Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 159
Filter
1.
J Inherit Metab Dis ; 47(1): 93-118, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37421310

ABSTRACT

Glycogen storage disorders (GSDs) are inherited disorders of metabolism resulting from the deficiency of individual enzymes involved in the synthesis, transport, and degradation of glycogen. This literature review summarizes the development of gene therapy for the GSDs. The abnormal accumulation of glycogen and deficiency of glucose production in GSDs lead to unique symptoms based upon the enzyme step and tissues involved, such as liver and kidney involvement associated with severe hypoglycemia during fasting and the risk of long-term complications including hepatic adenoma/carcinoma and end stage kidney disease in GSD Ia from glucose-6-phosphatase deficiency, and cardiac/skeletal/smooth muscle involvement associated with myopathy +/- cardiomyopathy and the risk for cardiorespiratory failure in Pompe disease. These symptoms are present to a variable degree in animal models for the GSDs, which have been utilized to evaluate new therapies including gene therapy and genome editing. Gene therapy for Pompe disease and GSD Ia has progressed to Phase I and Phase III clinical trials, respectively, and are evaluating the safety and bioactivity of adeno-associated virus vectors. Clinical research to understand the natural history and progression of the GSDs provides invaluable outcome measures that serve as endpoints to evaluate benefits in clinical trials. While promising, gene therapy and genome editing face challenges with regard to clinical implementation, including immune responses and toxicities that have been revealed during clinical trials of gene therapy that are underway. Gene therapy for the glycogen storage diseases is under development, addressing an unmet need for specific, stable therapy for these conditions.


Subject(s)
Carcinoma, Hepatocellular , Glycogen Storage Disease Type II , Glycogen Storage Disease Type I , Glycogen Storage Disease , Liver Neoplasms , Animals , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Glycogen Storage Disease/metabolism , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/therapy , Glycogen Storage Disease Type I/complications , Liver/metabolism , Glycogen/metabolism , Genetic Therapy/methods , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology
2.
FASEB J ; 37(11): e23216, 2023 11.
Article in English | MEDLINE | ID: mdl-37779422

ABSTRACT

Glycogen storage disease type Ib (GSD-Ib) is an autosomal recessive disorder caused by a deficiency in the glucose-6-phosphate (G6P) transporter (G6PT) that is responsible for transporting G6P into the endoplasmic reticulum. GSD-Ib is characterized by disturbances in glucose homeostasis, neutropenia, and neutrophil dysfunction. Although some studies have explored neutrophils abnormalities in GSD-Ib, investigations regarding monocytes/macrophages remain limited so far. In this study, we examined the impact of G6PT deficiency on monocyte-to-macrophage differentiation using bone marrow-derived monocytes from G6pt-/- mice as well as G6PT-deficient human THP-1 monocytes. Our findings revealed that G6PT-deficient monocytes exhibited immature differentiation into macrophages. Notably, the impaired differentiation observed in G6PT-deficient monocytes seemed to be associated with abnormal glucose metabolism, characterized by enhanced glucose consumption through glycolysis, even under quiescent conditions with oxidative phosphorylation. Furthermore, we observed a reduced secretion of inflammatory cytokines in G6PT-deficient THP-1 monocytes during the inflammatory response, despite their elevated glucose consumption. In conclusion, this study sheds light on the significance of G6PT in monocyte-to-macrophage differentiation and underscores its importance in maintaining glucose homeostasis and supporting immune response in GSD-Ib. These findings may contribute to a better understanding of the pathogenesis of GSD-Ib and potentially pave the way for the development of targeted therapeutic interventions.


Subject(s)
Glycogen Storage Disease Type I , Animals , Mice , Humans , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/metabolism , Glycogen Storage Disease Type I/therapy , Glucose-6-Phosphatase/metabolism , Glucose/metabolism , Macrophages/metabolism
3.
J Clin Invest ; 133(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37788110

