Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jan 1.
Published in final edited form as: Curr Gene Ther. 2015;15(4):338–347. doi: 10.2174/1566523215666150630132253

Preclinical Development of New Therapy for Glycogen Storage Diseases

Baodong Sun 1, Elizabeth D Brooks 1,2, Dwight D Koeberl 1,3,*
PMCID: PMC4682885  NIHMSID: NIHMS743922  PMID: 26122079

Abstract

Glycogen storage disease (GSD) consists of more than 10 discrete conditions for which the biochemical and genetic bases have been determined, and new therapies have been under development for several of these conditions. Gene therapy research has generated proof-of-concept for GSD types I (von Gierke disease) and II (Pompe disease). Key features of these gene therapy strategies include the choice of vector and regulatory cassette, and recently adeno-associated virus (AAV) vectors containing tissue-specific promoters have achieved a high degree of efficacy. Efficacy of gene therapy for Pompe disease depend upon the induction of immune tolerance to the therapeutic enzyme. Efficacy of von Gierke disease is transient, waning gradually over the months following vector administration. Small molecule therapies have been evaluated with the goal of improving standard of care therapy or ameliorating the cellular abnormalities associated with specific GSDs. The receptor-mediated uptake of the therapeutic enzyme in Pompe disease was enhanced by administration of β2 agonists. Rapamycin reduced the liver fibrosis observed in GSD III. Further development of gene therapy could provide curative therapy for patients with GSD, if efficacy from preclinical research is observed in future clinical trials and these treatments become clinically available.

Keywords: glycogen storage disease, adeno-associated virus (AAV), Pompe disease, von Gierke disease

Introduction

Glycogen storage disease (GSD) is caused by the inherited deficiency of an enzyme involved in either the degradation or synthesis of glycogen. More than 10 such conditions have been characterized, and the features of the well characterized GSDs are listed in the Table [1]. Typically a GSD involves primarily liver, striated muscle, or both tissues. Liver GSDs, including type I, III, IV, VI, and IX, usually cause hepatomegaly and hypoglycemia during fasting, while muscle GSDs cause exercise intolerance, cramping, progressive weakness with variable heart involvement in the form of cardiomyopathy (Table). With the exception of GSD II (Pompe disease), for which an enzyme replacement therapy (ERT) has been developed, therapy is limited to dietary modification or organ transplantation [1]. Therefore, new therapies are under development to address the unmet need for treatment in the GSDs.

Table 1.

Features of the Classical Glycogen Storage Diseases

Type/Eponym Enzyme Deficiency Presenting Clinical Features Primary Tissue Involvement Animal Model Vector Administered
I/von Gierke Glucose-6-phosphatase Growth retardation, hepatomegaly, hypoglycemia, lactic acidemia, hyperuricemia, and hyperlipidemia Liver, kidney Maltese dog, mouse AAV, Ad, LV, plasmid
II/Pompe Acid alpha-glucosidase Cardiomyopathy, myopathy, respiratory failure Heart, skeletal muscle, nerves Lapphund dog, mouse AAV, Ad, LV
III/Cori, Forbes Debrancher hepatomegaly, hypoglycemia, hyperlipidemia, and growth retardation Liver, heart, skeletal muscles Curly coated retriever dog, mouse None
IV/Anderson Branching Hepatosplenomegaly, failure to thrive Liver, heart, nerves Norwegian forest cat, mouse None
V/McArdle Muscle phosphorylase Exercise intolerance, muscle cramps Skeletal muscle Charlois cattle, sheep, mouse AAV, Ad
VII/Tarui Phosphofructokinase Exercise intolerance, muscle cramps, hemolytic anemia Skeletal muscle, erythrocytes English Springer spaniel dog, mouse None

Experiments with gene therapy have demonstrated proof-of-concept in several GSDs in animal models. Animal models that mimic human GSDs have been described, and confirmed by biochemical and genetic evaluations (Table). These models have been used to develop gene therapy for GSD I, II, and V. The goal of gene therapy for a GSD has been to stably replace the pertinent enzyme in the affected tissues. All of the major classes of vectors have been employed to deliver a therapeutic transgene in a GSD (Table), including adeno-associated virus (AAV), adenovirus (Ad), retroviral in the form of lentiviral (LV), and non-viral plasmid vectors. The features of each of these vector systems have been summarized [2], and relevant characteristics are presented below in the context of preclinical studies in the animal models for each GSD.

Potential for clinical development of gene therapy in Pompe disease

The deficiency of acid α-glucosidase (GAA) causes Pompe disease. Pompe disease stands out among lysosomal storage disorders due to the lack of devastating central nervous system involvement, rendering it much more amenable to treatment even later in life. GAA normally functions as an acid hydrolase that metabolizes lysosomal glycogen, and GAA deficiency causes lysosomal glycogen accumulation in virtually all tissues [3]. However, Pompe disease affects the heart and skeletal muscle primarily, and untreated infants with Pompe disease develop profound weakness and hypotonia. Infantile-onset Pompe disease causes death early in childhood from cardiorespiratory failure related to an underlying hypertrophic cardiomyopathy, if enzyme replacement therapy (ERT) is delayed or the patient fails to respond sustainably to ERT [4, 5]. Late-onset Pompe disease features progressive weakness without significant cardiomyopathy, and patients typically survive until late adulthood.

Pompe disease was demonstrated in Lapphund dogs with megaesophagus, exercise intolerance, and recurrent emesis [6]. Glycogen accumulations consisting of membrane bound vacuoles were present in the heart, skeletal, and smooth muscle. The genetic basis was a c.2237G>A change corresponding to the nonsense mutation p.W746* in the acid α-glucosidase gene [7].

Need for immune tolerance to GAA in Pompe disease

ERT with GAA, like other lysosomal enzymes, depends upon mannose-6-phophate receptor mediated uptake in affected tissues [8]. The availability of ERT with recombinant human (rh) GAA has prolonged survival and ameliorates cardiomyopathy of infantile Pompe disease [9]. Documented limitations of ERT in Pompe disease include the requirement for frequent intravenous infusions of high levels of GAA to achieve efficacy, degree of pre-ERT muscle damage, and the possibility of humoral immunity [10]. The rhGAA doses are markedly higher than doses required for ERT in other lysosomal storage disorders, reflecting the high threshold for correction of GAA deficiency in the skeletal muscle of Pompe disease patients [11].

ERT has prolonged survival in the majority of patients with infantile Pompe disease, although many patients die or remain very weak despite compliance with ERT. Pompe disease patients who lack any residual GAA protein are deemed cross-reacting immune material negative (CRIM-negative). Among the poor responders, were many CRIM-negative patients who formed high, sustained anti-rhGAA antibody titers (HSAT). Patients with HSAT demonstrated greatly increased mortality, in comparison with patients who formed no or only low titer antibodies [12]. In the first pilot study of ERT in Pompe disease using Chinese hamster ovary cell-derived rhGAA, the two patients who were CRIM-negative produced higher titers of anti-rhGAA antibodies than the third patient who was CRIM-positive. This corresponded with a markedly reduced efficacy of the treatment in the CRIM-negative patients [8]. Furthermore, the ability to prevent antibody formation in patients at risk for HSAT with immunosuppression, which significantly prolonged survival, confirmed the clinical relevance of HSAT [13]. The relevance of antibody formation to efficacy of therapy in Pompe disease has been emphasized by the poor response of CRIM-negative patients to ERT, which correlated with the onset of HSAT [10, 12].

Gene therapy has been developed as an alternative approach to therapy in Pompe disease, which can also prevent immune responses to GAA [14]. The availability of novel AAV serotypes with increased tropism for target tissues has markedly advanced the field of gene therapy in the past 12 years [15, 16]. AAV vectors provide an optimized gene delivery vehicle, because AAV is a non-pathogenic virus and vectors derived from AAV contain no viral genes. Furthermore, vectors packaged with the novel AAV serotypes elicit a lower immune response than the original AAV2 capsid [17]. AAV serotype 8 has been developed to pseudotype AAV2 vector genomes as AAV2/8 vectors, which feature an extremely high tropism for liver [15]. AAV2/8 vectors transduce liver with higher efficiency due to rapid uncoating and delivery of the genome to the transduced cells’ nuclei [18], and the entire liver can be transduced from an intravenous injection of an AAV2/8 vector [19]. Consequently, AAV2/8 vectors have been developed in preclinical models to effectively treat inherited metabolic diseases, such as Pompe disease. Importantly, AAV vectors have a favorable safety profile for clinical applications [16].

Multiple studies with AAV vectors encoding GAA have previously treated GAA-KO mice successfully [2024]. However, antibody formation remains a major obstacle for GAA gene transfer approaches in which constitutive – vs. tissue-specific – expression of the therapeutic transgene is utilized to achieve disease correction [21, 22, 25]. A highly active muscle-specific regulatory cassette achieved wide-spread correction of GAA deficiency in striated muscles, and reduced accumulated glycogen in those muscles. However, AAV vectors containing muscle specific regulatory cassettes have required high numbers of vector particles (vp) to achieve biochemical correction, >4×1013 vp/body weight [26]. Furthermore, muscle-specific expression provoked anti-GAA antibodies with negative effects upon efficacy [27]. Recently, a new approach to immune suppression was shown to prevent anti-body formation in response to an immunogenic AAV vector. This strategy used coreceptor blockade with a non-depleting anti-C44 monoclonal antibody to block activation of helper T cells, which successfully prevented antibody formation and improved the efficacy from gene therapy [28].

