Skeletal muscle effects of antisense oligonucleotides targeting glycogen synthase 1 in a mouse model of Pompe disease

Lan Weiss , Michele Carrer , Alyaa Shmara , Angela Martin , Hong Yin , Pallabi Pal , Cheng Cheng , Lac Ta , Victoria Boock , Yasamin Fazeli , Mindy Chang , Marvin Paguio , Jonathan Lee , Howard Yu , John Weiss , Tamar R Grossman , Nina Raben , Paymaan Jafar-Nejad , Virginia Kimonis

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (4) : e70314

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (4) : e70314 DOI: 10.1002/ctm2.70314
RESEARCH ARTICLE

Skeletal muscle effects of antisense oligonucleotides targeting glycogen synthase 1 in a mouse model of Pompe disease

Author information +
History +
PDF

Abstract

Pompe disease (PD) is a progressive myopathy caused by the aberrant accumulation of glycogen in skeletal and cardiac muscle resulting from the deficiency of the enzyme acid alpha-glucosidase (GAA). Administration of recombinant human GAA as enzyme replacement therapy (ERT) works well in alleviating the cardiac manifestations of PD but loses sustained benefit in ameliorating the skeletal muscle pathology. The limited efficacy of ERT in skeletal muscle is partially attributable to its inability to curb the accumulation of new glycogen produced by the muscle enzyme glycogen synthase 1 (GYS1). Substrate reduction therapies aimed at knocking down GYS1 expression represent a promising avenue to improve Pompe myopathy. However, finding specific inhibitors for GYS1 is challenging given the presence of the highly homologous GYS2 in the liver. Antisense oligonucleotides (ASOs) are chemically modified oligomers that hybridise to their complementary target RNA to induce their degradation with exquisite specificity. In the present study, we show that ASO-mediated Gys1 knockdown in the Gaa−/− mouse model of PD led to a robust reduction in glycogen accumulation in skeletal muscle. In addition, combining Gys1 ASO with ERT slightly further reduced glycogen content in muscle, eliminated autophagic buildup and lysosomal dysfunction, and improved motor function in Gaa−/− mice. Our results provide a strong foundation for validation of the use of Gys1 ASO, alone or in combination with ERT, as a therapy for PD. We propose that early administration of Gys1 ASO in combination with ERT may be the key to preventative treatment options in PD.

Keywords

antisense oligonucleotides (ASOs) / Enzyme replacement therapy (ERT) / Gaa-/- mouse model / glycogen synthase 1 (GYS1) / Pompe disease / skeletal muscle

Cite this article

Download citation ▾
Lan Weiss, Michele Carrer, Alyaa Shmara, Angela Martin, Hong Yin, Pallabi Pal, Cheng Cheng, Lac Ta, Victoria Boock, Yasamin Fazeli, Mindy Chang, Marvin Paguio, Jonathan Lee, Howard Yu, John Weiss, Tamar R Grossman, Nina Raben, Paymaan Jafar-Nejad, Virginia Kimonis. Skeletal muscle effects of antisense oligonucleotides targeting glycogen synthase 1 in a mouse model of Pompe disease. Clinical and Translational Medicine, 2025, 15(4): e70314 DOI:10.1002/ctm2.70314

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Tang H, Feuchtbaum L, Sciortino S, et al. The first year experience of newborn screening for pompe disease in California. Int J Neonatal Screen. 2020; 6: 9.

[2]

Raben N, Nichols RC, Boerkoel C, Plotz P. Genetic defects in patients with glycogenosis type II (acid maltase deficiency). Muscle Nerve Suppl. 1995; 3: S70-74.

[3]

Kroos MA, Van der Kraan M, Van Diggelen OP, et al. Glycogen storage disease type II: frequency of three common mutant alleles and their associated clinical phenotypes studied in 121 patients. J Med Genet. 1995; 32: 836-837.

[4]

Raben N, Nagaraju K, Lee E, et al. Targeted disruption of the acid alpha-glucosidase gene in mice causes an illness with critical features of both infantile and adult human glycogen storage disease type II. J Biol Chem. 1998; 273: 19086-19092.

[5]

Prater SN, Patel TT, Buckley AF, et al. Skeletal muscle pathology of infantile Pompe disease during long-term enzyme replacement therapy. Orphanet J Rare Dis. 2013; 8: 90.