ABSTRACT

Glycogen storage disease type 1a (GSD1a) is caused by a congenital deficiency of glucose-6-phosphatase-α (G6Pase-α, encoded by G6PC), which is primarily associated with life-threatening hypoglycemia. Although strict dietary management substantially improves life expectancy, patients still experience intermittent hypoglycemia and develop hepatic complications. Emerging therapies utilizing new modalities such as adeno-associated virus and mRNA with lipid nanoparticles are under development for GSD1a but potentially require complicated glycemic management throughout life. Here, we present an oligonucleotide-based therapy to produce intact G6Pase-α from a pathogenic human variant, G6PC c.648G>T, the most prevalent variant in East Asia causing aberrant splicing of G6PC. DS-4108b, a splice-switching oligonucleotide, was designed to correct this aberrant splicing, especially in liver. We generated a mouse strain with homozygous knockin of this variant that well reflected the pathophysiology of patients with GSD1a. DS-4108b recovered hepatic G6Pase activity through splicing correction and prevented hypoglycemia and various hepatic abnormalities in the mice. Moreover, DS-4108b had long-lasting efficacy of more than 12 weeks in mice that received a single dose and had favorable pharmacokinetics and tolerability in mice and monkeys. These findings together indicate that this oligonucleotide-based therapy could provide a sustainable and curative therapeutic option under easy disease management for GSD1a patients with G6PC c.648G>T.


Subject(s)
Glycogen Storage Disease Type I , Hypoglycemia , Humans , Mice , Animals , Oligonucleotides/genetics , Mice, Knockout , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/therapy , Glycogen Storage Disease Type I/complications , Liver/pathology , Glucose-6-Phosphatase/genetics , Hypoglycemia/genetics , Hypoglycemia/prevention & control
4.
Nat Rev Dis Primers ; 9(1): 46, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37679331

ABSTRACT

Glycogen storage diseases (GSDs) are a group of rare, monogenic disorders that share a defect in the synthesis or breakdown of glycogen. This Primer describes the multi-organ clinical features of hepatic GSDs and muscle GSDs, in addition to their epidemiology, biochemistry and mechanisms of disease, diagnosis, management, quality of life and future research directions. Some GSDs have available guidelines for diagnosis and management. Diagnostic considerations include phenotypic characterization, biomarkers, imaging, genetic testing, enzyme activity analysis and histology. Management includes surveillance for development of characteristic disease sequelae, avoidance of fasting in several hepatic GSDs, medically prescribed diets, appropriate exercise regimens and emergency letters. Specific therapeutic interventions are available for some diseases, such as enzyme replacement therapy to correct enzyme deficiency in Pompe disease and SGLT2 inhibitors for neutropenia and neutrophil dysfunction in GSD Ib. Progress in diagnosis, management and definitive therapies affects the natural course and hence morbidity and mortality. The natural history of GSDs is still being described. The quality of life of patients with these conditions varies, and standard sets of patient-centred outcomes have not yet been developed. The landscape of novel therapeutics and GSD clinical trials is vast, and emerging research is discussed herein.


Subject(s)
Glycogen Storage Disease Type II , Glycogen Storage Disease Type I , Glycogen Storage Disease , Humans , Quality of Life , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/therapy , Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/diagnosis , Glycogen Storage Disease Type I/therapy , Disease Progression
5.
J Inherit Metab Dis ; 46(6): 1147-1158, 2023 11.
Article in English | MEDLINE | ID: mdl-37467014

ABSTRACT

Glycogen storage disease type-Ia (GSD-Ia), characterized by impaired blood glucose homeostasis, is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC). Using the G6pc-R83C mouse model of GSD-Ia, we explored a CRISPR/Cas9-based double-strand DNA oligonucleotide (dsODN) insertional strategy that uses the nonhomologous end-joining repair mechanism to correct the pathogenic p.R83C variant in G6pc exon-2. The strategy is based on the insertion of a short dsODN into G6pc exon-2 to disrupt the native exon and to introduce an additional splice acceptor site and the correcting sequence. When transcribed and spliced, the edited gene would generate a wild-type mRNA encoding the native G6Pase-α protein. The editing reagents formulated in lipid nanoparticles (LNPs) were delivered to the liver. Mice were treated either with one dose of LNP-dsODN at age 4 weeks or with two doses of LNP-dsODN at age 2 and 4 weeks. The G6pc-R83C mice receiving successful editing expressed ~4% of normal hepatic G6Pase-α activity, maintained glucose homeostasis, lacked hypoglycemic seizures, and displayed normalized blood metabolite profile. The outcomes are consistent with preclinical studies supporting previous gene augmentation therapy which is currently in clinical trials. This editing strategy may offer the basis for a therapeutic approach with an earlier clinical intervention than gene augmentation, with the additional benefit of a potentially permanent correction of the GSD-Ia phenotype.


