prepared by the ethanol injection method. The average size, size distribution, and zeta potential of both the formulations are shown in Table 2. Overall, particle size of both the liposomal formulations was below 200 nm, indicating the formation of nanosized liposomes with a narrow size distribution and positive zeta potential to facilitate the complexation with negatively charged plasmid DNA via electrostatic interaction. The dynamic light scattering illustrated unimodal particle size distribution and positive zeta potential of liposomes containing DLinDMA as the cationic lipid (Figure 1).

Table 2. . Physicochemical properties of liposomal formulations.

Liposomal formulation Particle size (nm) Polydispersity index Zeta potential (mV)
DLinDMA liposomes 141.20 ± 10.45 0.121 ± 0.022 +33.50 ± 2.25
DOTAP liposomes 110.36 ± 7.83 0.117 ± 0.034 +32.10 ± 1.98

Figure 1. . Physicochemical characterization of DLinDMA-containing liposomal formulation.

Figure 1. 

(A) Electrostatic interaction between highly positive charge of cationic liposomes and negatively charged plasmid DNA to form lipoplexes. (B) Dynamic light scattering graphs illustrating unimodal particle size distribution and positive zeta potential of the liposomes.

Safety of cationic liposomal formulations

Cell viability (MTT) and cell proliferation (Cyquant) assays were performed to assess the cytotoxicity of DLinDMA- and DOTAP-containing liposomal formulations in HEK293 cells. Cells not treated with liposomes in their regular media and OptiMEM media were used as controls as the transfections were performed in serum-free media. Commercially available transfection reagents PEI Max and Transit LT1 were used as positive controls. The toxicity of DLinDMA liposomal formulation was observed at the concentrations ranging from 6.25 to 200 μM, whereas for DOTAP liposomal formulations, the toxicity was observed at the concentrations ranging from 12.5 to 200 μM (Figure 2A). Interestingly, the toxicity of commercially available transfection reagents such as PEI Max and Transit LT-1 at recommended 1:3 ratio of DNA:Transit LT1/PEI Max (3 μl reagent per 1 μg DNA) was similar compared with that of our liposomal formulations with higher ratio (Figure 2A). Similar to the cell viability studies, cell proliferation assay results also showed that these liposomal formulations are nontoxic at lower concentrations. A significant decrease in cell proliferation was only observed at the highest concentration 200 μM for DOTAP and at concentrations ranging from 25 to 200 μM for DLinDMA liposomal formulations (Figure 2B). As DLinDMA liposomes were less toxic compared with DOTAP, their cytotoxicity was also evaluated in SK-N-SH, a neuroblastoma cell line, at concentration range of 1.56–200 μM. Transit LT1 and PEI max were used as positive controls. The toxicity was observed at concentrations of 12.5 μM onward, which was similar to the effect observed in HEK293 cells (Figure 2C). These results indicate that liposomes with DOTAP and DLinDMA are relatively safe when compared with commercially available transfection reagents such as PEI Max and Transit LT1.

Figure 2. . Cytotoxicity studies of DLinDMA liposomes in HEK293 and SK-N-SH cell lines.

Figure 2. 

Blank liposomes cytotoxicity evaluated in HEK293 cells after 48 h by (A) MTT assay and (B) cell proliferation (Cyquant) assay with indicated concentrations (μM) of DOTAP and DLinDMA. (C) Blank DLinDMA liposomes cytotoxicity evaluated in SK-N-SH cells by MTT assay after 48 h with indicated concentrations (μM) of DLinDMA. Data represent mean ± standard error of mean of three individual experiments with n = 4 for each trial in HEK293 cell line and two individual experiments with n = 8 in SK-N-SH cells. Two-way ANOVA analysis in HEK293; One-way ANOVA analysis in SK-N-SH cell line; *p < 0.05, **p < 0.01 and ***p < 0.001.