[6]

Bellotti AS, Andreoli L, Ronchi D, Bresolin N, Comi GP, Corti S. Molecular approaches for the treatment of Pompe disease. Mol Neurobiol. 2020; 57: 1259-1280.

[7]

Xu S, Lun Y, Frascella M, et al. Improved efficacy of a next-generation ERT in murine Pompe disease. JCI Insight. 2019; 4: e125358.

[8]

Kuperus E, Kruijshaar ME, Wens SCA, et al. Long-term benefit of enzyme replacement therapy in Pompe disease: a 5-year prospective study. Neurology. 2017; 89: 2365-2373.

[9]

Alandy-Dy J, Wencel M, Hall K, et al. Variable clinical features and genotype-phenotype correlations in 18 patients with late-onset Pompe disease. Ann Transl Med. 2019; 7: 276.

[10]

Raben N, Fukuda T, Gilbert AL, et al. Replacing acid alpha-glucosidase in Pompe disease: recombinant and transgenic enzymes are equipotent, but neither completely clears glycogen from type II muscle fibers. Mol Ther. 2005; 11: 48-56.

[11]

Miller CM, Harris EN. Antisense oligonucleotides: treatment strategies and cellular internalization. RNA Dis. 2016; 3: e1393.

[12]

Drost MR, Schaart G, van Dijk P, et al. Both type 1 and type 2a muscle fibers can respond to enzyme therapy in Pompe disease. Muscle Nerve. 2008; 37: 251-255.

[13]

Taylor KM, Meyers E, Phipps M, et al. Dysregulation of multiple facets of glycogen metabolism in a murine model of Pompe disease. PLoS One. 2013; 8: e56181.

[14]

Canibano-Fraile R, Harlaar L, Dos Santos CA, et al. Lysosomal glycogen accumulation in Pompe disease results in disturbed cytoplasmic glycogen metabolism. J Inherit Metab Dis. 2023; 46: 101-115.

[15]

van der Ploeg A, Carlier PG, Carlier RY, et al. Prospective exploratory muscle biopsy, imaging, and functional assessment in patients with late-onset Pompe disease treated with alglucosidase alfa: the EMBASSY Study. Mol Genet Metab. 2016; 119: 115-123.

[16]

Clayton NP, Nelson CA, Weeden T, et al. Antisense oligonucleotide-mediated suppression of muscle glycogen synthase 1 synthesis as an approach for substrate reduction therapy of Pompe disease. Mol Ther Nucleic Acids. 2014; 3: e206.

[17]

Douillard-Guilloux G, Raben N, Takikita S, et al. Restoration of muscle functionality by genetic suppression of glycogen synthesis in a murine model of Pompe disease. Hum Mol Genet. 2010; 19: 684-696.

[18]

Meena NK, Randazzo D, Raben N, Puertollano R. AAV-mediated delivery of secreted acid alpha-glucosidase with enhanced uptake corrects neuromuscular pathology in Pompe mice. JCI Insight. 2023; 8: e170199.

[19]

Douillard-Guilloux G, Raben N, Takikita S, Batista L, Caillaud C, Richard E. Modulation of glycogen synthesis by RNA interference: towards a new therapeutic approach for glycogenosis type II. Hum Mol Genet. 2008; 17: 3876-3886.

[20]

Crooke ST, Baker BF, Pham NC, et al. The effects of 2'-O-methoxyethyl oligonucleotides on renal function in humans. Nucleic Acid Ther. 2018; 28: 10-22.

[21]

Crooke ST, Baker BF, Xia S, et al. Integrated assessment of the clinical performance of GalNAc(3)-conjugated 2'-O-methoxyethyl chimeric antisense oligonucleotides: i. human volunteer experience. Nucleic Acid Ther. 2019; 29: 16-32.

[22]

Crooke ST, Liang XH, Baker BF, Crooke RM. Antisense technology: a review. J Biol Chem. 2021; 296: 100416.

[23]

Shen W, De Hoyos CL, Migawa MT, et al. Chemical modification of PS-ASO therapeutics reduces cellular protein-binding and improves the therapeutic index. Nat Biotechnol. 2019; 37: 640-650.

[24]

Crooke ST, Vickers TA, Liang XH. Phosphorothioate modified oligonucleotide-protein interactions. Nucleic Acids Res. 2020; 48: 5235-5253.