Pompe disease can be effectively treated by converting the liver to a depot for secretion of GAA, accompanied by uptake of GAA in the heart and skeletal muscle [29, 30]. This concept was further developed with an AAV2/8 vector containing a liver-specific regulatory cassette to drive high-level GAA secretion from the liver [22]. This vector induced immune tolerance to prevent the anti-GAA antibody response in adult, immunocompetent GAA (−/−) mice. Preventing antibody formation in patients at risk for HSAT via immunosuppression has been demonstrated to significantly prolong survival [13]. AAV2/8-LSPhGAApA expressed hGAA exclusively in the liver and activated regulatory T (Treg) cells thereby preventing antibody formation [27]. Adaptive transfer of CD4+ T cells further confirmed the role of Tregs in achieving immune tolerance to GAA [31]. Immune tolerance to GAA from AAV2/8-LSPhGAApA might improve the response to ERT in patients at risk of HSAT [32], and in the long-term, at high enough doses, may even provide a curative therapy for Pompe disease. An intriguing variation on this theme was demonstrated by combining two vectors, one containing the liver-specific and one containing the constitutively active regulatory cassette, which suppressed antibody formation and achieved widespread correction of GAA deficiency [33]. AAV2/8-LSPhGAApA has undergone formal pharmacology-toxicology evaluation, and this vector was found to be safe and effective in the mouse model for Pompe disease [34].

Alternatively, an ex vivo approach to gene therapy was developed with, a lentiviral vector encoding human GAA that transduced cultured hematopoietic stem cells (HSCs), which were subsequently transplanted into a Pompe disease mouse model [35]. After mild conditioning to allow engraftment, transplantation of genetically engineered HSCs resulted in stable chimerism of approximately 35% hematopoietic cells that overexpressed acid alpha-glucosidase and in major clearance of glycogen in the heart, diaphragm, spleen, and liver. Despite the need for chemotherapy to deplete the patient’s own HSCs that entails some toxicity, this approach to gene therapy was efficacious and presumably induced immune tolerance to introduced GAA.

Potential for AAV vector-mediated gene therapy to reverse neuromuscular involvement

Neuromuscular involvement remains an important aspect of Pompe disease. For instance, although ERT has prolonged survival and prevented the need for invasive ventilation in patients with infantile Pompe disease [9], neuromuscular involvement has been documented in children treated with ERT [36, 37]. The main goal of immunomodulatory gene therapy is to improve upon ERT by inducing immune tolerance to GAA, and by continuous secretion of GAA accompanied by receptor-mediated uptake in the heart, skeletal muscle, and other affected tissues. One strategy that has had an effect upon neuromuscular involvement consists of up-regulating the cation-independent mannose-6-phosphate receptor (CI-MPR). CI-MPR mediates the uptake of GAA and the trafficking of GAA to the lysosomes. The addition of a β2 agonist to increase CI-MPR expression during ERT increased the clearance of accumulated glycogen from skeletal muscles and the brain, and improved muscle function, in comparison with ERT alone [38, 39]. Similarly, adjunctive β2 agonist treatment following low dose AAV2/8-LSPhGAApA reduced the accumulated glycogen in muscle and the brain, and improved muscle function, in comparison with vector alone [40]. Furthermore, clenbuterol by itself was sufficient to significantly reduced muscle glycogen content in mice with Pompe disease through a mechanism independent of CI-MPR [41]. Finally, a pilot study of adjunctive albuterol with ERT in patients with Pompe disease increased 6MWT distance, and was well-tolerated [42]. If adjunctive therapy with β2 agonists proves to be safe and efficacious in on-going clinical trials, it will be available to enhance the efficacy of gene therapy in the future.

Clinical trial of gene therapy in Pompe disease

Gene therapy for Pompe disease has been evaluated in a clinical trial with an AAV2/1 vector encoding human GAA [43]. Direct injection of the vector into the diaphragm was evaluated in 5 pediatric patients with respiratory failure. No toxicity or T cell responses against the vector were detected, although antibody responses to both the AAV capsid proteins and to GAA were observed (the latter in a single participant). Modest improvements in volitional ventilatory performance measures were suggested. While invasive, this approach to gene therapy did target a muscle highly relevant to respiratory failure and further patient enrollment was proposed.

Alternative treatment approaches for Pompe disease

A novel approach in the form of substrate reduction therapy (by inhibiting glycogen synthesis) has been proposed as a potential adjuvant therapy for Pompe disease [44]. A phosphorodiamidate morpholino oligonucleotide (PMO) designed to invoke exon skipping and premature stop codon usage in the transcript for muscle specific glycogen synthase (Gys1) was conjugated to a cell penetrating peptide (GS-PPMO) to facilitate PMO delivery to muscle. GS-PPMO systemic administration to Pompe mice reduced glycogen synthase expression in the quadriceps, and the diaphragm, and to a lesser extent in the heart, but not the liver. These reductions correlated with significant decreases in lysosomal glycogen in the quadriceps, diaphragm, and heart of mice with Pompe disease. The lack of toxicity detected indicated that substrate reduction by GS-PPMO-mediated inhibition of muscle specific glycogen synthase might be useful in Pompe disease, and in other glycogen storage diseases involving muscle.

Progress and Challenges for Gene Therapy in GSD I

Liver and kidney involvement predominant in GSD Ia, which results from the deficiency of the catalytic subunit of glucose-6-phophosphatase-α (G6Pase) due to mutations in G6PC [45]. The importance of early diagnosis and treatment of GSD Ia is emphasized by the severe complications of untreated GSD Ia, including life threatening hypoglycemia, severe lactic acidosis, growth failure, renal Fanconi syndrome, and pancreatitis [1]. The acute complications of GSD Ia have responded favorably to dietary therapy that prevents hypoglycemia, including renal tubular dysfunction and growth failure, even though dietary therapy has significant limitations [46]. Long-term complications of GSD Ia frequently fail to respond to dietary therapy, including growth retardation, proteinuria occasionally progressing to renal failure, osteopenia, formation of hepatic adenomas and hepatocellular carcinoma, and rarely pancreatitis or pulmonary hypertension [47]. Dietary therapy consists of ingesting uncooked cornstarch at regular intervals around-the-clock, and requires constant vigilance to avoid acute metabolic decompensation accompanied by life-threatening hypoglycemia and lactic acidosis [48].

GSD Ia was reported in Maltese terriers that grew poorly in association with hypoglycemia, hepatomegaly, and nephromegaly [49]. Liver and kidney histology revealed glycogen storage, and G6Pase activity was reduced to nearly undetectable levels, confirming a diagnosis of GSD Ia. The genetic basis was a M121I missense mutation in the gene encoding G6PC [50].

Liver correction has efficacy in GSD Ia

The severe complications stemming from liver involvement in GSD Ia suggest that correction of hepatic abnormalities should be a primary goal for gene therapy [51]. As individuals with GSD Ia survive longer, complications such as increased size and number of liver adenomas and development of hepatocellular carcinoma have been observed [52]. Liver transplantation has been performed in patients with type I disease because multiple liver adenomas bear the risk of malignant transformation and/or inducing poor metabolic control. Hypoglycemia and other biochemical abnormalities were corrected after transplantation and height increased [53], confirming the efficacy of restoring G6Pase expression in the liver in GSD Ia. Glycogen breakdown was associated with increased glycolytic enzymes including glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in G6Pase (−/−) mouse liver [54]. GAPDH has been associated with non-enzymatic moonlighting activities including the induction of apoptosis [55], and apoptosis was increased in G6pase (−/−) mouse liver [54]. Hepatocellular abnormalities including increased apoptosis represent a challenge for liver-directed gene therapy in GSD Ia.

AAV vectors containing a G6Pase regulatory cassette / promoter have corrected G6Pase deficiency in the liver of animal models of GSD Ia, and these vectors contain sequence elements that regulate G6Pase expression appropriately [56, 57]. A double-stranded AAV2/8 vector containing a human G6Pase mini-gene demonstrated early efficacy in G6pase (−/−) mice and dogs with GSD Ia [57]. A single-stranded AAV vector containing a larger human G6Pase regulatory cassette also featured high efficacy in G6pase (−/−) mice, and the longer regulatory sequence featured greater activity than the G6Pase minimal promoter [58]. Other vectors that have been evaluated in the G6pase (−/−) mouse model have included helper-dependent adenovirus (Ad) vector encoding canine G6Pase [59, 60] and a lentiviral (LV) feline immunodeficiency virus vector encoding murine G6Pase that rescued neonatal G6pase (−/−) mice [61]. While both the Ad and LV vectors prolonged survival and prevented hypoglycemia in the majority of treated mice, each remains limited by significant concerns related to potential toxicity. Finally, complications of GSD Ib, caused by glucose-6-phosphate transporter deficiency, were incompletely reversed in experiments with an AAV vector, and longer-term surviving mice developed hepatocellular carcinoma related to inadequate correction [62].