Transfection efficiency of DLinDMA liposomes is higher than DOTAP liposomes

We used fluorescence microscopy to determine the transfection efficiency of DOTAP- and DLinDMA liposomes in HEK293 and SK-N-SH cells. The transfection efficiency of liposomes in both the cell lines was studied at different ratio of DNA to DLinDMA/DOTAP liposomes at 1:5, 1:10, 1:20, 1:40 and 1:60 (Figure 3A–C). The number of EGFP-expressing cells transfected with different ratios of liposomes was quantified (Figure 3D & E). The results show that transfection efficiency was higher with DLinDMA liposomes compared with DOTAP liposomes in HEK293 cells (Figure 3D). In addition, prominent transfection was observed with DLinDMA liposomes in SK-N-SH cells, a neuroblastoma cell line that is difficult to transfect (Figure 3E). Due to the higher transfection efficiency and lower cytotoxicity of DLinDMA liposomes compared with DOTAP liposomes, liposomes containing DLinDMA as the cationic lipid were selected to perform further experiments.

Figure 3. . DLinDMA liposomes show higher transfection efficiency than DOTAP liposomes.

Figure 3. 

 Representative fluorescent images demonstrating transfection efficiency with (A) DOTAP liposomes and (B) DLinDMA liposomes at 1:5, 1:10, 1:20, 1:40 and 1:60 mass ratio in HEK293 cells after 48 h of transfection. n = 6; Scale bar: 200 μm. (C) Representative fluorescent images demonstrating transfection efficiency with DLinDMA liposomes at 1:5, 1:10, 1:20, 1:40 and 1:60 mass ratio after 48 h of transfection in SK-N-SH cells. n = 4; Scale bar: 200 μm. (D) Quantification of transfection efficiency of DLinDMA and DOTAP liposomes in HEK293 cells shown above. (E) Quantification of transfection efficiency of DLinDMA liposomes in SK-N-SH cells shown above. Scale bar: 200 μm. Graphical data represent mean ± standard error of mean of three individual experiments with n = 3 for each trial. One-way analysis of variance with post hoc Tukey; *p < 0.05, **p < 0.01 and ***p < 0.001.

DLinDMA formed functional lipoplexes

The efficiency of the liposomes to form a complex binding with DNA was evaluated using electrophoretic mobility shift assay (EMSA). Naked pEGFP-N3 was used as a control and DNA to DLinDMA liposomes forming a complex with the at 1:1, 1:5, 1:10, 1:20, 1:40 and 1:60 mass ratio were studied. The liposomal formulations at 1:20, 1:40 and 1:60 mass ratio of DNA:DLinDMA showed the plasmid DNA complexation with no DNA migration as a result of binding and neutralization of the negatively charged DNA by the cationic liposomes (Figure 4). Based on the data from cytotoxicity assay, cellular transfections and EMSA, we decided to use DLinDMA liposomes at the ratio of 1:20 that corresponds to DLinDMA liposomes at 32 μM for immunofluorescence microscopy or 16 μM for immunoprecipitation experiments.

Figure 4. . Electrophoretic mobility shift assay gel image for indicated ratios of plasmid DNA:DLinDMA containing cationic liposomes.

Figure 4. 

Representative image from two independent experiments.

DLinDMA liposomes efficiently deliver laforin in cells

After studying the transfection efficiency of pEGFP-N3 in HEK293 and SK-N-SH cells, we evaluated the transfection of FLAG laforin in SK-N-SH neuroblastoma cells using cationic liposomes at DNA:DLinDMA mass ratio of 1:20. FLAG laforin-transfected cells were immunolabeled using fluorescent-dye conjugated secondary antibody to anti-FLAG antibody. As seen in Figure 5, fluorescence exhibited by cells transfected with pEGFP-N3 as well as FLAG-laforin indicated that cells were effectively transfected with DLinDMA cationic liposomes. In addition, cells transfected with pEGFP-N3 and FLAG-laforin showed similar cell density and cellular morphology when compared with control. As compared with HEK293 cells, SK-N-SH cells are difficult to transfect which implies that 1:20 mass ratio of DNA:liposomes are safe and effective vehicle for gene delivery. Furthermore, in order to assess intracellular protein persistence and length of protein expression, we performed DLinDMA liposomal transfection and studied the protein expression after 5- and 10-day periods in HEK293 cells by fluorescence microscopy and western blotting. The EGFP expression in HEK293 cells increased from days 5 to 10 (Figure 5B). The western blotting data showed that the EGFP or FLAG-laforin protein expression was stable after 5- and 10-days of transfection using DLinDMA liposomes in HEK293 cells (Figure 5C).