Subject(s)
Glycogen Storage Disease Type I , Oligonucleotides , Mice , Animals , Oligonucleotides/metabolism , CRISPR-Cas Systems , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/therapy , Glycogen Storage Disease Type I/metabolism , Liver/metabolism , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism
6.
Clin Nephrol ; 99(4): 197-202, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36871226

ABSTRACT

Type 1 glycogen storage disease (GSDI) is a rare autosomal recessive disorder caused by glucose-6-phosphatase (G6Pase) deficiency. We discuss a case of a 29-year-old gentleman who had GSDI with metabolic complications of hypoglycemia, hypertriglyceridemia, hyperuricemia, and short stature. He also suffered from advanced chronic kidney disease, nephrotic range proteinuria, and hepatic adenomas. He presented with acute pneumonia and refractory metabolic acidosis despite treatment with isotonic bicarbonate infusion, reversal of hypoglycemia, and lactic acidosis. He eventually required kidney replacement therapy. The case report highlights the multiple contributing mechanisms and challenges to managing refractory metabolic acidosis in a patient with GSDI. Important considerations for dialysis initiation, decision for long-term dialysis modality and kidney transplantation for patients with GSDI are also discussed in this case report.


Subject(s)
Acidosis , Glycogen Storage Disease Type I , Hypoglycemia , Renal Insufficiency, Chronic , Male , Humans , Adult , Renal Dialysis/adverse effects , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/diagnosis , Glycogen Storage Disease Type I/therapy , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Hypoglycemia/complications , Hypoglycemia/therapy
7.
Pediatr Endocrinol Diabetes Metab ; 28(3): 207-212, 2022.
Article in English | MEDLINE | ID: mdl-35620924

ABSTRACT

Glycogen storage disease type 1b (GSD 1b) is an inherited metabolic defect caused by a deficiency of microsomal glucose-6-phosphate (G6P) transport protein across the endoplasmic reticulum membrane. Patients with GSD 1b have hypoglycemia episodes, lactate acidosis, hypertriglyceridemia, hypercholesterolemia, hyperuricemia, neutropenia and in imaging studies hepatomegaly and/or nephromegaly. The primary goals of treatment are to maintain proper blood glucose levels and to increase the number of properly functioning neutrophils. The aim of the study was a retrospective analysis of the clinical picture and treatment results of pediatric patients with type 1b glycogen storage disease from Poland. The study included 13 patients from 3 clinical centers, with a median age at diagnosis as 5 months. In 11/13 patients, the diagnosis was confirmed by molecular test, by the presence of pathogenic variants on both alleles of the SLC37A4 gene. Ten out of 13 patients developed the first symptoms in the form of severe infection (sepsis and/or pneumonia) already in the neonatal-infant period. A hypoglycemia episode was observed before diagnosis in 8/13 patients, of which 4/8 patients presented symptoms in the form of generalized relaxation and/or seizures. Two patients developed hypertension, and 4/13 required long-term treatment of inflammatory bowel disease.


Subject(s)
Glycogen Storage Disease Type I , Hypoglycemia , Antiporters/genetics , Blood Glucose , Carrier Proteins , Glucose-6-Phosphate , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/diagnosis , Glycogen Storage Disease Type I/therapy , Humans , Infant , Infant, Newborn , Lactates , Monosaccharide Transport Proteins/genetics , Poland , Retrospective Studies
8.
BMJ Case Rep ; 14(8)2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34380672

ABSTRACT

Glycogen storage disease type 1a (GSD 1a) is a metabolic disorder caused by deficiency of an enzyme required for glycogen breakdown, causing hypoglycaemia and lactic acidosis. Metabolic derangements cause disease manifestations affecting the kidneys, liver and platelet function. Physiological changes in pregnancy worsen fasting intolerance and increase reliance on exogenous glucose to avoid lactic acidosis. Fetal macrosomia and declining respiratory function result in high rates of caesarean sections. We report the multidisciplinary team (MDT) management of a 25-year-old woman with GSD 1a in an unplanned pregnancy. Existing percutaneous endoscopic gastrostomy tube feeding, alongside high-calorie drinks and intravenous dextrose during labour, managed the risks of hypoglycaemia and lactic acidosis. Metabolic parameters were regularly monitored and fortnightly growth scans were assessed for macrosomia. Allopurinol was continued throughout the pregnancy to reduce the risk of hyperuricaemia. MDT management optimised maternal and fetal care throughout pregnancy and labour, resulting in a successful vaginal delivery.