Efficacy from gene therapy gradually waned in GSD Ia

AAV vector-mediated gene therapy has effectively treated GSD Ia, due to relative long-term stability of episomal vector genomes in hepatocytes [56, 57, 63, 64]. Chromosomal integration of AAV vector genomes occurs very infrequently, and consequently AAV vector genomes are gradually lost from dividing cells [65, 66]. Gradual loss of AAV vector genomes requires the eventual readministration of AAV to maintain transgene expression in the liver. However, anti-AAV antibody formation dictates that any subsequent re-administration would require delivery by a new AAV serotype [67, 68]. Re-administration will be facilitated by the fact that a number of AAV serotypes are available, several of which transduce the liver with high efficiency. Several of these serotypes have reproducibly transduced the liver in both murine and canine GSD Ia, including AAV2/8 and AAV2/9 [57, 69].

The ability to re-administer AAV vectors will be limited by the number of AAV serotypes available for clinical use. The limited ability to re-administer an AAV vector of a particular serotype is a significant hurdle given studies that demonstrated transient expression from AAV vectors in the GSD Ia liver [64, 69, 70]. Mortality from uncorrected GSD Ia has required early treatment; however, AAV vector administration to young mice accomplished a high level of liver transduction followed by declining numbers of vector genomes over the ensuing months [56, 71]. For example, an AAV2/8 vector decreased from >2 copies per liver cell at 3 weeks of age to 0.3 copies per liver cell at 26 weeks of age in G6pase (−/−) mice [56]. Similarly, an AAV2/8 vector was administered to a GSD Ia puppy at one day of age, and prevented hypoglycemia for three hours at one month of age; however, by two months of age the dog became hypoglycemic after one hour of fasting [70]. Chou and colleagues also highlighted the transience of AAV vector-mediated expression by measuring a 12-fold decline in G6Pase expression in mice from age 2 to 6 weeks [72]. A later study revealed that hypoglycemia during fasting was corrected for >18 months, but G6Pase activity in the liver declined by >90% between ages 6 and 18 months [73]. The gradual loss of efficacy from AAV vectors underlies the failure to completely reverse growth failure associated with GSD Ia in animal models [74]. These studies showed that AAV vector genomes are gradually lost over the weeks following vector administration to young animals with GSD Ia, which presents a hurdle to gene therapy for genetic diseases in general. Very recently an integrating lentiviral vector prevented the formation of hepatocellular carcinoma in mice with a liver-specific knockout of G6Pase, as well as reversing biochemical abnormalities of GSDIa, indicating that an integrating vector might have unique advantages in this disorder [75].

Potential for Gene Therapy in Other GSDs

GSD III

Mutations in the AGL gene causes genetic deficiency of glycogen debranching enzyme (GDE), resulting in accumulation of non-membrane glycogen with short outer chains in liver and muscle. Most patients have involvement of both liver and muscle (type IIIa), although some patients (~15%) have only liver involvement (type IIIb) [76]. Liver symptoms occur early in childhood; liver cirrhosis and hepatocellular carcinoma (HCC) have been reported in some cases [7779]. Progressive skeletal myopathy and cardiomyopathy are a major cause of morbidity in adults. Muscle weakness is present during childhood and becomes more predominant in the third or fourth decade of life. Sudden death caused by cardiac arrhythmias or cardiac failure has been reported [76, 8082]. Currently there is no effective treatment for the disease. Dietary interventions include control of hypoglycemia by frequent meals high in carbohydrates with cornstarch supplements or nocturnal gastric drip feedings. High protein diet is recommended for patients with myopathy [76, 83, 84]. A case report showed that a combined treatment of synthetic ketone bodies, in conjunction with a ketogenic and high-protein diet, improved cardiomyopathy in a patient with GSD III [85]. A recent study indicated that a high-fat, high-protein, and low-carbohydrate diet could be a beneficial therapeutic choice for GSD III with cardiomyopathy [86].

Animal models of GSD III

GSD III was characterized in curly coated retrievers that presented with exercise intolerance and lethargy at >12 months of age [87]. Liver transaminases, alkaline phosphatase and creatine kinase were elevated in serum by 6 months of age. Accumulations of non-membrane bound glycogen with short outer chains of α1,4-linked glucose in liver and skeletal muscle were accompanied by absence of glycogen debranching enzyme. A deletion of an adenine in exon 32 of the canine AGL gene predicted a truncation of the debranching enzyme by 126 amino acid residues [87]. Furthermore, progressive age-related liver fibrosis leading to cirrhosis was described in the CCR up to 16 months of age [88], which mimicked liver involvement in the human disorder [89]. Skeletal muscle damage caused by progressive accumulation of glycogen was observed in the affected dogs after age of 12 months [88]. Taken together, the curly coated retriever dogs represent an accurate model of GSD IIIa for improving our understanding of the disease progression and developing therapy.

Two Agl knockout mouse models were reported recently by two different groups. The first model was generated by Liu et al [90] by removing the exons after exon 5 in the Agl gene. The GDE expression was undetectable in the liver, muscle, and heart of the homozygous affected mice, which was accompanied by massive glycogen accumulation in these tissues. Reduced motor activity and muscle weakness were observed after one year of age; hepatomegaly and progressive liver fibrosis were found in the affected mice. Hypoglycemia appeared only at a young age (3 weeks) but no hyperlipidemia was observed in this model [90]. More recently, a second GSD III mouse model was generated by deletion of the carboxyl end of the glucosidase domain and the glycogen-binding domain of the GDE protein [91]. In addition to the presentation of increased glycogen accumulation in the liver, skeletal and cardiac muscles, and brain, the affected mice showed a more severe phenotype than that of the first model by Liu et al., including the decreased survival rate and appearance of muscle weakness and exercise intolerance at young age [91]. In conclusion, both mouse models represent human GSD IIIa and will be useful for therapy development for GSD III.

Challenges for gene therapy in GSD III

The human AGL gene is 85 kb in length and composed of 35 exons, encoding a 7.4-kb mRNA that includes a 4596-bp coding region and a 2371-bp 3′ untranslated sequence to express a 175 kDa GDE protein [92]. Thus, a minimum size of 5 kb GDE expression cassette (promoter + GDE coding sequence + polyA signal) needs to be packaged into a gene therapy vector for in vivo gene delivery. This >5 kb insert size exceeds the 4.7 kb AAV vector packaging limit [93] and would result in significant reduction in viral vector titer if packaged into lentiviral vectors [94]. Although the 36 kb genome of adenoviral vectors provides sufficient space for inserting large sequences, severe toxicity and strong immune responses induced by the viral vectors after systemic administration limit its application for treatment of patients with genetic diseases like GSD III [2]. Non-viral or DNA-based vectors offer no-size-limit advantage and are efficient systems for in vitro and ex vivo gene delivery, but there has been little success in achieving efficacious and safe in vivo gene transfer [2].

An AAV-based gene therapy approach that could be useful for GSD III is the hybrid-dual AAV (HD-AAV) system that was developed to expand AAV packaging capacity by splitting a large therapeutic gene into two independent vectors. High-level gene expression was achieved in mouse muscles upon systemic co-administration of the two vectors through the mechanisms of trans-splicing and homologous recombination (overlapping) [95, 96].

Alternative treatment approaches for GSD III

Enzyme replacement therapy (ERT) with GDE will not be effective for treatment of GSD III due to the lack of receptor-mediated enzyme uptake by the affected tissues. Recently we reported that recombinant human acid-alpha glucosidase (rhGAA, Alglucosidase alfa), an FDA approved therapy for Pompe disease, significantly reduced glycogen levels in primary muscles from patients with GSD IIIa [97]. Further study is needed to test the treatment efficacy of rhGAA in animal models of GSD III.

In another study daily administration of rapamycin, a specific inhibitor of mTOR, significantly reduced glycogen content in the liver and muscle, and effectively prevented liver fibrosis and muscle damage in the GSD IIIa dogs [98]. These data suggest that use of small molecule drugs like rapamycin to inhibit glycogen synthesis could be as an alternative approach for treatment of GSD III.

GSD IV

Mutations in the glycogen branching enzyme gene (GBE1) cause GSD IV which is characterized by the accumulation of amylopectin-like, poorly soluble polysaccharide deposits (polyglucosan bodies, PB) in multiple tissues including liver, skeletal and cardiac muscles, and the central nervous system (CNS). The typical presentation of hepatic form of GSD IV includes hepatomegaly and progressive liver cirrhosis that lead to death in early childhood. Four neuromuscular forms have been identified based on the age of disease onset and disease severity: perinatal form, congenital form, juvenile form, and adult form [76, 99]. Liver transplantation is the only treatment option for patients with liver cirrhosis. There are no other effective therapies available for this disease.

Animal models of GSD IV

GSD IV was described in Norwegian forest cats presented with early demise, although surviving cats appeared normal until the onset of progressive neurological decline at 5 months of age [100]. Affected kittens developed hypoglycemia, and glucose administration in the neonatal period promoted the survival of affected kittens to adulthood. Glycogen accumulations in skeletal muscle and neurons prompted the analysis of glycogen branching activity in skeletal muscle, which was severely deficient. An underlying mutation in the GBE1 gene was delineated, consisting of a 6.2 kb deletion and 332 bp insertion that altered splicing of the mRNA and decreased glycogen branching enzyme in liver and muscle [100].