Figure 5. . DLinDMA liposome-mediated protein expression is stable in cells.

Figure 5. 

(A) Representative images of HEK293 cells transfected with pEGFP-N3 and FLAG laforin with DLinDMA liposomes (1:20 ratio) obtained by florescence microscopy. n = 3; Scale bar: 100 μm. (B) Representative fluorescence images of stable EGFP expression in HEK293 cells transfected with pEGFP-N3:DLinDMA liposomes (1:20 ratio) for 5 and 10 days. n = 3; Scale bar: 200 μm. (C) Representative western blots to observe the stable expression of GFP (27 kDa) or laforin (40 kDa) after 5- and 10-day transfections using DLinDMA liposomes (1:20 ratio) in HEK293 cells. β-actin (42 kDa) was used as loading control. n = 3.

DLinDMA lipoplexes deliver functional laforin with intact glucan phosphatase activity

Protein expression studies were performed using western blotting to confirm the presence of laforin in SK-N-SH cells transfected using the cationic liposomes. The cellular expression of laforin was confirmed in whole cell lysates by employing FLAG antibody that showed the presence of laforin in the samples transfected with FLAG laforin and mutant FLAG C266S laforin whereas, as expected, no bands were observed in control and blank liposome lanes (Figure 6A). Furthermore, IP laforin was assessed by western blot using laforin antibody. The results show the expression of laforin in the cells transfected with FLAG laforin and mutant FLAG C266S laforin (Figure 6B).

Figure 6. . DLinDMA lipoplexes deliver functional laforin.

Figure 6. 

(A) SK-N-SH cells were transfected with FLAG laforin and mutant FLAG C266S laforin using DLinDMA liposomes (1:20 ratio). Representative western blots for protein expression of laforin (40 kDa) and β-actin (42 kDa) in whole cell lysate are shown. n = 4. (B) Representative western blot for expression of FLAG-laforin (40 kDa) in IP samples. n = 5. (C) Glucan phosphatase activity of functionally active laforin transfected using DLinDMA liposomes. Graphical data represent mean ± standard error of mean of three individual experiments with n = 5 for each trial. One-way analysis of variance; ***p < 0.001.

IP: Immunoprecipitated.

Malachite green-based glucan phosphatase assay was used to determine whether the lipoplex-mediated transfected laforin is functionally active. The assay measures glucan phosphatase activity of laforin as it uses phosphorylated glucan polymer amylopectin as a substrate that is similar to laforin’s endogenous substrate glycogen [5,39]. Phosphate is released in the presence of laforin and forms a complex with ammonium molybdate in malachite green reagent and the resulting color can be measured at 620 nm. A standard curve was generated before the experiment and picomol of phosphate released was calculated using the standard curve. Glucan phosphatase activity was significant in FLAG laforin-transfected samples compared with control, blank liposomes and the cells transfected with mutant FLAG C266S laforin that is catalytically inactive (Figure 6C). In addition, we performed silencing of endogenous laforin in HEK293 cells and then introduced DLinDMA liposomal FLAG-laforin to confirm that the expression and activity observed is from laforin delivered through liposomes. Our data showed robust FLAG-laforin in siRNA-treated cells (Supplementary Figure 1) confirming data from Figure 6A and C. These results show that the DLinDMA cationic liposomes are not only efficient in delivering laforin DNA into the cells but also indicate that the expression and functional activity of laforin is intact.

Biocompatibility of DLinDMA liposomes analyzed by hemolytic potential

The effect of varying concentrations of DLinDMA in developed cationic liposomes was evaluated on mice RBCs by performing the in vitro hemolysis study. As shown in Figure 7, cationic liposomes exhibited negligible hemolysis (<5%) at all the tested concentrations (25, 50 and 100 μM) of cationic lipid (DLinDMA), in comparison to the positive control (SLS), which demonstrated 100% hemolysis. Notably, rapid and complete redispersion of RBCs following centrifugation implied that the DLinDMA cationic liposomes did not change the surface characteristics of RBCs at any of the tested concentrations.

Figure 7. . In vitro hemolysis study results of indicated DLinDMA concentrations of cationic liposomes.

Figure 7. 

PBS: Phosphate-buffered saline; SLS: Sodium lauryl sulfate.