Subject(s)
Glycogen Storage Disease Type I , Hypoglycemia , Labor, Obstetric , Pregnancy Complications , Adult , Cesarean Section , Female , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/therapy , Humans , Hypoglycemia/etiology , Pregnancy , Pregnancy Complications/therapy
9.
Arq Gastroenterol ; 58(1): 87-92, 2021.
Article in English | MEDLINE | ID: mdl-33909803

ABSTRACT

BACKGROUND: Glycogen storage disease (GSD) type 1b is a multisystemic disease in which immune and infectious complications are present, in addition to the well-known metabolic manifestations of GSD. Treatment with granulocyte-colony stimulating factor (G-CSF) is often indicated in the management of neutropenia and inflammatory bowel disease. OBJECTIVE: To report on the demographics, genotype, clinical presentation, management, and complications of pediatric patients with glycogen storage disease type 1b (GSD 1b), with special attention to immune-related complications. METHODS: Retrospective case series of seven patients with GSD 1b diagnosed and followed at a tertiary university hospital in Brazil, from July/2000 until July/2016. RESULTS: Mean age at referral was fourteen months. Diagnosis of GSD 1b was based on clinical and laboratory findings and supported by genetic studies in five cases. All patients presented suffered from neutropenia, managed with G-CSF - specifically Filgrastim. Hospitalizations for infections were frequent. Two patients developed inflammatory bowel disease. Six patients remained alive, one died at age 14 years and 9 months. The mean age at the end of the follow-up was 11.5 years. Compliance to treatment was suboptimal: poor compliance to medications, starch and dietetic management of GSD were documented, and outpatient appointments were frequently missed. CONCLUSION: Managing GSD 1b is challenging not only for the chronic and multisystemic nature of this disease, but also for the additional demands related dietary restrictions, use of multiple medications and the need for frequent follow-up visits; furthermore in Brazil, the difficulties are increased in a scenario where we frequently care for patients with unfavorable socioeconomic status and with irregular supply of medications in the public health system.


Subject(s)
Glycogen Storage Disease Type I , Neutropenia , Adolescent , Brazil , Child , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/therapy , Granulocyte Colony-Stimulating Factor , Humans , Retrospective Studies
10.
Arq. gastroenterol ; 58(1): 87-92, Jan.-Mar. 2021. tab
Article in English | LILACS | ID: biblio-1248988

ABSTRACT

ABSTRACT BACKGROUND Glycogen storage disease (GSD) type 1b is a multisystemic disease in which immune and infectious complications are present, in addition to the well-known metabolic manifestations of GSD. Treatment with granulocyte-colony stimulating factor (G-CSF) is often indicated in the management of neutropenia and inflammatory bowel disease. OBJECTIVE To report on the demographics, genotype, clinical presentation, management, and complications of pediatric patients with glycogen storage disease type 1b (GSD 1b), with special attention to immune-related complications. METHODS Retrospective case series of seven patients with GSD 1b diagnosed and followed at a tertiary university hospital in Brazil, from July/2000 until July/2016. RESULTS Mean age at referral was fourteen months. Diagnosis of GSD 1b was based on clinical and laboratory findings and supported by genetic studies in five cases. All patients presented suffered from neutropenia, managed with G-CSF - specifically Filgrastim. Hospitalizations for infections were frequent. Two patients developed inflammatory bowel disease. Six patients remained alive, one died at age 14 years and 9 months. The mean age at the end of the follow-up was 11.5 years. Compliance to treatment was suboptimal: poor compliance to medications, starch and dietetic management of GSD were documented, and outpatient appointments were frequently missed. CONCLUSION Managing GSD 1b is challenging not only for the chronic and multisystemic nature of this disease, but also for the additional demands related dietary restrictions, use of multiple medications and the need for frequent follow-up visits; furthermore in Brazil, the difficulties are increased in a scenario where we frequently care for patients with unfavorable socioeconomic status and with irregular supply of medications in the public health system.


