Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Inherit Metab Dis ; 46(1): 55-65, 2023 01.
Article in English | MEDLINE | ID: mdl-36220785

ABSTRACT

Protein catabolism ultimately yields toxic ammonia, which must be converted to urea by the liver for renal excretion. In extrahepatic tissues, ammonia is temporarily converted primarily to glutamine for subsequent hepatic extraction. Urea cycle disorders (UCDs) are inborn errors of metabolism causing impaired ureagenesis, leading to neurotoxic accumulation of ammonia and brain glutamine. Treatment includes dietary protein restriction and oral "ammonia scavengers." These scavengers chemically combine with glutamine and glycine to yield excretable products, creating an alternate pathway of waste nitrogen disposal. The amino acid transporter SLC6A19 is responsible for >95% of absorption and reabsorption of free neutral amino acids in the small intestine and kidney, respectively. Genetic SLC6A19 deficiency causes massive neutral aminoaciduria but is typically benign. We hypothesized that inhibiting SLC6A19 would open a novel and effective alternate pathway of waste nitrogen disposal. To test this, we crossed SLC6A19 knockout (KO) mice with spfash mice, a model of ornithine transcarbamylase (OTC) deficiency. Loss of SLC6A19 in spfash mice normalized plasma ammonia and brain glutamine and increased median survival in response to a high protein diet from 7 to 97 days. While induced excretion of amino acid nitrogen is likely the primary therapeutic mechanism, reduced intestinal absorption of dietary free amino acids, and decreased muscle protein turnover due to loss of SLC6A19 may also play a role. In summary, the results suggest that SLC6A19 inhibition represents a promising approach to treating UCDs and related aminoacidopathies.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Amino Acid Transport Systems, Neutral , Ornithine Carbamoyltransferase Deficiency Disease , Mice , Animals , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Glutamine , Nitrogen/metabolism , Ammonia , Disease Models, Animal , Mice, Knockout , Urea/metabolism , Ornithine Carbamoyltransferase/genetics , Amino Acid Transport Systems, Neutral/genetics
2.
J Proteome Res ; 21(1): 151-163, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34843255

ABSTRACT

Microscale-based separations are increasingly being applied in the field of metabolomics for the analysis of small-molecule metabolites. These methods have the potential to provide improved sensitivity, less solvent waste, and reduced sample-size requirements. Ion-pair free microflow-based global metabolomics methods, which we recently reported, were further compared to analytical flow ion-pairing reagent containing methods using a sample set from a urea cycle disorder (UCD) mouse model. Mouse urine and brain homogenate samples representing healthy, diseased, and disease-treated animals were analyzed by both methods. Data processing was performed using univariate and multivariate techniques followed by analyte trend analysis. The microflow methods performed comparably to the analytical flow ion-pairing methods with the ability to separate the three sample groups when analyzed by partial least-squares analysis. The number of detected metabolic features present after each data processing step was similar between the microflow-based methods and the ion-pairing methods in the negative ionization mode. The observed analyte trend and coverage of known UCD biomarkers were the same for both evaluated approaches. The 12.5-fold reduction in sample injection volume required for the microflow-based separations highlights the potential of this method to support studies with sample-size limitations.


Subject(s)
Metabolomics , Urea Cycle Disorders, Inborn , Animals , Chromatography, Liquid/methods , Mass Spectrometry/methods , Metabolomics/methods , Mice , Solvents/chemistry , Urea Cycle Disorders, Inborn/diagnosis
3.
JCI Insight ; 3(14)2018 07 26.
Article in English | MEDLINE | ID: mdl-30046012

ABSTRACT

The neuropathological effects of phenylketonuria (PKU) stem from the inability of the body to metabolize excess phenylalanine (Phe), resulting in accumulation of Phe in the blood and brain. Since the kidney normally reabsorbs circulating amino acids with high efficiency, we hypothesized that preventing the renal uptake of Phe might provide a disposal pathway that could lower systemic Phe levels. SLC6A19 is a neutral amino acid transporter responsible for absorption of the majority of free Phe in the small intestine and reuptake of Phe by renal proximal tubule cells. Transgenic KO mice lacking SLC6A19 have elevated levels of Phe and other amino acids in their urine but are otherwise healthy. Here, we crossed the Pahenu2 mouse model of PKU with the Slc6a19-KO mouse. These mutant/KO mice exhibited abundant excretion of Phe in the urine and an approximately 70% decrease in plasma Phe levels. Importantly, brain Phe levels were decreased by 50%, and the levels of key neurotransmitters were increased in the mutant/KO mice. In addition, a deficit in spatial working memory and markers of neuropathology were corrected. Finally, treatment of Pahenu2 mice with Slc6a19 antisense oligonucleotides lowered Phe levels. The results suggest that inhibition of SLC6A19 may represent a novel approach for the treatment of PKU and related aminoacidopathies.