The first GSD IV mouse model that recapitulated the clinical features of the severe fetal neuromuscular form of GSD-IV was reported by Lee et al [101]. The homozygous affected mice carry a stop codon mutation (E609X) in the GBE1 gene resulted in GBE ablation that led to poor ventricular function in late gestation, and ultimately caused heart failure, fetal hydrops and embryonic lethality [101]. Soon after this report, Akman et al [102] reported another two mouse models of GSD IV. Homozygous deletion of exon 7 of the Gbe1 gene in the Gbe1−/− mice completely eliminated GBE activity in tissues, leading to significant in utero PB accumulation and death at birth [102]. In contrast, the mice homozygous for the insertion of a phosphoglycerate kinase-Neomycin cassette within intron 7 of the Gbe1 gene (Gbe1neo/neo) exhibit a phenotype similar to juvenile neuromuscular form GSD IV, with reduced GBE enzyme activity and accumulation of PG in the liver, skeletal muscle, heart, and brain. The Gbe1neo/neo mice died between 3 to 9 months of age [102].

Therapeutic approaches for GSD IV

The formation of PB is the hallmark of GSD IV. The critical determinant of PB formation is an increase in the ratio of glycogen synthase (GS) to GBE, which leads to uncontrolled elongation of α-1,4-glycosidic glycogen chains without adequate α-1,6-glycosidic branching activity [103105]. Thus, restoration of GBE activity or inhibition of GS expression will be two potential treatment approaches for correction of the abnormal GS/GBE ratio and prevention of PB formation in GSD IV. Increased expression of GBE could be achieved by traditional gene therapy approach by delivery of a GBE expression cassette. The 2.1-kb coding sequence of human GBE1 gene could be easily packaged into an AAV vector. Widespread GBE expression could be achieved by systemic administration of an AAV2/9 vector, the most efficient AAV serotype for whole-body gene transfer, into GSD IV mice or cats, to test the treatment efficacy [102, 106109]. Kakhlon et al demonstrated that inactivation of GS by rapamycin prevented PB formation in GBE-knocked down neurons derived from E18 rats [110]. Other approaches, such as antisense oligonucleotides, AAV-mediated shRNA, or screening of small molecule drugs, should be considered for inhibition of GS expression and prevent of PB formation in GSD IV. It should be noted that both treatment approaches, by increasing GBE expression and by inhibiting GS activity, are preventive therapies that require starting treatment at young age to achieve best outcomes. To clear the existing PB in the affected tissues at advanced stage of GSD IV disease, a glycogen-degrading enzyme will be needed.

GSD V

GSD V, also known as McArdle disease, is frequently detected in the second to third decade of life by exercise intolerance with muscle cramping accompanied by elevated serum creatine kinase [1]. This is due to a mutation in the muscle phosphorylase gene (PYGM) most commonly associated with a change from an arginine to a stop codon in exon 1 (p.R50X) [111]. There is no effective treatment for this disease, but many patients are able to perform moderate, sustained exercise on a carbohydrate-rich diet with carbohydrate ingestion shortly before exercise [112].

Animal models of GSD V

There are currently 3 known GSD V animal models. The first was discovered in Charlois cattle with a C to T substitution in codon 489 leading to an arginine to tryptophan substitution in the myophosphorylase gene [113]. Affected cattle display signs of rhabdomyolysis including exercise intolerance and myoglobinuria [114]. A second large animal GSD V model was discovered in a flock of Merino sheep exhibiting exercise intolerance [115]. Affected sheep have a splice site mutation at the 3′ end of intron 19 of the myophosphorylase gene leading to disruption in the reading frame and premature truncation of the myophosphorylase protein. Upon biopsy, an increase of muscle glycogen and absence of muscle glycogen phosphorylase was discovered [115]. A mouse model was recently created using a knock-in for the common human mutation, p.R50X in the PYGM gene encoding for myophosphorylase. This mouse model demonstrates many features of GSD V including elevated creatine kinase serum concentrations, increased muscle glycogen, absence of myophosphorylase protein and activity in muscles and poor exercise performance [116].

Therapeutic approaches for GSD V

To date, only the ovine GSD V model has been used to test novel therapeutics using gene therapy and pharmaceuticals. Adenovirus 5 (Ad5) and AAV vectors expressing myophosphorylase were both used with some efficacy in sheep with GSD V. Different promoters were also utilized, namely, an Ad5 vector was evaluated containing either Rous Sarcoma virus (RSV) or Cytomegalovirus (CMV) promoters, and an AAV vector was evaluated containing a CMV promoter. All three vectors effectively transduced sheep muscle when injected intramuscularly in young sheep, but the Ad5 vector containing the CMV promoter featured the greatest sustained transduction. However, myophosphorylase activity waned over time in all cases (Howell et al 2008). The same study also demonstrated that damage to muscle fibers caused by injection with vectors, including a positive control vector expressing LacZ that caused re-expression of non-muscle isoforms of glycogen phosphorylase. Further studies have been performed using pharmaceuticals such as valproate and notexin to cause similar muscle damage and thus re-expression of non-muscle isoforms of myophosphorylase in the GSD V ovine model [117, 118].

GSD VII

GSD VII is inherited as an autosomal recessive trait characterized by a deficiency of the muscle isoenzyme of phosphofructokinase (PFKM). There are several mutations in the PFKM gene found on chromosome 12 that have been described to cause this disease in patients, but there have been no specific genotype-phenotype correlations found to date [119]. The typical clinical presentation of the classic form of GSD VII consists of muscle cramps, exercise intolerance, myoglobinuria, hyperuricemia and hemolytic anemia. The anemia occurs because erythrocytes express both PFKM and the liver isoenzyme of PFK (PFKL); PFKL accounts for about 50% of normal PFK activity in erythrocytes [1]. Other presentations of GSD VII include a severe infantile form leading to early death in childhood, a late onset form and a hemolytic form that lacks muscle symptoms [119]. There is no specific treatments for this disease, and therapy remains mainly palliative,

Animal models of GSD VII

An animal model for GSD VII was reported as an autosomal recessive trait demonstrated in three families of English Springer spaniels that had hemolytic crises and rhabdomyolysis during exercise in association with PFKM deficiency [120, 121]. The mutation in the PFKM gene was demonstrated as a nonsense mutation that caused premature termination [122]. Other dog breeds have been reported to have the same mutation including American Cocker spaniels and whippets. [123, 124]. More recently, PFK deficiency was described in Wachtelhunds exhibiting exercise intolerance, hemolytic anemia and pigmenturia, but apparently is not caused by the same mutation as that seen in other breeds [125]. These large animal models of PFK deficiency provide an opportunity to develop muscle-directed gene therapy for GSD VII, but have not been used in any specific therapeutic studies.

Two mouse models have been reported with PFKM deficiency, the first was developed by lexicon laboratories and has demonstrated an impairment in insulin secretion and decrease in fat stores [126] [127]. The second model is a Pfkm(−/−) and displays a severe cardiac and hematologic disorder accompanied by skeletal muscle symptoms similar to PFK human patients [128]. Neither has been developed for gene therapy models to the authors’ knowledge.

Therapeutic approaches for GSD VII

Based on findings by Bruser et al 2012, pharmacologic agents that block the inhibitory allosteric binding site of muscle 6-phosphofructokinase to ADP may be therapeutic, but have yet to be tested in animal models [129].

Conclusion

Gene therapy can be efficacious in GSD, if the transgene stably expresses sufficient quantities of the therapeutic enzyme in involved tissues. Small molecule therapies show promise as both palliative and adjunctive therapies for specific GSDs. The development of new therapies will benefit affected human patients, if pharmacology-toxicology studies and clinical trials replicate these proof-of-principle experiments.

Acknowledgments

GSD research at Duke University has been supported by the Children’s Fund for GSD Research, Children’s Miracle Network, the Association for Glycogen Storage Disease, the Alice and YT Chen Pediatrics Genetics and Genomics Research Center, Genzyme Corporation, the Muscular Dystrophy Association, and the National Institutes of Health. DDK was supported by NIH Grant R01AR065873 from the National Institute of Arthritis and Musculoskeletal and Skin Disorders.