Discussion

Gene therapy is a promising therapeutic platform and can be used for introduction of a functional gene, silencing of a mutant gene using siRNA technology and introducing a disease-modifying gene by using gene-editing technology. Gene therapy for rare genetic disorders such as cystic fibrosis, Duchenne muscular dystrophy, McArdle disease, ataxia telangiectasia and mucopolysaccharidoses have been under clinical investigation [40,41]. Gene therapy could be an excellent therapeutic avenue for LD as there is no cure available for this fatal disease. As LD occurs due to mutations in EPM2A or EMP2B genes and loss of function, delivering a functional copy of the gene via gene therapy could be a promising therapeutic approach. To our knowledge, this is the first investigation exploring the use of cationic liposome-based gene delivery as a treatment strategy for LD. In this manuscript, we developed and characterized DLinDMA cationic liposomes as a nanoformulation to deliver laforin DNA and explored its efficacy and safety.

Our study showed that the lipoplexes containing cationic lipids and laforin DNA were both safe and effective in cells tested at working concentration of mass ratio 1:20. The liposomes were also found to be relatively safe in both HEK293 and SK-N-SH cells when compared with commercially available transfection reagents such as PEI Max and Transit LT-1 based on the cytotoxicity data (Figure 2). The results of in vitro cell culture assays of cationic liposomes in HEK293 and SK-N-SH cells to study their transfection potential, expression and functional activity were promising. Transfection efficiency was comparable to commercially available transfection reagents such as PEI Max and Transit LT1 (Figure 3). Electrophoretic mobility shift assay results demonstrated better DNA entrapment ability of liposomes beginning at mass ratio of 1:20 of DNA and liposomes (Figure 4). From our immunofluorescence images, it was confirmed that these liposomes were able to deliver laforin DNA (Figure 5A). In addition, stable EGFP and laforin expression was observed in cellular persistence studies of liposome-gene delivery performed for 5- and 10 days (Figure 5B & C). Western blot results showed expression of FLAG indicating the liposome-mediated delivery of FLAG laforin into cells (Figure 6A & B). Glucan phosphatase activity assay showed that laforin delivered via cationic liposomes retained its biological activity (Figure 6C). In addition, in vitro hemolysis study resulted in negligible hemolysis by DLinDMA liposomes even at a higher concentration of cationic lipids (Figure 7), suggesting their systemic administration would be safe. Based on the results obtained, our approach could be a promising strategy among several research studies for LD that are at preclinical level.

Current investigational therapies for the treatment of LD aim to reduce the activity of GYS enzyme by small molecules or by using ASOs and/or aim to breakdown LBs by introducing amylase into the brain [25,42]. Metformin is an activator of AMPK and AMPK activation is known to inhibit glycogen synthesis and activate autophagy among its many other cellular effects [20]. In animal studies, metformin was shown to improve the neuropathological symptoms, reduced seizures and also decreased the number of LBs [43]. Wide spectrum of AEDs such as levetiracetam, sodium valproate, topiramate and benzodiazepines are therapeutically considered drugs for LD [17]. Though AEDs ameliorate seizure symptoms, they are unable to stop the disease progression. The high-fat, low-carb ketogenic diet has been around for many years as a therapeutic treatment for epileptic seizures [44,45]. However, a small population clinical trial indicated that ketogenic diet did not provide symptomatic relief and was unable to stop the disease progression [44].

Investigational ASO therapy in LD is currently in development to target the mRNA encoding brain GYS by Ionis pharmaceuticals [46]. The company has a US FDA-approved therapy for spinal muscular dystrophy, which is administered intrathecally. Although targeting and inhibiting GYS is a possible treatment for LD, this strategy could also lead to significant side effects as glycogen is known to play an important role in physiological brain functions such as learning and memory [47]. Dosage titration and targeting of the ASO will be crucial as glycogen storage disease type 0 with mutations in GYS1 or GYS2 are known to be debilitating [48]. Another therapy in development for LD is to introduce amylase into the brain to clear LBs [25]. α-Amylase is the only enzyme known to digest LBs and their degradation is mediated by breaking the α-1,4 glycosidic linkages in glycogen. An antibody–enzyme fusion called VAL-0417 is being developed by Valerion therapeutics, which utilizes α-amylase as an enzyme to therapeutically degrade neuronal glycogen and prevent the formation of LBs [25]. Intracerebroventricular injection of VAL-0417 in Lafora mouse models showed that LD can also be prevented by reducing the production of glycogen [25]. However, it is unclear whether this drug can reverse the effects. Another caveat is that we do not know yet whether LBs are a cause or consequence of LD similar to brain deposits in several other neurodegenerative diseases and thus clearing LBs may not provide desired clinical benefits. All the above-stated proposed therapies are an indirect approach to treat LD and do not address the principal cause of mutations in laforin or malin. As LD is a genetic disease resulting in the loss of function of proteins, gene therapy to express functional protein can be an excellent therapeutic strategy. Therefore, further advancement of gene technology in the clinical settings for LD requires optimization of gene delivery, target engagement, stability, prolonged efficacy and safety profile.