RESUMO CONTEXTO Glicogenose (GSD) tipo 1b é uma doença multissistêmica em que complicações imunológicas e infecciosas estão presentes, além das manifestações metabólicas bem conhecidas da GSD. O tratamento com fator estimulador de colônias de granulócitos (G-CSF) é frequentemente indicado no tratamento da neutropenia e doença inflamatória intestinal. OBJETIVO Relatar sobre a dados demográficos, genótipo, apresentação clínica, manejo e complicações de pacientes pediátricos com GSD tipo 1b (GSD 1b), com atenção especial às complicações relacionadas ao sistema imunológico. MÉTODOS Série de casos retrospectiva de sete pacientes com GSD 1b diagnosticados e acompanhados em um hospital universitário terciário no Brasil, de julho/2000 a julho/2016. RESULTADOS A idade média no encaminhamento foi de 14 meses. O diagnóstico de GSD 1b foi baseado em achados clínicos e laboratoriais e apoiado por estudos genéticos em cinco casos. Todos os pacientes apresentaram neutropenia, tratada com G-CSF - especificamente Filgrastim. As hospitalizações por infecções foram frequentes. Dois pacientes desenvolveram doença inflamatória intestinal. Seis pacientes permanecem vivos, um morreu aos 14 anos e 9 meses de idade. A média de idade ao final do acompanhamento foi de 11,5 anos. A adesão ao tratamento foi sub-ótima: má adesão aos medicamentos, amido e manejo dietético de GSD foram documentados, e consultas ambulatoriais foram frequentemente perdidas. CONCLUSÃO O manejo da GSD 1b é um desafio, não apenas pela natureza crônica e multissistêmica desta doença, mas também pelas demandas adicionais relacionadas a restrições dietéticas, uso de múltiplos medicamentos e a necessidade de consultas de acompanhamento frequentes; no Brasil, isso ainda é dificultado em um cenário em que frequentemente atendemos pacientes com situação socioeconômica desfavorável e com oferta irregular de medicamentos no sistema público de saúde.


Subject(s)
Humans , Child , Adolescent , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/therapy , Neutropenia , Brazil , Retrospective Studies , Granulocyte Colony-Stimulating Factor
11.
J Inherit Metab Dis ; 44(1): 118-128, 2021 01.
Article in English | MEDLINE | ID: mdl-32474930

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) is an inherited metabolic disease caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC) which plays a critical role in blood glucose homeostasis by catalyzing the hydrolysis of glucose-6-phosphate (G6P) to glucose and phosphate in the terminal step of glycogenolysis and gluconeogenesis. Patients with GSD-Ia manifest life-threatening fasting hypoglycemia along with the excessive accumulation of hepatic glycogen and triglycerides which results in hepatomegaly and a risk of long-term complications such as hepatocellular adenoma and carcinoma (HCA/HCC). The etiology of HCA/HCC development in GSD-Ia, however, is unknown. Recent studies have shown that the livers in model animals of GSD-Ia display impairment of autophagy, a cellular recycling process which is critical for energy metabolism and cellular homeostasis. However, molecular mechanisms of autophagy impairment and its involvement in pathogenesis in GSD-Ia are still under investigation. Here, we summarize the latest advances for signaling pathways implicated in hepatic autophagy impairment and the roles of autophagy in hepatic tumorigenesis in GSD-Ia. In addition, recent evidence has illustrated that autophagy plays an important role in hepatic metabolism and liver-directed gene therapy mediated by recombinant adeno-associated virus (rAAV). Therefore, we highlight the possible role of hepatic autophagy in metabolic control and rAAV-mediated gene therapy for GSD-Ia. In this review, we also provide potential therapeutic strategies for GSD-Ia on the basis of molecular mechanisms underlying hepatic autophagy impairment in GSD-Ia.