References

  • 1.Chen YT. Glycogen Storage Diseases. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The Metabolic and Molecular Bases of Inherited Disease. 8. New York: McGraw-Hill; 2001. pp. 1521–51. [Google Scholar]
  • 2.Kay MA. State-of-the-art gene-based therapies: the road ahead. Nat Rev Genet. 2011 May;12(5):316–28. doi: 10.1038/nrg2971. [DOI] [PubMed] [Google Scholar]
  • 3.Ponce E, Witte DP, Hirschhorn R, Huie ML, Grabowski GA. Murine Acid alpha-Glucosidase : Cell-Specific mRNA Differential Expression during Development and Maturation. Am J Pathol. 1999 Apr;154(4):1089–96. doi: 10.1016/s0002-9440(10)65361-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hirschhorn R, Reuser AJJ. Glycogen Storage Disease Type II: Acid à-Glucosidase (Acid Maltase) Deficiency. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The Metabolic and Molecular Basis for Inherited Disease. 8. New York: McGraw-Hill; 2001. pp. 3389–419. [Google Scholar]
  • 5.Kishnani PS, Hwu WL, Mandel H, Nicolino M, Yong F, Corzo D. A retrospective, multinational, multicenter study on the natural history of infantile-onset Pompe disease. J Pediatr. 2006;148(5):671–6. doi: 10.1016/j.jpeds.2005.11.033. [DOI] [PubMed] [Google Scholar]
  • 6.Walvoort HC. Glycogen storage disease type II in the Lapland dog. Vet Q. 1985;7(3):187–90. doi: 10.1080/01652176.1985.9693981. [DOI] [PubMed] [Google Scholar]
  • 7.Seppala EH, Reuser AJ, Lohi H. A nonsense mutation in the acid alpha-glucosidase gene causes Pompe disease in Finnish and Swedish Lapphunds. PLoS One. 2013;8(2):e56825. doi: 10.1371/journal.pone.0056825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Amalfitano A, Bengur AR, Morse RP, et al. Recombinant human acid à-glucosidase enzyme therapy for infantile glycogen storage disease type II: Results of a phase I/II clinical trial. Genet Med. 2001 May;3(2):132–8. doi: 10.109700125817-200103000-00007. [DOI] [PubMed] [Google Scholar]
  • 9.Kishnani PS, Corzo D, Nicolino M, et al. Recombinant human acid {alpha}-glucosidase: Major clinical benefits in infantile-onset Pompe disease. Neurology. 2007;68(2):99–109. doi: 10.1212/01.wnl.0000251268.41188.04. [DOI] [PubMed] [Google Scholar]
  • 10.Kishnani PS, Goldenberg PC, DeArmey SL, et al. Cross-reactive immunologic material status affects treatment outcomes in Pompe disease infants. Mol Genet Metab. 2010 Jan;99(1):26–33. doi: 10.1016/j.ymgme.2009.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Desnick RJ. Enzyme replacement and enhancement therapies for lysosomal diseases. J Inh Metab Dis. 2004;27(3):385–410. doi: 10.1023/B:BOLI.0000031101.12838.c6. [DOI] [PubMed] [Google Scholar]
  • 12.Banugaria SG, Prater SN, Ng YK, et al. The impact of antibodies on clinical outcomes in diseases treated with therapeutic protein: lessons learned from infantile Pompe disease. Genet Med. 2011 Aug;13(8):729–36. doi: 10.1097/GIM.0b013e3182174703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mendelsohn NJ, Messinger YH, Rosenberg AS, Kishnani PS. Elimination of antibodies to recombinant enzyme in Pompe’s disease. N Engl J Med. 2009 Jan 8;360(2):194–5. doi: 10.1056/NEJMc0806809. [DOI] [PubMed] [Google Scholar]
  • 14.Koeberl DD, Kishnani PS. Immunomodulatory gene therapy in lysosomal storage disorders. Curr Gene Ther. 2009 Dec;9(6):503–10. doi: 10.2174/156652309790031094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gao GP, Alvira MR, Wang L, Calcedo R, Johnston J, Wilson JM. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Nat Acad Sci USA. 2002;99(18):11854–9. doi: 10.1073/pnas.182412299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Asokan A, Schaffer DV, Samulski RJ. The AAV vector toolkit: poised at the clinical crossroads. Mol Ther. 2012 Apr;20(4):699–708. doi: 10.1038/mt.2011.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Vandenberghe LH, Wang L, Somanathan S, et al. Heparin binding directs activation of T cells against adeno-associated virus serotype 2 capsid. Nat Med. 2006;12(8):967–71. doi: 10.1038/nm1445. [DOI] [PubMed] [Google Scholar]
  • 18.Thomas CE, Storm TA, Huang Z, Kay MA. Rapid uncoating of vector genomes is the key to efficient liver transduction with pseudotyped adeno-associated virus vectors. J Virol. 2004;78(6):3110–22. doi: 10.1128/JVI.78.6.3110-3122.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nakai H, Fuess S, Storm TA, Muramatsu S, Nara Y, Kay MA. Unrestricted hepatocyte transduction with adeno-associated virus serotype 8 vectors in mice. J Virol. 2005;79(1):214–24. doi: 10.1128/JVI.79.1.214-224.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sun B, Zhang H, Franco LM, et al. Efficacy of an adeno-associated virus 8-pseudotyped vector in glycogen storage disease type II. Mol Ther. 2005;11(1):57–65. doi: 10.1016/j.ymthe.2004.10.004. [DOI] [PubMed] [Google Scholar]
  • 21.Sun B, Zhang H, Franco LM, et al. Correction of glycogen storage disease type II by an adeno-associated virus vector containing a muscle-specific promoter. Mol Ther. 2005;11(6):889–98. doi: 10.1016/j.ymthe.2005.01.012. [DOI] [PubMed] [Google Scholar]
  • 22.Franco LM, Sun B, Yang X, et al. Evasion of immune responses to introduced human acid alpha-glucosidase by liver-restricted expression in glycogen storage disease type II. Mol Ther. 2005;12(5):876–84. doi: 10.1016/j.ymthe.2005.04.024. [DOI] [PubMed] [Google Scholar]
  • 23.Fraites TJ, Schleissing MR, Shanely RA, et al. Correction of the enzymatic and functional deficits in a model of pompe disease using adeno-associated virus vectors. Mol Ther. 2002 May;5(5):571–8. doi: 10.1006/mthe.2002.0580. [DOI] [PubMed] [Google Scholar]
  • 24.Mah C, Pacak CA, Cresawn KO, et al. Physiological Correction of Pompe Disease by Systemic Delivery of Adeno-associated Virus Serotype 1 Vectors. Mol Ther. 2007;15(3):501–7. doi: 10.1038/sj.mt.6300100. [DOI] [PubMed] [Google Scholar]
  • 25.Cresawn KO, Fraites TJ, Wasserfall C, et al. Impact of humoral immune response on distribution and efficacy of recombinant adeno-associated virus-derived acid alpha-glucosidase in a model of glycogen storage disease type II. Hum Gene Ther. 2005;16(1):68–80. doi: 10.1089/hum.2005.16.68. [DOI] [PubMed] [Google Scholar]
  • 26.Sun B, Young SP, Li P, et al. Correction of multiple striated muscles in murine Pompe disease through adeno-associated virus-mediated gene therapy. Mol Ther. 2008;16(8):1366–71. doi: 10.1038/mt.2008.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Sun B, Li S, Bird A, et al. Antibody formation and mannose-6-phosphate receptor expression impact the efficacy of muscle-specific transgene expression in murine Pompe disease. J Gene Med. 2010 Nov;12(11):881–91. doi: 10.1002/jgm.1511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Han SO, Li S, Brooks ED, et al. Enhanced efficacy from gene therapy in Pompe disease using coreceptor blockade. Hum Gene Ther. 2015 Jan;26(1):26–35. doi: 10.1089/hum.2014.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Amalfitano A, McVie-Wylie AJ, Hu H, et al. Systemic correction of the muscle disorder glycogen storage disease type II after hepatic targeting of a modified adenovirus vector encoding human acid-alpha-glucosidase. Proc Natl Acad Sci USA. 1999;96(16):8861–6. doi: 10.1073/pnas.96.16.8861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pauly DF, Fraites TJ, Toma C, et al. Intercellular transfer of the virally derived precursor form of acid alpha-glucosidase corrects the enzyme deficiency in inherited cardioskeletal myopathy Pompe disease. Hum Gene Ther. 2001;12(5):527–38. doi: 10.1089/104303401300042447. [DOI] [PubMed] [Google Scholar]