Subject(s)
Amino Acid Transport Systems, Neutral/analysis , Amino Acid Transport Systems, Neutral/metabolism , Amino Acids, Neutral/metabolism , Biological Transport/drug effects , Phenylketonurias/therapy , Amines , Amino Acid Transport Systems, Neutral/genetics , Amino Acids, Neutral/blood , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Genetic Diseases, Inborn/therapy , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Memory, Short-Term , Mice , Mice, Knockout , Morpholinos/pharmacology , Oligonucleotides/pharmacology , Phenylalanine/blood , Phenylalanine/metabolism , Phenylketonurias/pathology , Renal Reabsorption/drug effects
4.
Mol Genet Metab ; 117(4): 447-55, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26857995

ABSTRACT

Mucolipidoses II and III (ML II and ML III) are lysosomal disorders in which the mannose 6-phosphate recognition marker is absent from lysosomal hydrolases and other glycoproteins due to mutations in GNPTAB, which encodes two of three subunits of the heterohexameric enzyme, N-acetylglucosamine-1-phosphotransferase. Both disorders are caused by the same gene, but ML II represents the more severe phenotype. Bone manifestations of ML II include hip dysplasia, scoliosis, rickets and osteogenesis imperfecta. In this study, we sought to determine whether a recombinant adeno-associated viral vector (AAV2/8-GNPTAB) could confer high and prolonged gene expression of GNPTAB and thereby influence the pathology in the cartilage and bone tissue of a GNPTAB knock out (KO) mouse model. The results demonstrated significant increases in bone mineral density and content in AAV2/8-GNPTAB-treated as compared to non-treated KO mice. We also showed that IL-6 (interleukin-6) expression in articular cartilage was reduced in AAV2/8-GNPTAB treated ML II mice. Together, these data suggest that AAV-mediated expression of GNPTAB in ML II mice can attenuate bone loss via inhibition of IL-6 production. This study emphasizes the value of the MLII KO mouse to recapitulate the clinical manifestations of the disease and highlights its amenability to therapy.


Subject(s)
Bone Demineralization, Pathologic/etiology , Dependovirus/genetics , Gene Expression , Genetic Vectors/genetics , Mucolipidoses/genetics , Mucolipidoses/pathology , Transduction, Genetic , Transferases (Other Substituted Phosphate Groups)/genetics , Animals , Bone Demineralization, Pathologic/diagnosis , Bone Demineralization, Pathologic/therapy , Bone Density , Disease Models, Animal , Gene Order , Gene Targeting , Genetic Loci , Genetic Therapy , Genetic Vectors/administration & dosage , Genotype , Humans , Mice , Mice, Knockout , Mucolipidoses/therapy , Phenotype
5.
Mol Genet Metab ; 109(4): 339-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23867524

ABSTRACT

Enzyme replacement therapy is often hampered by the rapid clearance and degradation of the administered enzyme, limiting its efficacy and requiring frequent dosing. Encapsulation of therapeutic molecules into red blood cells (RBCs) is a clinically proven approach to improve the pharmacokinetics and efficacy of biologics and small molecule drugs. Here we evaluated the ability of RBCs encapsulated with phenylalanine hydroxylase (PAH) to metabolize phenylalanine (Phe) from the blood and confer sustained enzymatic activity in the circulation. Significant quantities of PAH were successfully encapsulated within murine RBCs (PAH-RBCs) with minimal loss of endogenous hemoglobin. While intravenously administered free PAH enzyme was rapidly eliminated from the blood within a few hours, PAH-RBCs persisted in the circulation for at least 10days. A single injection of PAH-RBCs was able to decrease Phe levels by nearly 80% in normal mice. These results demonstrate the ability of enzyme-loaded RBCs to metabolize circulating amino acids and highlight the potential to treat disorders of amino acid metabolism.