Subject(s)
Autophagy , Carcinoma, Hepatocellular/prevention & control , Genetic Therapy/methods , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Liver Neoplasms/prevention & control , Animals , Carcinogenesis/pathology , Carcinoma, Hepatocellular/enzymology , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Glucose/metabolism , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/enzymology , Homeostasis , Humans , Liver/enzymology , Liver Neoplasms/enzymology , Mice , Mice, Knockout , Signal Transduction
12.
Mol Ther ; 29(4): 1602-1610, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33359667

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia), deficient in glucose-6-phosphatase-α (G6PC), is characterized by impaired glucose homeostasis and a hallmark of fasting hypoglycemia. We have developed a recombinant adeno-associated virus (rAAV) vector-mediated gene therapy for GSD-Ia that is currently in a phase I/II clinical trial. While therapeutic expression of the episomal rAAV-G6PC clinical vector is stable in mice, the long-term durability of expression in humans is currently being established. Here we evaluated CRISPR/Cas9-based in vivo genome editing technology to correct a prevalent pathogenic human variant, G6PC-p.R83C. We have generated a homozygous G6pc-R83C mouse strain and shown that the G6pc-R83C mice manifest impaired glucose homeostasis and frequent hypoglycemic seizures, mimicking the pathophysiology of GSD-Ia patients. We then used a CRISPR/Cas9-based gene editing system to treat newborn G6pc-R83C mice and showed that the treated mice grew normally to age 16 weeks without hypoglycemia seizures. The treated G6pc-R83C mice, expressing ≥ 3% of normal hepatic G6Pase-α activity, maintained glucose homeostasis, displayed normalized blood metabolites, and could sustain 24 h of fasting. Taken together, we have developed a second-generation therapy in which in vivo correction of a pathogenic G6PC-p.R83C variant in its native genetic locus could lead to potentially permanent, durable, long-term correction of the GSD-Ia phenotype.


Subject(s)
Gene Editing , Genetic Therapy , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Animals , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/genetics , Glucose/genetics , Glucose/metabolism , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/metabolism , Glycogen Storage Disease Type I/pathology , Humans , Liver/metabolism , Liver/pathology , Mice
13.
O.F.I.L ; 31(4)2021.
Article in Spanish | IBECS | ID: ibc-224763

ABSTRACT

La glucogenosis tipo Ib es una patología metabólica rara de carácter hereditario originada por deficiencias en el transportador microsomal de glucosa-6-fosfatasa. Considerando la baja incidencia de esta enfermedad, los casos clínicos suponen la fuente principal de información sobre su manejo. La neutropenia secundaria a la glucogenosis tipo Ib se asocia con la aparición de infecciones y enfermedad inflamatoria intestinal. El tratamiento es la administración de factores estimulantes de colonias (CSF) diariamente. La literatura previa describe la caída abrupta e inmediata del recuento de neutrófilos tras la interrupción del tratamiento con CSF. Se presenta el caso clínico de una mujer tratada con filgrastim cada 48 horas a dosis menores que no ha sido descrita previamente, estabilizando el recuento de neutrófilos por encima de 1 x 109 células/L sin aparición de infecciones. Por otra parte, se muestra el impacto negativo del contacto público, junto a medidas adoptadas que evitaron la aparición de infecciones durante la pandemia de la COVID-19. Por tanto, este caso clínico puede aportar información a diferentes niveles sobre el manejo de los pacientes con glucogenosis tipo Ib. (AU)


Glycogen storage disease type Ib is a rare hereditary metabolic pathology caused by deficiencies in the microsomal transporter of glucose-6-phosphatase. Considering the low incidence of this disease, clinical cases are the main source of information on its management. Neutropenia secondary to glycogenosis type Ib is associated with the appearance of infections and inflammatory bowel disease. The treatment is daily administration of colony stimulating factors (CSF). Previous literature describes the abrupt and immediate drop in the neutrophil count after discontinuation of CSF treatment. We present a case of a woman treated with filgrastim every 48 hours using a lower dose that has not been previously described. The neutrophil count was stabilized above 1 x 109 cells/L without the appearance of infections. On the other hand, the negative impact of public contact along with measures to prevent the appearance of infections during the COVID-19 pandemic was showed. Therefore, this case report can provide information at different levels on the management of patients with glycogen storage disease type Ib (AU).