  • 31.Zhang P, Luo X, Bird A, Li S, Koeberl DD. Deficiency in MyD88 signaling results in decreased antibody responses to an adeno-associated virus vector in murine Pompe disease. Biores Open Acc. 2012;1(3):109–14. doi: 10.1089/biores.2012.0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sun B, Bird A, Young SP, Kishnani PS, Chen YT, Koeberl DD. Enhanced response to enzyme replacement therapy in pompe disease after the induction of immune tolerance. Am J Hum Genet. 2007;81(5):1042–9. doi: 10.1086/522236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhang P, Sun B, Osada T, et al. Immunodominant liver-specific expression suppresses transgene-directed immune responses in murine pompe disease. Hum Gene Ther. 2012 May;23(5):460–72. doi: 10.1089/hum.2011.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wang G, Young SP, Bali D, et al. Assessment of toxicity and biodistribution of recombinant AAV2/8 vector-mediated immunomodulatory gene therapy in mice with Pompe disease. Mol Ther Meth Clin Dev. 2014;1:14018–27. doi: 10.1038/mtm.2014.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.van Til NP, Stok M, Aerts Kaya FS, et al. Lentiviral gene therapy of murine hematopoietic stem cells ameliorates the Pompe disease phenotype. Blood. 2010 Jul;115(26):5329–37. doi: 10.1182/blood-2009-11-252874. [DOI] [PubMed] [Google Scholar]
  • 36.Nicolino M, Byrne B, Wraith JE, et al. Clinical outcomes after long-term treatment with alglucosidase alfa in infants and children with advanced Pompe disease. Genet Med. 2009 Mar;11(3):210–9. doi: 10.1097/GIM.0b013e31819d0996. [DOI] [PubMed] [Google Scholar]
  • 37.Prater SN, Banugaria SG, Dearmey SM, et al. The emerging phenotype of long-term survivors with infantile Pompe disease. Genet Med. 2012 Sep;14(9):800–10. doi: 10.1038/gim.2012.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Koeberl DD, Luo X, Sun B, et al. Enhanced efficacy of enzyme replacement therapy in Pompe disease through mannose-6-phosphate receptor expression in skeletal muscle. Mol Genet Metab. 2011 Jun;103(2):107–12. doi: 10.1016/j.ymgme.2011.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Koeberl DD, Li S, Dai J, Thurberg BL, Bali D, Kishnani PS. beta2 Agonists enhance the efficacy of simultaneous enzyme replacement therapy in murine Pompe disease. Mol Genet Metab. 2012 Feb;105(2):221–7. doi: 10.1016/j.ymgme.2011.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Li S, Sun B, Nilsson MI, et al. Adjunctive beta2-agonists reverse neuromuscular involvement in murine Pompe disease. FASEB J. 2013 Jan;27(1):34–44. doi: 10.1096/fj.12-207472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Farah BL, Madden L, Li S, et al. Adjunctive beta2-agonist treatment reduces glycogen independently of receptor-mediated acid alpha-glucosidase uptake in the limb muscles of mice with Pompe disease. FASEB J. 2014 May;28(5):2272–80. doi: 10.1096/fj.13-244202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Koeberl DD, Austin S, Case LE, et al. Adjunctive albuterol enhances the response to enzyme replacement therapy in late-onset Pompe disease. FASEB J. 2014 May;28(5):2171–6. doi: 10.1096/fj.13-241893. [DOI] [PubMed] [Google Scholar]
  • 43.Smith BK, Collins SW, Conlon TJ, et al. Phase I/II trial of adeno-associated virus-mediated alpha-glucosidase gene therapy to the diaphragm for chronic respiratory failure in Pompe disease: initial safety and ventilatory outcomes. Hum Gene Ther. 2013 Jun;24(6):630–40. doi: 10.1089/hum.2012.250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.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 Nucl Ac. 2014;3:e206. doi: 10.1038/mtna.2014.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Froissart R, Piraud M, Boudjemline AM, et al. Glucose-6-phosphatase deficiency. Orphanet J Rare Dis. 2011;6:27. doi: 10.1186/1750-1172-6-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chen YT, Cornblath M, Sidbury JB. Cornstarch therapy in type I glycogen-storage disease. New Engl J Med. 1984;310(3):171–5. doi: 10.1056/NEJM198401193100306. [DOI] [PubMed] [Google Scholar]
  • 47.Koeberl DD, Kishnani PS, Bali D, Chen YT. Emerging therapies for glycogen storage disease type I. Trends Endocrinol Metab. 2009;20(5):252–8. doi: 10.1016/j.tem.2009.02.003. [DOI] [PubMed] [Google Scholar]
  • 48.Wolfsdorf JI, Crigler JF., Jr Effect of continuous glucose therapy begun in infancy on the long-term clinical course of patients with type I glycogen storage disease. J Pediatr Gastroenterol Nutr. 1999;29(2):136–43. doi: 10.1097/00005176-199908000-00008. [DOI] [PubMed] [Google Scholar]
  • 49.Brix AE, Howerth EW, McConkie-Rosell A, et al. Glycogen storage disease type Ia in two littermate Maltese puppies. Vet Pathol. 1995;32(5):460–5. doi: 10.1177/030098589503200502. [DOI] [PubMed] [Google Scholar]
  • 50.Kishnani PS, Bao Y, Wu JY, Brix AE, Lin JL, Chen YT. Isolation and nucleotide sequence of canine glucose-6-phosphatase mRNA: identification of mutation in puppies with glycogen storage disease type Ia. Biochem Mol Med. 1997;61(2):168–77. doi: 10.1006/bmme.1997.2600. [DOI] [PubMed] [Google Scholar]
  • 51.Chou JY, Mansfield BC. Gene therapy for type I glycogen storage diseases. Curr Gene Ther. 2007;7(2):79–88. doi: 10.2174/156652307780363152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Franco LM, Krishnamurthy V, Bali D, et al. Hepatocellular carcinoma in glycogen storage disease type Ia: A case series. J Inh Metab Dis. 2005;28(2):153–62. doi: 10.1007/s10545-005-7500-2. [DOI] [PubMed] [Google Scholar]
  • 53.Matern D, Starzl TE, Arnaout W, et al. Liver transplantation for glycogen storage disease types I, III, and IV. Eur J Pediatr. 1999;158(Suppl 2):S43–S8. doi: 10.1007/pl00014320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sun B, Li S, Yang L, et al. Activation of glycolysis and apoptosis in glycogen storage disease type Ia. Mol Genet Metab. 2009 Aug;97(4):267–71. doi: 10.1016/j.ymgme.2009.04.003. [DOI] [PubMed] [Google Scholar]
  • 55.Sriram G, Martinez JA, McCabe ER, Liao JC, Dipple KM. Single-gene disorders: what role could moonlighting enzymes play? Am J Hum Genet. 2005;76(6):911–24. doi: 10.1086/430799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Koeberl DD, Sun BD, Damodaran TV, et al. Early, sustained efficacy of adeno-associated virus vector-mediated gene therapy in glycogen storage disease type Ia. Gene Ther. 2006;13(17):1281–9. doi: 10.1038/sj.gt.3302774. [DOI] [PubMed] [Google Scholar]
  • 57.Koeberl DD, Pinto C, Sun B, et al. AAV vector-mediated reversal of hypoglycemia in canine and murine glycogen storage disease type Ia. Mol Ther. 2008;16(4):665–72. doi: 10.1038/mt.2008.15. [DOI] [PubMed] [Google Scholar]
  • 58.Lee YM, Pan CJ, Koeberl DD, Mansfield BC, Chou JY. The upstream enhancer elements of the G6PC promoter are critical for optimal G6PC expression in murine glycogen storage disease type Ia. Mol Genet Metab. 2013 Nov;110(3):275–80. doi: 10.1016/j.ymgme.2013.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Koeberl DD, Sun B, Bird A, Chen Y, Oka K, Chan L. Efficacy of helper-dependent adenovirus vector-mediated gene therapy in murine glycogen storage disease type Ia. Mol Ther. 2007;15(7):1253–8. doi: 10.1038/sj.mt.6300188. [DOI] [PubMed] [Google Scholar]
  • 60.Crane B, Luo X, Demaster A, et al. Rescue administration of a helper-dependent adenovirus vector with long-term efficacy in dogs with glycogen storage disease type Ia. Gene Ther. 2012 Apr;19(4):443–52. doi: 10.1038/gt.2011.86. [DOI] [PubMed] [Google Scholar]
  • 61.Grinshpun A, Condiotti R, SNW, et al. Neonatal Gene Therapy of Glycogen Storage Disease Type Ia Using a Feline Immunodeficiency Virus-based Vector. Mol Ther. 2010 Jun 22; doi: 10.1038/mt.2010.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yiu WH, Pan CJ, Mead PA, Starost MF, Mansfield BC, Chou JY. Normoglycemia alone is insufficient to prevent long-term complications of hepatocellular adenoma in glycogen storage disease type Ib mice. J Hepatol. 2009 Nov;51(5):909–17. doi: 10.1016/j.jhep.2008.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Yiu WH, Mead PA, Jun HS, Mansfield BC, Chou JY. Oxidative stress mediates nephropathy in type Ia glycogen storage disease. Lab Invest. 2010 Apr;90(4):620–9. doi: 10.1038/labinvest.2010.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Luo X, Hall G, Li S, et al. Hepatorenal correction in murine glycogen storage disease type I with a double-stranded adeno-associated virus vector. Mol Ther. 2011 Nov;19(11):1961–70. doi: 10.1038/mt.2011.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.McCarty DM, Young SM, Jr, Samulski RJ. Integration of adeno-associated virus (AAV) and recombinant AAV vectors. Annu Rev Genet. 2004;38:819–45. doi: 10.1146/annurev.genet.37.110801.143717. [DOI] [PubMed] [Google Scholar]