[25]

Ramms B, Patel S, Sun X, et al. Interventional hepatic apoC-III knockdown improves atherosclerotic plaque stability and remodeling by triglyceride lowering. JCI Insight. 2022; 7: e158414.

[26]

Walsh M, Bethune C, Smyth A, et al. Phase 2 study of the factor XI antisense inhibitor IONIS-FXI(Rx) in patients with ESRD. Kidney Int Rep. 2022; 7: 200-209.

[27]

Nitschke S, Sullivan MA, Mitra S, et al. Glycogen synthase downregulation rescues the amylopectinosis of murine RBCK1 deficiency. Brain. 2022; 145: 2361-2377.

[28]

Karam C, Brown D, Yang M, et al. Long-term treatment effects of inotersen on health-related quality of life in patients with hATTR amyloidosis with polyneuropathy: analysis of the open-label extension of the NEURO-TTR trial. Muscle Nerve. 2022; 66: 438-446.

[29]

Karam C, Brown D, Yang M, et al. Factors associated with increased health-related quality-of-life benefits in hereditary transthyretin amyloidosis polyneuropathy patients treated with inotersen. Muscle Nerve. 2022; 66: 319-328.

[30]

Raben N, Nagaraju K, Lee E, Plotz P. Modulation of disease severity in mice with targeted disruption of the acid alpha-glucosidase gene. Neuromuscul Disord. 2000; 10: 283-291.

[31]

Raben N, Ralston E, Chien YH, et al. Differences in the predominance of lysosomal and autophagic pathologies between infants and adults with Pompe disease: implications for therapy. Mol Genet Metab. 2010; 101: 324-331.

[32]

Ullman JC, Mellem KT, Xi Y, et al. Small-molecule inhibition of glycogen synthase 1 for the treatment of Pompe disease and other glycogen storage disorders. Sci Transl Med. 2024; 16: eadf1691.

[33]

Roman W, Gomes ER. Nuclear positioning in skeletal muscle. Semin Cell Dev Biol. 2018; 82: 51-56.

[34]

Hansson KA, Eftestol E. Scaling of nuclear numbers and their spatial arrangement in skeletal muscle cell size regulation. Mol Biol Cell. 2023; 34: pe3.

[35]

Lim JA, Sun B, Puertollano R, Raben N. Therapeutic benefit of autophagy modulation in Pompe disease. Mol Ther. 2018; 26: 1783-1796.

[36]

Pugsley HR. Assessing autophagic flux by measuring LC3, p62, and LAMP1 co-localization using multispectral imaging flow cytometry. J Vis Exp. 2017(125): 55637.

[37]

Pugsley HR. Quantifying autophagy: measuring LC3 puncta and autolysosome formation in cells using multispectral imaging flow cytometry. Methods. 2017; 112: 147-156.

[38]

Korlimarla A, Lim JA, Kishnani PS, Sun B. An emerging phenotype of central nervous system involvement in Pompe disease: from bench to bedside and beyond. Ann Transl Med. 2019; 7: 289.

[39]

Barker SJ, Thayer MB, Kim C, et al. Targeting the transferrin receptor to transport antisense oligonucleotides across the mammalian blood-brain barrier. Sci Transl Med. 2024; 16: eadi2245.

[40]

Crawford L, Rosch J, Putnam D. Concepts, technologies, and practices for drug delivery past the blood-brain barrier to the central nervous system. J Control Release. 2016; 240: 251-266.

[41]

Geary RS, Norris D, Yu R, Bennett CF. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev. 2015; 87: 46-51.

[42]

Chien YH, Hwu WL, Lee NC. Pompe disease: early diagnosis and early treatment make a difference. Pediatr Neonatol. 2013; 54: 219-227.

[43]

Chen S, Heendeniya SN, Le BT, et al.. Splice-modulating antisense oligonucleotides as therapeutics for inherited metabolic diseases. BioDrugs. 2024; 38: 177-203.

[44]

Crooke ST, Liang XH, Crooke RM, Baker BF, Geary RS. Antisense drug discovery and development technology considered in a pharmacological context. Biochem Pharmacol. 2021; 189: 114196.

[45]

Boros BD, Schoch KM, Kreple CJ, Miller TM. Antisense oligonucleotides for the study and treatment of ALS. Neurotherapeutics. 2022; 19: 1145-1158.