Liposomal delivery is favored over viral vector-based delivery as any gene delivery formulations need the ease of preparation and use. For AAV-based delivery, loss of gene upon infection with adenovirus serves as a drawback [49]. Transformation of host cells leading to immunogenicity and cancer are also potential hazards of viral vectors. Therefore, liposomal gene delivery is preferred over vector-based delivery for its ease of preparation and safety. This is evident as mRNA vaccines for COVID-19 by both Pfizer/BioNTech and NIAID/Moderna employ lipid nanoparticles as carriers [50]. Therefore, we intend to further study the laforin-lipoplexes in animal models and plan on continuing to develop formulation strategies that will effectively deliver genes in brain. Results from natural history and functional status study of patients with LD is currently ongoing and will provide a clinical benchmark for various LD therapeutics in development [51].

Conclusion

Our study is a novel attempt in preclinical development of gene therapy via DLinDMA liposomes for the treatment of LD. Our findings from various cell culture assays demonstrate that delivering gene through cationic liposomes can be a safe and effective strategy to restore functional protein for the treatment of rare genetic diseases such as LD. Further investigation is warranted to show that this study can be recapitulated in animals before the translation could be achieved for the gene therapy of LD and other neurodegenerative diseases.

Future perspective

The field of nanotechnology has been growing exponentially offering new opportunities for improving the diagnosis and treatment of diseases. Our current work is in vitro proof-of-concept study of EPM2A-loaded DLinDMA liposomes for the treatment of LD. Since LD is a neurodegenerative disease, development of liposomes and investigating their efficiency to deliver to brain is warranted. Subsequent preclinical in vivo studies will aid in validating the potential of these liposomes in leading their way to clinical trials.

Summary points.

  • Cationic liposomes with DOTAP or DLinDMA were formulated and their cytotoxicity and transfection efficiency was evaluated.

  • Optimized DLinDMA liposomes showed unimodal size distribution and high-positive surface charge, which would aid in incorporating the negatively charged DNA.

  • Transfection efficiencies of plasmid DNA-encapsulated liposomes were found to be effective in HEK293 and SK-N-SH cells at 1:20 mass ratio of DNA:DLinDMA.

  • Electrophoretic mobility shift assay results depicted binding of pEGFP-N3 with liposomes, indicating successful complexation between cationic liposomes and negatively charged DNA.

  • Western blotting showed expression of laforin in SK-N-SH cells. Most importantly, glucan phosphatase assay signified that laforin delivered via DLinDMA liposomes retains its functional activity.

  • DLinDMA liposomes showed negligible hemolysis suggesting their relative systemic safety.

  • Based on the results obtained in vitro, cationic DLinDMA liposomes could be a promising nonviral vector to efficiently deliver plasmid DNA to carry out further investigational studies for the treatment of rare genetic diseases such as Lafora disease.

Supplementary Material

Footnotes

Author contributions

Conceptualization was by VV Dukhande and K Patel. Methodology was performed by VV Dukhande, K Patel, HP Vemana, A Saraswat, SP Bhutkar. Validation was performed by VV Dukhande and K Patel. Formal analysis was performed by HP Vemana, A Saraswat. Investigation was performed by HP Vemana, A Saraswat, SP Bhutkar. Resources were provided VV Dukhande and K Patel. Data curation was performed by HP Vemana, A Saraswat, SP Bhutkar. Original draft preparation was performed by HP Vemana, A Saraswat. Review and editing were performed by VV Dukhande and K Patel. Visualization was performed by VV Dukhande and K Patel. Supervision was performed by VV Dukhande and K Patel. Project administration was performed by VV Dukhande and K Patel.