  • 66.Schnepp BC, Clark KR, Klemanski DL, Pacak CA, Johnson PR. Genetic fate of recombinant adeno-associated virus vector genomes in muscle. J Virol. 2003 Mar;77(6):3495–504. doi: 10.1128/JVI.77.6.3495-3504.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Halbert CL, Standaert TA, Aitken ML, Alexander IE, Russell DW, Miller AD. Transduction by adeno-associated virus vectors in the rabbit airway: efficiency, persistence, and readministration. J Virol. 1997;71(8):5932–41. doi: 10.1128/jvi.71.8.5932-5941.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Halbert CL, Rutledge EA, Allen JM, Russell DW, Miller AD. Repeat transduction in the mouse lung by using adeno-associated virus vectors with different serotypes. J Virol. 2000;74(3):1524–32. doi: 10.1128/jvi.74.3.1524-1532.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Demaster A, Luo X, Curtis S, et al. Long-term efficacy following readministration of an adeno-associated virus vector in dogs with glycogen storage disease type Ia. Hum Gene Ther. 2012 Apr;23(4):407–18. doi: 10.1089/hum.2011.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Weinstein DA, Correia CE, Conlon T, et al. Adeno-associated virus-mediated correction of a canine model of glycogen storage disease type ia. Hum Gene Ther. 2010 Jul;21(7):903–10. doi: 10.1089/hum.2009.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Cunningham SC, Spinoulas A, Carpenter KH, Wilcken B, Kuchel PW, Alexander IE. AAV2/8-mediated Correction of OTC Deficiency Is Robust in Adult but Not Neonatal Spf(ash) Mice. Mol Ther. 2009 Aug;17(8):1340–6. doi: 10.1038/mt.2009.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yiu WH, Lee YM, Peng WT, et al. Complete normalization of hepatic G6PC deficiency in murine glycogen storage disease type Ia using gene therapy. Mol Ther. 2010 Jun;18(6):1076–84. doi: 10.1038/mt.2010.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lee YM, Jun HS, Pan CJ, et al. Prevention of hepatocellular adenoma and correction of metabolic abnormalities in murine glycogen storage disease type Ia by gene therapy. Hepatology. 2012 Nov;56(5):1719–29. doi: 10.1002/hep.25717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Brooks ED, Little D, Arumugam R, et al. Pathogenesis of growth failure and partial reversal with gene therapy in murine and canine Glycogen Storage Disease type Ia. Mol Genet Metab. 2013 Jun;109(2):161–70. doi: 10.1016/j.ymgme.2013.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Clar J, Mutel E, Gri B, et al. Hepatic lentiviral gene transfer prevents the long-term onset of hepatic tumours of glycogen storage disease type 1a in mice. Hum Mol Genet. 2015 Apr 15;24(8):2287–96. doi: 10.1093/hmg/ddu746. [DOI] [PubMed] [Google Scholar]
  • 76.Chen YT, Kishnani PS, Koeberl DD. Glycogen storage diseases. New York: McGraw-Hill; 2009. [Google Scholar]
  • 77.Demo E, Frush D, Gottfried M, et al. Glycogen storage disease type III-hepatocellular carcinoma a long-term complication? J Hepatol. 2007 Mar;46(3):492–8. doi: 10.1016/j.jhep.2006.09.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Siciliano M, De Candia E, Ballarin S, et al. Hepatocellular carcinoma complicating liver cirrhosis in type IIIa glycogen storage disease. J Clin Gastroenterol. 2000 Jul;31(1):80–2. doi: 10.1097/00004836-200007000-00020. [DOI] [PubMed] [Google Scholar]
  • 79.Labrune P, Trioche P, Duvaltier I, Chevalier P, Odievre M. Hepatocellular adenomas in glycogen storage disease type I and III: a series of 43 patients and review of the literature. J Pediatr Gastroenterol Nutr. 1997 Mar;24(3):276–9. doi: 10.1097/00005176-199703000-00008. [DOI] [PubMed] [Google Scholar]
  • 80.Lee PJ, Deanfield JE, Burch M, Baig K, McKenna WJ, Leonard JV. Comparison of the functional significance of left ventricular hypertrophy in hypertrophic cardiomyopathy and glycogenosis type III. Am J Cardiol. 1997 Mar 15;79(6):834–8. doi: 10.1016/s0002-9149(96)00885-5. [DOI] [PubMed] [Google Scholar]
  • 81.Lucchiari S, Pagliarani S, Salani S, et al. Hepatic and neuromuscular forms of glycogenosis type III: nine mutations in AGL. Hum Mut. 2006 Jun;27(6):600–1. doi: 10.1002/humu.9426. [DOI] [PubMed] [Google Scholar]
  • 82.Coleman RA, Winter HS, Wolf B, Gilchrist JM, Chen YT. Glycogen storage disease type III (glycogen debranching enzyme deficiency): correlation of biochemical defects with myopathy and cardiomyopathy. Ann nt Med. 1992 Jun 1;116(11):896–900. doi: 10.7326/0003-4819-116-11-896. [DOI] [PubMed] [Google Scholar]
  • 83.Kishnani PS, Austin SL, Arn P, et al. Glycogen storage disease type III diagnosis and management guidelines. Genet Med. 2010 Jul;12(7):446–63. doi: 10.1097/GIM.0b013e3181e655b6. [DOI] [PubMed] [Google Scholar]
  • 84.Derks TG, Smit GP. Dietary management in glycogen storage disease type III: what is the evidence? J Inherit Metab Dis. 2015 May;38(3):545–50. doi: 10.1007/s10545-014-9756-x. [DOI] [PubMed] [Google Scholar]
  • 85.Valayannopoulos V, Bajolle F, Arnoux JB, et al. Successful treatment of severe cardiomyopathy in glycogen storage disease type III with D,L-3-hydroxybutyrate, ketogenic and high protein diet. Pediatr Res. 2011 Dec;70(6):638–41. doi: 10.1203/PDR.0b013e318232154f. [DOI] [PubMed] [Google Scholar]
  • 86.Brambilla A, Mannarino S, Pretese R, Gasperini S, Galimberti C, Parini R. Improvement of Cardiomyopathy After High-Fat Diet in Two Siblings with Glycogen Storage Disease Type III. JIMD Rep. 2014 Oct 17;:91–5. doi: 10.1007/8904_2014_343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gregory BL, Shelton GD, Bali DS, Chen YT, Fyfe JC. Glycogen storage disease type IIIa in curly-coated retrievers. J Vet Intern Med. 2007;21(1):40–6. doi: 10.1892/0891-6640(2007)21[40:gsdtii]2.0.co;2. [DOI] [PubMed] [Google Scholar]
  • 88.Yi H, Thurberg BL, Curtis S, et al. Characterization of a canine model of glycogen storage disease type IIIa. Dis Model Mech. 2012 Nov;5(6):804–11. doi: 10.1242/dmm.009712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Markowitz AJ, Chen YT, Muenzer J, Delbuono EA, Lucey MR. A man with type III glycogenosis associated with cirrhosis and portal hypertension. Gastroenterology. 1993;105(6):1882–5. doi: 10.1016/0016-5085(93)91088-y. [DOI] [PubMed] [Google Scholar]
  • 90.Liu KM, Wu JY, Chen YT. Mouse model of glycogen storage disease type III. MolGenMetab. 2014 Apr;111(4):467–76. doi: 10.1016/j.ymgme.2014.02.005. [DOI] [PubMed] [Google Scholar]
  • 91.Pagliarani S, Lucchiari S, Ulzi G, et al. Glycogen storage disease type III: A novel Agl knockout mouse model. Biochim Biophys Acta. 2014 Nov;1842(11):2318–28. doi: 10.1016/j.bbadis.2014.07.029. [DOI] [PubMed] [Google Scholar]
  • 92.Bao Y, Dawson TL, Jr, Chen YT. Human glycogen debranching enzyme gene (AGL): complete structural organization and characterization of the 5′ flanking region. Genomics. 1996 Dec 1;38(2):155–65. doi: 10.1006/geno.1996.0611. [DOI] [PubMed] [Google Scholar]
  • 93.Wu Z, Yang H, Colosi P. Effect of genome size on AAV vector packaging. Mol Ther. 2010 Jan;18(1):80–6. doi: 10.1038/mt.2009.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kumar M, Keller B, Makalou N, Sutton RE. Systematic determination of the packaging limit of lentiviral vectors. Hum Gene Ther. 2001 Oct 10;12(15):1893–905. doi: 10.1089/104303401753153947. [DOI] [PubMed] [Google Scholar]
  • 95.Ghosh A, Yue Y, Lai Y, Duan D. A hybrid vector system expands adeno-associated viral vector packaging capacity in a transgene-independent manner. Mol Ther. 2008 Jan;16(1):124–30. doi: 10.1038/sj.mt.6300322. [DOI] [PubMed] [Google Scholar]
  • 96.Ghosh A, Yue Y, Duan D. Efficient transgene reconstitution with hybrid dual AAV vectors carrying the minimized bridging sequences. Hum Gene Ther. 2011 Jan;22(1):77–83. doi: 10.1089/hum.2010.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Sun B, Fredrickson K, Austin S, et al. Alglucosidase alfa enzyme replacement therapy as a therapeutic approach for glycogen storage disease type III. Mol Gen Metab. 2013 Feb;108(2):145–7. doi: 10.1016/j.ymgme.2012.12.002. [DOI] [PubMed] [Google Scholar]
  • 98.Yi H, Brooks ED, Thurberg BL, Fyfe JC, Kishnani PS, Sun B. Correction of glycogen storage disease type III with rapamycin in a canine model. J Mol Med (Berl) 2014 Jun;92(6):641–50. doi: 10.1007/s00109-014-1127-4. [DOI] [PubMed] [Google Scholar]