Subject(s)
Enzyme Replacement Therapy , Erythrocytes/enzymology , Phenylalanine Hydroxylase/genetics , Phenylalanine/blood , Phenylketonurias/enzymology , Animals , Drug Delivery Systems , Hemoglobins/metabolism , Humans , Liver/enzymology , Liver/metabolism , Mice , Phenylalanine Hydroxylase/pharmacokinetics , Phenylketonurias/blood , Phenylketonurias/genetics , Phenylketonurias/therapy
6.
Pediatr Endocrinol Rev ; 11 Suppl 1: 99-109, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24380128

ABSTRACT

Lysosomal storage diseases (LSDs) are a group of single-gene disorders that have proven to be highly informative in revealing the merits of gene transfer as a technology platform. Over the past several years considerable progress has been made in delivering therapeutic genes to peripheral tissues as well as the central nervous system. The current leading vectors for direct genetic modification of target cells in vivo are derived from adeno-associated viruses (AAV) and lentiviruses. These vectors are capable of conferring widespread, robust, and sustained expression of a given gene in several mouse models of LSDs. Here we review recent progress using recombinant AAV and lentiviruses to treat various LSDs and the remaining challenges to translate the results in mice to human patients.


Subject(s)
Genetic Therapy/methods , Lysosomal Storage Diseases/therapy , Animals , Gene Transfer Techniques , Humans , Lysosomal Storage Diseases/genetics
7.
J Gene Med ; 13(6): 324-32, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21674735

ABSTRACT

BACKGROUND: The secretory form of acid sphingomyelinase (ASM) is postulated to play a key role in the retention and aggregation of lipoproteins in the subendothelial space of the arterial wall by converting sphingomyelin in lipoproteins into ceramide. The present study aimed to determine whether the level of circulating ASM activity affects lesion development in mouse model of atherosclerosis. METHODS: Apolipoprotein E deficient (ApoE(-/-) ) mice were injected intravenously with a recombinant adeno-associated virus (AAV8-ASM) that constitutively expressed high levels of human ASM in liver and plasma. RESULTS: Plasma sphingomyelin levels were reduced at early but not later time points after the administration of AAV8-ASM despite persistently elevated circulating ASM. No change in serum lipoprotein levels was observed. Thirteen or 17 weeks after the administration of AAV8-ASM, the amount of plaque formation in the aortic sinus was comparable to that of mice treated with a control AAV. CONCLUSIONS: Unexpectedly, the lesion area of the entire aorta was reduced significantly in the AAV8-ASM virus-treated group. Hepatic expression and secretion of ASM into the circulation did not accelerate or exacerbate, but rather decreased, lesion formation in ApoE(-/-) mice. Thus, plasma ASM activity does not appear to be rate limiting for plaque formation during atherogenesis.


Subject(s)
Aorta/pathology , Apolipoproteins E/genetics , Dependovirus/metabolism , Plaque, Atherosclerotic/enzymology , Sphingomyelin Phosphodiesterase/metabolism , Analysis of Variance , Animals , Histological Techniques , Humans , Lipoproteins/blood , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/pathology , Sphingomyelin Phosphodiesterase/administration & dosage , Sphingomyelin Phosphodiesterase/blood
8.
PLoS One ; 5(6): e11239, 2010 Jun 21.
Article in English | MEDLINE | ID: mdl-20574539

ABSTRACT

BACKGROUND: Obesity is characterized by the accumulation of fat in the liver and other tissues, leading to insulin resistance. We have previously shown that a specific inhibitor of glucosylceramide synthase, which inhibits the initial step in the synthesis of glycosphingolipids (GSLs), improved glucose metabolism and decreased hepatic steatosis in both ob/ob and diet-induced obese (DIO) mice. Here we have determined in the DIO mouse model the efficacy of a related small molecule compound, Genz-112638, which is currently being evaluated clinically for the treatment of Gaucher disease, a lysosomal storage disorder. METHODOLOGY/PRINCIPAL FINDINGS: DIO mice were treated with the Genz-112638 for 12 to 16 weeks by daily oral gavage. Genz-112638 lowered HbA1c levels and increased glucose tolerance. Whole body adiposity was not affected in normal mice, but decreased in drug-treated obese mice. Drug treatment also significantly lowered liver triglyceride levels and reduced the development of hepatic steatosis. We performed hyperinsulinemic-euglycemic clamps on the DIO mice treated with Genz-112638 and showed that insulin-mediated suppression of hepatic glucose production increased significantly compared to the placebo treated mice, indicating a marked improvement in hepatic insulin sensitivity. CONCLUSIONS/SIGNIFICANCE: These results indicate that GSL inhibition in obese mice primarily results in an increase in insulin action in the liver, and suggests that GSLs may have an important role in hepatic insulin resistance in conditions of obesity.