Subject(s)
Humans , Female , Young Adult , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/diagnosis , Glycogen Storage Disease Type I/drug therapy , Glycogen Storage Disease Type I/therapy , Filgrastim/therapeutic use , Neutropenia , Coronavirus Infections/immunology
14.
Biochem Biophys Res Commun ; 527(3): 824-830, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32430177

ABSTRACT

The current phase I/II clinical trial for human glycogen storage disease type-Ia (GSD-Ia) (NCT03517085) uses a recombinant adeno-associated virus (rAAV) vector expressing a codon-optimized human glucose-6-phosphatase-α (G6Pase-α or G6PC). DNA sequence changes introduced by codon-optimization can negatively impact gene expression. We therefore generated a novel variant in which a single amino acid change, S298C, is introduced into the native human G6PC sequence. Short term gene transfer study in G6pc-/- mice showed that the rAAV-G6PC-S298C vector is 3-fold more efficacious than the native rAAV-G6PC vector. We have shown previously that restoring 3% of normal hepatic G6Pase-α activity in G6pc-/- mice prevents hepatocellular adenoma/carcinoma (HCA/HCC) development and that mice harboring <3% of normal hepatic G6Pase-α activity are at risk of tumor development. We have also shown that G6Pase-α deficiency leads to hepatic autophagy impairment that can contribute to hepatocarcinogenesis. We now undertake a long-term (66-week) preclinical characterization of the rAAV-G6PC-S298C vector in GSD-Ia gene therapy. We show that the increased efficacy of rAAV-G6PC-S298C has enabled the G6pc-/- mice treated with a lower dose of this vector to survive long-term. We further show that mice expressing ≥3% of normal hepatic G6Pase-α activity do not develop hepatic tumors or autophagy impairment but mice expressing <3% of normal hepatic G6Pase-α activity display impaired hepatic autophagy with one developing HCA/HCC nodules. Our study shows that the rAAV-G6PC-S298C vector provides equal or greater efficacy to the codon optimization approach, offering a valuable alternative vector for clinical translation in human GSD-Ia.


Subject(s)
Genetic Therapy , Genetic Vectors/therapeutic use , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Point Mutation , Animals , Autophagy , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/genetics , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/pathology , Humans , Liver/metabolism , Liver/pathology , Mice
18.
Hum Gene Ther ; 30(10): 1263-1273, 2019 10.
Article in English | MEDLINE | ID: mdl-31319709

ABSTRACT

Glycogen storage diseases (GSDs) type I (GSDI) and type III (GSDIII), the most frequent hepatic GSDs, are due to defects in glycogen metabolism, mainly in the liver. In addition to hypoglycemia and liver pathology, renal, myeloid, or muscle complications affect GSDI and GSDIII patients. Currently, patient management is based on dietary treatment preventing severe hypoglycemia and increasing the lifespan of patients. However, most of the patients develop long-term pathologies. In the past years, gene therapy for GSDI has generated proof of concept for hepatic GSDs. This resulted in a recent clinical trial of adeno-associated virus (AAV)-based gene replacement for GSDIa. However, the current limitations of AAV-mediated gene transfer still represent a challenge for successful gene therapy in GSDI and GSDIII. Indeed, transgene loss over time was observed in GSDI liver, possibly due to the degeneration of hepatocytes underlying the physiopathology of both GSDI and GSDIII and leading to hepatic tumor development. Moreover, multitissue targeting requires high vector doses to target nonpermissive tissues such as muscle and kidney. Interestingly, recent pharmacological interventions or dietary regimen aiming at the amelioration of the hepatocyte abnormalities before the administration of gene therapy demonstrated improved efficacy in GSDs. In this review, we describe the advances in gene therapy and the limitations to be overcome to achieve efficient and safe gene transfer in GSDs.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type III/therapy , Glycogen Storage Disease Type I/therapy , Hypoglycemia/therapy , Animals , Clinical Trials as Topic , Dependovirus/metabolism , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glucose-6-Phosphatase/metabolism , Glycogen/biosynthesis , Glycogen Storage Disease Type I/enzymology , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/pathology , Glycogen Storage Disease Type III/enzymology , Glycogen Storage Disease Type III/genetics , Glycogen Storage Disease Type III/pathology , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Hypoglycemia/enzymology , Hypoglycemia/genetics , Hypoglycemia/pathology , Liver/enzymology , Liver/pathology , Transgenes
19.
J Inherit Metab Dis ; 42(3): 470-479, 2019 05.
Article in English | MEDLINE | ID: mdl-30714174