  • 99.Paradas C, Akman HO, Ionete C, et al. Branching enzyme deficiency: expanding the clinical spectrum. JAMA Neurol. 2014 Jan;71(1):41–7. doi: 10.1001/jamaneurol.2013.4888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Fyfe JC, Kurzhals RL, Hawkins MG, et al. A complex rearrangement in GBE1 causes both perinatal hypoglycemic collapse and late-juvenile-onset neuromuscular degeneration in glycogen storage disease type IV of Norwegian forest cats. Mol Genet Metab. 2007;90(4):383–92. doi: 10.1016/j.ymgme.2006.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Lee YC, Chang CJ, Bali D, Chen YT, Yan YT. Glycogen-branching enzyme deficiency leads to abnormal cardiac development: novel insights into glycogen storage disease IV. Hum Mol Genet. 2011 Feb 1;20(3):455–65. doi: 10.1093/hmg/ddq492. [DOI] [PubMed] [Google Scholar]
  • 102.Akman HO, Sheiko T, Tay SK, Finegold MJ, Dimauro S, Craigen WJ. Generation of a novel mouse model that recapitulates early and adult onset glycogenosis type IV. Hum Mol Genet. 2011 Nov 15;20(22):4430–9. doi: 10.1093/hmg/ddr371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Raben N, Danon M, Lu N, et al. Surprises of genetic engineering: a possible model of polyglucosan body disease. Neurology. 2001 Jun 26;56(12):1739–45. doi: 10.1212/wnl.56.12.1739. [DOI] [PubMed] [Google Scholar]
  • 104.Pederson BA, Csitkovits AG, Simon R, et al. Overexpression of glycogen synthase in mouse muscle results in less branched glycogen. Bioch Biophys Res Commun. 2003 Jun 13;305(4):826–30. doi: 10.1016/s0006-291x(03)00862-3. [DOI] [PubMed] [Google Scholar]
  • 105.Lossos A, Barash V, Soffer D, et al. Hereditary branching enzyme dysfunction in adult polyglucosan body disease: a possible metabolic cause in two patients. Ann Neurol. 1991 Nov;30(5):655–62. doi: 10.1002/ana.410300505. [DOI] [PubMed] [Google Scholar]
  • 106.Samaranch L, Salegio EA, San Sebastian W, et al. Adeno-associated virus serotype 9 transduction in the central nervous system of nonhuman primates. Hum Gene Ther. 2012 Apr;23(4):382–9. doi: 10.1089/hum.2011.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Manfredsson FP, Rising AC, Mandel RJ. AAV9: a potential blood-brain barrier buster. Mol Ther. 2009 Mar;17(3):403–5. doi: 10.1038/mt.2009.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Inagaki K, Fuess S, Storm TA, et al. Robust systemic transduction with AAV9 vectors in mice: efficient global cardiac gene transfer superior to that of AAV8. Mol Ther. 2006 Jul;14(1):45–53. doi: 10.1016/j.ymthe.2006.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Fyfe JC, Kurzhals RL, Hawkins MG, et al. A complex rearrangement in GBE1 causes both perinatal hypoglycemic collapse and late-juvenile-onset neuromuscular degeneration in glycogen storage disease type IV of Norwegian forest cats. Mol Genet Metab. 2007 Apr;90(4):383–92. doi: 10.1016/j.ymgme.2006.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kakhlon O, Glickstein H, Feinstein N, et al. Polyglucosan neurotoxicity caused by glycogen branching enzyme deficiency can be reversed by inhibition of glycogen synthase. J Neurochem. 2013 Oct;127(1):101–13. doi: 10.1111/jnc.12277. [DOI] [PubMed] [Google Scholar]
  • 111.Lucia A, Ruiz JR, Santalla A, et al. Genotypic and phenotypic features of McArdle disease: insights from the Spanish national registry. J Neurol Neurosurg Psychiatry. 2012 Mar;83(3):322–8. doi: 10.1136/jnnp-2011-301593. [DOI] [PubMed] [Google Scholar]
  • 112.Lucia A, Nogales-Gadea G, Perez M, Martin MA, Andreu AL, Arenas J. McArdle disease: what do neurologists need to know? Nat Clin Pract Neurol. 2008 Oct;4(10):568–77. doi: 10.1038/ncpneuro0913. [DOI] [PubMed] [Google Scholar]
  • 113.Tsujino S, Shanske S, Valberg SJ, Cardinet GH, 3rd, Smith BP, DiMauro S. Cloning of bovine muscle glycogen phosphorylase cDNA and identification of a mutation in cattle with myophosphorylase deficiency, an animal model for McArdle’s disease. Neuromuscul Disord. 1996 Jan;6(1):19–26. doi: 10.1016/0960-8966(95)00014-3. [DOI] [PubMed] [Google Scholar]
  • 114.Angelos S, Valberg SJ, Smith BP, et al. Myophosphorylase deficiency associated with rhabdomyolysis and exercise intolerance in 6 related Charolais cattle. Muscle Nerve. 1995 Jul;18(7):736–40. doi: 10.1002/mus.880180710. [DOI] [PubMed] [Google Scholar]
  • 115.Tan P, Allen JG, Wilton SD, Akkari PA, Huxtable CR, Laing NG. A splice-site mutation causing ovine McArdle’s disease. Neuromuscul Disord. 1997 Jul;7(5):336–42. doi: 10.1016/s0960-8966(97)00062-x. [DOI] [PubMed] [Google Scholar]
  • 116.Nogales-Gadea G, Pinos T, Lucia A, et al. Knock-in mice for the R50X mutation in the PYGM gene present with McArdle disease. Brain. 2012 Jul;135(Pt 7):2048–57. doi: 10.1093/brain/aws141. [DOI] [PubMed] [Google Scholar]
  • 117.Howell JM, Quinlivan R, Sewry C. Investigation of possible treatment regimes for McArdle’s disease using the sheep model of the disease. Neuromuscul Disord. 2008 Oct;18(9–10):828. [Google Scholar]
  • 118.Howell JM, Walker KR, Creed KE, et al. Phosphorylase re-expression, increase in the force of contraction and decreased fatigue following notexin-induced muscle damage and regeneration in the ovine model of McArdle disease. Neuromuscul Disord. 2014 Feb;24(2):167–77. doi: 10.1016/j.nmd.2013.10.003. [DOI] [PubMed] [Google Scholar]
  • 119.Musumeci O, Bruno C, Mongini T, et al. Clinical features and new molecular findings in muscle phosphofructokinase deficiency (GSD type VII) Neuromuscul Disord. 2012 Apr;22(4):325–30. doi: 10.1016/j.nmd.2011.10.022. [DOI] [PubMed] [Google Scholar]
  • 120.Giger U, Reilly MP, Asakura T, Baldwin CJ, Harvey JW. Autosomal recessive inherited phosphofructokinase deficiency in English springer spaniel dogs. Anim Genet. 1986;17(1):15–23. doi: 10.1111/j.1365-2052.1986.tb03184.x. [DOI] [PubMed] [Google Scholar]
  • 121.Giger U, Argov Z, Schnall M, Bank WJ, Chance B. Metabolic myopathy in canine muscle-type phosphofructokinase deficiency. Muscle Nerve. 1988;11(12):1260–5. doi: 10.1002/mus.880111210. [DOI] [PubMed] [Google Scholar]
  • 122.Smith BF, Stedman H, Rajpurohit Y, et al. Molecular basis of canine muscle type phosphofructokinase deficiency. J Biol Chem. 1996;271(33):20070–4. doi: 10.1074/jbc.271.33.20070. [DOI] [PubMed] [Google Scholar]
  • 123.Giger U, Smith BF, Woods CB, Patterson DF, Stedman H. Inherited phosphofructokinase deficiency in an American cocker spaniel. J Am Vet Med Assoc. 1992 Nov 15;201(10):1569–71. [PubMed] [Google Scholar]
  • 124.Gerber K, Harvey JW, D’Agorne S, Wood J, Giger U. Hemolysis, myopathy, and cardiac disease associated with hereditary phosphofructokinase deficiency in two Whippets. Vet Clin Pathol. 2009 Mar;38(1):46–51. doi: 10.1111/j.1939-165X.2008.00089.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hillstrom A, Tvedten H, Rowe A, Giger U. Hereditary phosphofructokinase deficiency in wachtelhunds. J Am Anim Hosp Assoc. 2011 Mar-Apr;47(2):145–50. doi: 10.5326/JAAHA-MS-5619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Richard AM, Webb DL, Goodman JM, et al. Tissue-dependent loss of phosphofructokinase-M in mice with interrupted activity of the distal promoter: impairment in insulin secretion. Am J Physiol Endocrinol Metab. 2007 Sep;293(3):E794–801. doi: 10.1152/ajpendo.00168.2007. [DOI] [PubMed] [Google Scholar]
  • 127.Getty-Kaushik L, Viereck JC, Goodman JM, et al. Mice deficient in phosphofructokinase-M have greatly decreased fat stores. Obesity. 2010 Mar;18(3):434–40. doi: 10.1038/oby.2009.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Garcia M, Pujol A, Ruzo A, et al. Phosphofructo-1-kinase deficiency leads to a severe cardiac and hematological disorder in addition to skeletal muscle glycogenosis. PLoS Genet. 2009 Aug;5(8):e1000615. doi: 10.1371/journal.pgen.1000615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Bruser A, Kirchberger J, Schoneberg T. Altered allosteric regulation of muscle 6-phosphofructokinase causes Tarui disease. Biochem Biophys Res Commun. 2012 Oct 12;427(1):133–7. doi: 10.1016/j.bbrc.2012.09.024. [DOI] [PubMed] [Google Scholar]

RESOURCES