Financial & competing interests disclosure

This research was supported by an award from the National Institute of General Medical Sciences of the NIH under Award Number SC2GM125550 to VV Dukhande and by funds from the College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA to VV Dukhande and K Patel. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.No writing assistance was utilized in the production of this manuscript.

No writing assistance was utilized in the production of this manuscript.

Ethical conduct of research

The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all human or animal experimental investigations.

References

Papers of special note have been highlighted as: • of interest; •• of considerable interest

  • 1.Minassian BA. Lafora’s disease: towards a clinical, pathologic, and molecular synthesis. Pediatr. Neurol. 25(1), 21–29 (2001). [DOI] [PubMed] [Google Scholar]
  • 2.Minassian BA, Lee JR, Herbrick JA et al. Mutations in a gene encoding a novel protein tyrosine phosphatase cause progressive myoclonus epilepsy. Nat. Genet. 20(2), 171–174 (1998). [DOI] [PubMed] [Google Scholar]; •• Demonstrates that mutations in EPM2A gene result in detrimental effects in laforin protein, which effects glycogen metabolism and causes progressive myoclonus epilepsy of Lafora type.
  • 3.Minassian BA, Ianzano L, Meloche M et al. Mutation spectrum and predicted function of laforin in Lafora’s progressive myoclonus epilepsy. Neurology 55(3), 341–346 (2000). [DOI] [PubMed] [Google Scholar]
  • 4.Chan EM, Young EJ, Ianzano L et al. Mutations in NHLRC1 cause progressive myoclonus epilepsy. Nat. Genet. 35(2), 125–127 (2003). [DOI] [PubMed] [Google Scholar]
  • 5.Worby CA, Gentry MS, Dixon JE. Laforin, a dual specificity phosphatase that dephosphorylates complex carbohydrates. J. Biol. Chem. 281(41), 30412–30418 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gentry MS, Worby CA, Dixon JE. Insights into Lafora disease: malin is an E3 ubiquitin ligase that ubiquitinates and promotes the degradation of laforin. Proc. Natl Acad. Sci. USA 102(24), 8501–8506 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wang W, Roach PJ. Glycogen and related polysaccharides inhibit the laforin dual-specificity protein phosphatase. Biochem. Biophys. Res. Commun. 325(3), 726–730 (2004). [DOI] [PubMed] [Google Scholar]
  • 8.Gentry MS, Dowen RH 3rd, Worby CA, Mattoo S, Ecker JR, Dixon JE. The phosphatase laforin crosses evolutionary boundaries and links carbohydrate metabolism to neuronal disease. J. Cell Biol. 178(3), 477–488 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Sullivan M, Nitschke S, Steup M, Minassian B, Nitschke F. Pathogenesis of Lafora disease: transition of soluble glycogen to insoluble polyglucosan. IJMS 18(8), 1743 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dukhande VV, Sherwood AR, Gentry MS. Laforin. UCSD-Nature Molecule Pages (2010). [Google Scholar]
  • 11.Gentry MS, Romá-Mateo C, Sanz P. Laforin, a protein with many faces: glucan phosphatase, adapter protein, et alii. FEBS J. 280(2), 525–537 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]; • Provides a review summary that laforin is involved in several physiological processes in addition to its phosphatase activity.
  • 12.Duran J, Gruart A, Garcia-Rocha M, Delgado-Garcia JM, Guinovart JJ. Glycogen accumulation underlies neurodegeneration and autophagy impairment in Lafora disease. Hum. Mol. Genet. 23(12), 3147–3156 (2014). [DOI] [PubMed] [Google Scholar]
  • 13.DePaoli-Roach AA, Tagliabracci VS, Segvich DM, Meyer CM, Irimia JM, Roach PJ. Genetic depletion of the malin e3 ubiquitin ligase in mice leads to Lafora bodies and the accumulation of insoluble laforin. J. Biol. Chem. 285(33), 25372–25381 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Raththagala M, Brewer MK, Parker MW et al. Structural mechanism of laforin function in glycogen dephosphorylation and Lafora disease. Mol. Cell 57(2), 261–272 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Nitschke F, Wang P, Schmieder P et al. Hyperphosphorylation of glucosyl C6 carbons and altered structure of glycogen in the neurodegenerative epilepsy Lafora disease. Cell Metab. 17(5), 756–767 (2013). [DOI] [PubMed] [Google Scholar]