Subject(s)
Diet/adverse effects , Glucosyltransferases/antagonists & inhibitors , Insulin/metabolism , Liver/drug effects , Liver/metabolism , Obesity/metabolism , Pyrrolidines/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Body Weight/drug effects , Eating/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Fatty Liver/drug therapy , Fatty Liver/metabolism , Glucose/metabolism , Glucose Clamp Technique , Glycated Hemoglobin/metabolism , Hyperinsulinism/metabolism , Male , Mice , Obesity/enzymology , Obesity/etiology , Obesity/physiopathology , Pyrrolidines/therapeutic use , Sphingolipids/metabolism
9.
J Bone Miner Metab ; 28(5): 510-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20300790

ABSTRACT

Studies examining the effects of hypoxia upon osteoclast biology have consistently revealed a stimulatory effect; both osteoclast differentiation and resorption activity have been shown to be enhanced in the presence of hypoxia. In the present study we examined the effects of the hypoxia mimetics dimethyloxallyl glycine (DMOG) and desferrioxamine (DFO) upon osteoclastogenesis. In contrast to hypoxia, our studies revealed a dose-dependent inhibition of osteoclast formation from macrophages treated with DMOG and DFO. Moreover, expression of a constitutively active form of hypoxia-inducible factor 1alpha (HIF-1alpha) did not enhance osteoclastogenesis and actually attenuated the differentiation process. DMOG did not affect cell viability or receptor activator of nuclear factor kappaB ligand (RANKL)-dependent phosphorylation of mitogen-activated protein (MAP) kinases. However, RANKL-dependent transcription of tartrate-resistant acid phosphatase (TRAP) was reduced in the presence of DMOG. Additionally, DMOG promoted transcription of the pro-apoptotic mediator B-Nip3. These studies suggest that a hypoxia-responsive factor other than HIF-1alpha is necessary for enhancing the formation of osteoclasts in hypoxic settings.


Subject(s)
Cell Differentiation/drug effects , Glycine , Osteoclasts , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Animals , Cell Line , Deferoxamine/pharmacology , Female , Gene Expression Regulation , Glycine/chemistry , Glycine/pharmacology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/drug effects , Osteoclasts/physiology , RANK Ligand/metabolism , Rats , Rats, Sprague-Dawley , Siderophores/pharmacology , Vascular Endothelial Growth Factor A/metabolism
10.
Hepatology ; 50(1): 85-93, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19444873

ABSTRACT

UNLABELLED: Steatosis in the liver is a common feature of obesity and type 2 diabetes and the precursor to the development of nonalcoholic steatohepatitis (NASH), cirrhosis, and liver failure. It has been shown previously that inhibiting glycosphingolipid (GSL) synthesis increases insulin sensitivity and lowers glucose levels in diabetic rodent models. Here we demonstrate that inhibiting GSL synthesis in ob/ob mice not only improved glucose homeostasis but also markedly reduced the development of hepatic steatosis. The ob/ob mice were treated for 7 weeks with a specific inhibitor of glucosylceramide synthase, the initial enzyme involved in the synthesis of GSLs. Besides lowering glucose and hemoglobin A1c (HbA1c) levels, drug treatment also significantly reduced the liver/body weight ratio, decreased the accumulation of triglycerides, and improved several markers of liver pathology. Drug treatment reduced liver glucosylceramide (GL1) levels in the ob/ob mouse. Treatment also reduced the expression of several genes associated with hepatic steatosis, including those involved in lipogenesis, gluconeogenesis, and inflammation. In addition, inhibiting GSL synthesis in diet-induced obese mice both prevented the development of steatosis and partially reversed preexisting steatosis. CONCLUSION: These data indicate that inhibiting GSL synthesis ameliorates the liver pathology associated with obesity and diabetes, and may represent a novel strategy for treating fatty liver disease and NASH.