ABSTRACT

Glycogen storage disease type-Ia (GSD-Ia), caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC), is characterized by impaired glucose homeostasis with a hallmark hypoglycemia, following a short fast. We have shown that G6pc-deficient (G6pc-/-) mice treated with recombinant adeno-associated virus (rAAV) vectors expressing either wild-type (WT) (rAAV-hG6PC-WT) or codon-optimized (co) (rAAV-co-hG6PC) human (h) G6Pase-α maintain glucose homeostasis if they restore ≥3% of normal hepatic G6Pase-α activity. The co vector, which has a higher potency, is currently being used in a phase I/II clinical trial for human GSD-Ia (NCT03517085). While routinely used in clinical therapies, co vectors may not always be optimal. Codon-optimization can impact RNA secondary structure, change RNA/DNA protein-binding sites, affect protein conformation and function, and alter posttranscriptional modifications that may reduce potency or efficacy. We therefore sought to develop alternative approaches to increase the potency of the G6PC gene transfer vectors. Using an evolutionary sequence analysis, we identified a Ser-298 to Cys-298 substitution naturally found in canine, mouse, rat, and several primate G6Pase-α isozymes, that when incorporated into the WT hG6Pase-α sequence, markedly enhanced enzymatic activity. Using G6pc-/- mice, we show that the efficacy of the rAAV-hG6PC-S298C vector was 3-fold higher than that of the rAAV-hG6PC-WT vector. The rAAV-hG6PC-S298C vector with increased efficacy, that minimizes the potential problems associated with codon-optimization, offers a valuable vector for clinical translation in human GSD-Ia.


Subject(s)
Genetic Therapy/methods , Glucose-6-Phosphatase/genetics , Glucose/metabolism , Glycogen Storage Disease Type I/therapy , Animals , Dependovirus/genetics , Disease Models, Animal , Dogs , Genetic Vectors/administration & dosage , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/enzymology , Homeostasis , Humans , Liver/enzymology , Mice , Mice, Knockout , Rats
20.
J Inherit Metab Dis ; 42(3): 459-469, 2019 05.
Article in English | MEDLINE | ID: mdl-30637773

ABSTRACT

Hepatocellular adenoma/carcinoma (HCA/HCC) is a long-term complication of glycogen storage disease type-Ia (GSD-Ia), which is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC), a key enzyme in gluconeogenesis. Currently, there is no therapy to address HCA/HCC in GSD-Ia. We have previously shown that a recombinant adeno-associated virus (rAAV) vector-mediated G6PC gene transfer to 2-week-old G6pc-/- mice prevents HCA development. However, it remains unclear whether G6PC gene transfer at the tumor developing stage of GSD-Ia can prevent tumor initiation or abrogate the pre-existing tumors. Using liver-specific G6pc-knockout (L-G6pc-/-) mice that develop HCA/HCC, we now show that treating the mice at the tumor-developing stage with rAAV-G6PC restores hepatic G6Pase-α expression, normalizes glucose homeostasis, and prevents de novo HCA/HCC development. The rAAV-G6PC treatment also normalizes defective hepatic autophagy and corrects metabolic abnormalities in the nontumor liver tissues of both tumor-free and tumor-bearing mice. However, gene therapy cannot restore G6Pase-α expression in the HCA/HCC lesions and fails to abrogate any pre-existing tumors. We show that the expression of 11 ß-hydroxysteroid dehydrogenase type-1 that mediates local glucocorticoid activation is downregulated in HCA/HCC lesions, leading to impairment in glucocorticoid signaling critical for gluconeogenesis activation. This suggests that local glucocorticoid action downregulation in the HCA/HCC lesions may suppress gene therapy mediated G6Pase-α restoration. Collectively, our data show that rAAV-mediated gene therapy can prevent de novo HCA/HCC development in L-G6pc-/- mice at the tumor developing stage, but it cannot reduce any pre-existing tumor burden.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Genetic Therapy/methods , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Liver Neoplasms/prevention & control , Animals , Carcinoma, Hepatocellular/enzymology , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Glucose/metabolism , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/enzymology , Homeostasis , Humans , Liver/enzymology , Liver Neoplasms/enzymology , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...