  • 16.Nitschke F, Ahonen SJ, Nitschke S, Mitra S, Minassian BA. Lafora disease - from pathogenesis to treatment strategies. Nat. Rev. Neurol. 14(10), 606–617 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Goldenberg MM. Overview of drugs used for epilepsy and seizures. P T. 35(7), 392–415 (2010). [PMC free article] [PubMed] [Google Scholar]
  • 18.Michelucci R, Pasini E, Riguzzi P, Andermann E, Kälviäinen R, Genton P. Myoclonus and seizures in progressive myoclonus epilepsies: pharmacology and therapeutic trials. Epileptic Disord. 18(S2), 145–153 (2016). [DOI] [PubMed] [Google Scholar]
  • 19.Goldsmith D, Minassian BA. Efficacy and tolerability of perampanel in ten patients with Lafora disease. Epilepsy Behav. 62, 132–135 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yang Y, Zhu B, Zheng F et al. Chronic metformin treatment facilitates seizure termination. Biochem. Biophys. Res. Commun. 484(2), 450–455 (2017). [DOI] [PubMed] [Google Scholar]
  • 21.Mikati MA, Tabbara F. Managing Lafora body disease with vagal nerve stimulation. Epileptic Disord. 19(1), 82–86 (2017). [DOI] [PubMed] [Google Scholar]
  • 22.Hajnsek S, Petelin Gadze Z, Borovecki F et al. Vagus nerve stimulation in Lafora body disease. Epilepsy Behav. Case Rep. 1, 150–152 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bennett CF, Krainer AR, Cleveland DW. Antisense oligonucleotide therapies for neurodegenerative diseases. Annu. Rev. Neurosci. 42(1), 385–406 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Goldsmith D, Minassian BA. Extraneurological sparing in long-lived typical Lafora disease. Epilepsia Open 3(2), 295 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Brewer MK, Uittenbogaard A, Austin GL et al. Targeting pathogenic Lafora bodies in Lafora disease using an antibody-enzyme fusion. Cell Metab. 30(4), 689–705.e6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Confirms that α-amylase is capable of degrading Lafora bodies and antibody enzyme fusion therapy can be used for the treatment of Lafora disease.
  • 26.Austin GL, Simmons ZR, Klier JE et al. Central nervous system delivery and biodistribution analysis of an antibody–enzyme fusion for the treatment of Lafora disease. Mol. Pharmaceutics 16(9), 3791–3801 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Shahryari A, Saghaeian Jazi M, Mohammadi S et al. Development and clinical translation of approved gene therapy products for genetic disorders. Front. Genet. 10, 868 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bates M. Advances in gene therapy offer hope for rare disorders. IEEE Pulse 10(6), 9–12 (2019). [DOI] [PubMed] [Google Scholar]
  • 29.Lechardeur D, Lukacs GL. Intracellular barriers to non-viral gene transfer. Curr. Gene Ther. 2(2), 183–194 (2002). [DOI] [PubMed] [Google Scholar]
  • 30.Ruponen M, Honkakoski P, Rönkkö S, Pelkonen J, Tammi M, Urtti A. Extracellular and intracellular barriers in non-viral gene delivery. J. Control. Rel. 93(2), 213–217 (2003). [DOI] [PubMed] [Google Scholar]
  • 31.Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat. Rev. Genet. 4(5), 346–358 (2003). [DOI] [PubMed] [Google Scholar]
  • 32.Ramamoorth M, Narvekar A. Non viral vectors in gene therapy - an overview. J. Clin. Diagn. Res. 9(1), GE01–GE06 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Patil S, Gao Y-G, Lin X et al. The Development of functional non-viral vectors for gene delivery. Int. J. Mol. Sci. 20(21), 5491 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Faheem AM, Elkordy AA, Hassan E, Carr-Wilkinson J. Preparation, characterisation and cell transfection of cationic liposomes in gene therapy. Br. J. Pharm. 4(1), 618 (2019). [Google Scholar]