[46]

Bailey JK, Shen W, Liang XH, Crooke ST. Nucleic acid binding proteins affect the subcellular distribution of phosphorothioate antisense oligonucleotides. Nucleic Acids Res. 2017; 45: 10649-10671.

[47]

Ersoz E, Demir-Dora D. Unveiling the potential of antisense oligonucleotides: mechanisms, therapies, and safety insights. Drug Dev Res. 2024; 85: e22187.

[48]

Lim KR, Maruyama R, Yokota T. Eteplirsen in the treatment of Duchenne muscular dystrophy. Drug Des Devel Ther. 2017; 11: 533-545.

[49]

Mendell JR, Rodino-Klapac LR, Sahenk Z, et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol. 2013; 74: 637-647.

[50]

Sazani P, Ness KP, Weller DL, Poage D, Nelson K, Shrewsbury AS. Chemical and mechanistic toxicology evaluation of exon skipping phosphorodiamidate morpholino oligomers in mdx mice. Int J Toxicol. 2011; 30: 322-333.

[51]

Frank DE, Schnell FJ, Akana C, et al. Increased dystrophin production with golodirsen in patients with Duchenne muscular dystrophy. Neurology. 2020; 94: e2270-e2282.

[52]

Clemens PR, Rao VK, Connolly AM, et al. Safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy amenable to exon 53 skipping: a phase 2 randomized clinical trial. JAMA Neurol. 2020; 77: 982-991.

[53]

Finkel RS, Mercuri E, Darras BT, et al. Nusinersen versus Sham control in infantile-onset spinal muscular atrophy. N Engl J Med. 2017; 377: 1723-1732.

[54]

Hua Y, Sahashi K, Hung G, et al. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes Dev. 2010; 24: 1634-1644.

[55]

van der Wal E, Bergsma AJ, Pijnenburg JM, van der Ploeg AT, Pijnappel W. Antisense oligonucleotides promote exon inclusion and correct the common c.-32-13T>G GAA splicing variant in Pompe disease. Mol Ther Nucleic Acids. 2017; 7: 90-100.

[56]

van der Wal E, Bergsma AJ, van Gestel TJM, et al. GAA deficiency in Pompe disease is alleviated by exon inclusion in iPSC-derived skeletal muscle cells. Mol Ther Nucleic Acids. 2017; 7: 101-115.

[57]

Goina E, Peruzzo P, Bembi B, Dardis A, Buratti E. glycogen reduction in myotubes of late-onset Pompe disease patients using antisense technology. Mol Ther. 2017; 25: 2117-2128.

[58]

Moulton HM, Moulton JD. Morpholinos and their peptide conjugates. Biochimica et Biophysica Acta. 2010;1798: 2296-2303.

[59]

Baker BF, Lot SS, Condon TP, et al. 2'-O-(2-Methoxy)ethyl-modified anti-intercellular adhesion molecule 1 (ICAM-1) oligonucleotides selectively increase the ICAM-1 mRNA level and inhibit formation of the ICAM-1 translation initiation complex in human umbilical vein endothelial cells. J Biol Chem. 1997; 272: 11994-12000.

[60]

Raben N, Lu N, Nagaraju K, et al. Conditional tissue-specific expression of the acid alpha-glucosidase (GAA) gene in the GAA knockout mice: implications for therapy. Hum Mol Genet. 2001; 10: 2039-2047.

[61]

Baik AD, Calafati P, Zhang X, et al. Cell type-selective targeted delivery of a recombinant lysosomal enzyme for enzyme therapies. Mol Ther. 2021; 29: 3512-3524.

[62]

Tresse E, Salomons FA, Vesa J, et al. VCP/p97 is essential for maturation of ubiquitin-containing autophagosomes and this function is impaired by mutations that cause IBMPFD. Autophagy. 2010; 6: 217-227.

[63]

Cheng C, Weiss L, Leinonen H, et al. VCP/p97 inhibitor CB-5083 modulates muscle pathology in a mouse model of VCP inclusion body myopathy. J Transl Med. 2022; 20: 21.

[64]

Yin HZ, Wang HL, Ji SG, et al. Rapid intramitochondrial Zn2+ accumulation in CA1 hippocampal pyramidal neurons after transient global ischemia: a possible contributor to mitochondrial disruption and cell death. J Neuropathol Exp Neurol. 2019; 78: 655-664.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

9

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/