  • 35.Shim G, Kim M-G, Park JY, Oh Y-K. Application of cationic liposomes for delivery of nucleic acids. Asian J. Pharm. Sci. 8(2), 72–80 (2013). [Google Scholar]; • Demonstrates the importance and advantages of cationic liposomes for gene delivery, which could help in developing the treatment strategies for rare genetic disorders.
  • 36.Bae Y-U, Huh J-W, Kim B-K, Park HY, Seu Y-B, Doh K-O. Enhancement of liposome mediated gene transfer by adding cholesterol and cholesterol modulating drugs. Biochim. Biophys. Acta 1858(12), 3017–3023 (2016). [DOI] [PubMed] [Google Scholar]
  • 37.Du Z, Munye MM, Tagalakis AD, Manunta MDI, Hart SL. The role of the helper lipid on the DNA transfection efficiency of lipopolyplex formulations. Sci. Rep. 4(1), 7107 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dukhande VV, Rogers DM, Roma-Mateo C et al. Laforin, a dual specificity phosphatase involved in Lafora disease, is present mainly as monomeric form with full phosphatase activity. PLoS ONE 6(8), e24040 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sherwood AR, Johnson MB, Delgado-Escueta AV, Gentry MS. A bioassay for Lafora disease and laforin glucan phosphatase activity. Clin. Biochem. 46(18), 1869–1876 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Alton EWFW, Armstrong DK, Ashby D et al. Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir. Med. 3(9), 684–691 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.McNamara EL, Taylor RL, Clayton JS et al. Systemic AAV8-mediated delivery of a functional copy of muscle glycogen phosphorylase (Pygm) ameliorates disease in a murine model of McArdle disease. Hum. Mol. Genet. 29(1), 20–30 (2020). [DOI] [PubMed] [Google Scholar]
  • 42.Tang B, Frasinyuk MS, Chikwana VM et al. Discovery and development of small-molecule inhibitors of glycogen synthase. J. Med. Chem. 63(7), 3538–3551 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; • Proves the important role of glycogen synthase inhibitors in suppressing accumulation of glycogen, which could be a potential treatment of Lafora disease.
  • 43.Sánchez-Elexpuru G, Serratosa JM, Sanz P, Sánchez MP. 4-Phenylbutyric acid and metformin decrease sensitivity to pentylenetetrazol-induced seizures in a malin knockout model of Lafora disease. Neuroreport 28(5), 268–271 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cardinali S, Canafoglia L, Bertoli S et al. A pilot study of a ketogenic diet in patients with Lafora body disease. Epilepsy Res. 69(2), 129–134 (2006). [DOI] [PubMed] [Google Scholar]
  • 45.Israelian L, Wang P, Gabrielian S, Zhao X, Minassian BA. Ketogenic diet reduces Lafora bodies in murine Lafora disease. Neurol. Genet. 6(6), e533 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hospital for Sick Children HSC. Ionis Pharmaceuticals Inc.: US20190194666 (2019).
  • 47.Rich LR, Harris W, Brown AM. The role of brain glycogen in supporting physiological function. Front. Neurosci. 13, 1176 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wolfsdorf JI, Weinstein DA. Glycogen storage diseases. Rev. Endocr. Metab. Disord. 4(1), 95–102 (2003). [DOI] [PubMed] [Google Scholar]
  • 49.Colella P, Ronzitti G, Mingozzi F. Emerging issues in AAV-mediated in vivo gene therapy. Mol. Ther. Methods Clin. Dev. 8, 87–104 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ye T, Zhong Z, García-Sastre A, Schotsaert M, Geest BGD. Current status of COVID-19 (Pre)clinical vaccine development. Angew. Chem. Int. Ed. Engl. 59(43), 18885–18897 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Brewer MK, Gentry MS. The 3rd International Lafora Workshop: evidence for a cure. Epilepsy Behav. 81, 125–127 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Provides evidence that injection of antisense oligonucleotides targeting glycogen synthase reduced Lafora bodies and a natural history study on a small cohort of patients is initiated by Ionis Pharmaceuticals.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from Nanomedicine are provided here courtesy of Taylor & Francis

RESOURCES