Subject(s)
Dioxanes/pharmacology , Dioxanes/therapeutic use , Fatty Liver/metabolism , Glycosphingolipids/antagonists & inhibitors , Glycosphingolipids/biosynthesis , Obesity/drug therapy , Obesity/metabolism , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Animals , Male , Mice , Mice, Inbred C57BL
11.
Mol Ther ; 16(8): 1400-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18545223

ABSTRACT

Peripheral neuropathy is a particularly debilitating complication of both type 1 and type 2 diabetes characterized by sensory and motor neuron damage and decreased circulating levels of insulin-like growth factor 1 (IGF-1). Quite often, an early hyperalgesia is followed by hypoalgesia and muscle weakness. Hypoalgesia can lead to significant morbidity for which there is no current treatment. Hyperglycemic, streptozotocin (STZ)-induced rodent models reproduce these symptoms. We investigated whether increasing systemic IGF-1 could improve neuronal function in hyper- and hypoalgesic STZ-treated mice. Increased circulating levels of IGF-1 were achieved by delivering a plasmid or adeno-associated viral (AAV) vector bearing mouse IGF-1 to the liver. Treating mice in the hyperalgesia stage prevented later hypoalgesia. Treating mice in the hypoalgesia stage reversed existing hypoalgesia. This latter effect could be seen by merely restoring IGF-1 serum levels to normalcy, which was possible to achieve by IGF-1 gene therapy or insulin treatment. Sensory nerve functional correction was seen to be correlated with attenuated Schwann cell vacuolization and demyelination in peripheral sensory nerve fibers. A further increase in serum IGF-1 levels with gene therapy also improved motor function, consistent with the observed prevention of both muscle atrophy and peripheral motor nerve fiber demyelination. These results suggest that the restoration of systemic levels of IGF-1 may prove to be a highly effective therapeutic modality for treating diabetic peripheral neuropathy.


Subject(s)
Diabetic Neuropathies/therapy , Genetic Therapy/methods , Hyperalgesia/therapy , Insulin-Like Growth Factor I/physiology , Animals , Body Weight/physiology , Dependovirus/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/etiology , Diabetic Neuropathies/physiopathology , Disease Models, Animal , Genetic Vectors/genetics , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Neurons/cytology , Motor Neurons/metabolism , Motor Neurons/physiology
12.
Nat Biotechnol ; 26(5): 549-51, 2008 May.
Article in English | MEDLINE | ID: mdl-18438402

ABSTRACT

Pulmonary delivery of plasmid DNA (pDNA)/cationic liposome complexes is associated with an acute unmethylated CG dinucleotide (CpG)-mediated inflammatory response and brief duration of transgene expression. We demonstrate that retention of even a single CpG in pDNA is sufficient to elicit an inflammatory response, whereas CpG-free pDNA vectors do not. Using a CpG-free pDNA expression vector, we achieved sustained (>or=56 d) in vivo transgene expression in the absence of lung inflammation.


Subject(s)
CpG Islands/genetics , Gene Targeting/methods , Genetic Therapy/methods , Inflammation/genetics , Inflammation/prevention & control , Lung/metabolism , Plasmids/genetics , Plasmids/therapeutic use , Animals
13.
Diabetes ; 56(5): 1210-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17470562

ABSTRACT

Previous reports have shown that glycosphingolipids can modulate the activity of the insulin receptor, and studies in transgenic mice suggest a link between altered levels of various gangliosides and the development of insulin resistance. Here, we show that an inhibitor of glycosphingolipid synthesis can improve glucose control and increase insulin sensitivity in two different diabetic animal models. In the Zucker diabetic fatty rat, the glucosylceramide synthase inhibitor (1R,2R)-nonanoic acid[2-(2',3'-dihydro-benzo [1, 4] dioxin-6'-yl)-2-hydroxy-1-pyrrolidin-1-ylmethyl-ethyl]- amide-l-tartaric acid salt (Genz-123346) lowered glucose and A1C levels and improved glucose tolerance. Drug treatment also prevented the loss of pancreatic beta-cell function normally observed in the Zucker diabetic fatty rat and preserved the ability of the animals to secrete insulin. In the diet-induced obese mouse, treatment with Genz-123346 normalized A1C levels and improved glucose tolerance. Analysis of the phosphorylation state of the insulin receptor and downstream effectors showed increased insulin signaling in the muscles of the treated Zucker diabetic fatty rats and diet-induced obese mice. These results suggest that inhibiting glycosphingolipid synthesis can significantly improve insulin sensitivity and glucose homeostasis and may therefore represent a novel therapeutic approach for the treatment of type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/physiopathology , Dioxanes/therapeutic use , Glucosylceramides/metabolism , Glycosphingolipids/biosynthesis , Hypoglycemic Agents/therapeutic use , Insulin/blood , Pyrrolidines/therapeutic use , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/blood , Disease Models, Animal , Glycated Hemoglobin/metabolism , Lipids/blood , Liver/metabolism , Male , Obesity/blood , Rats , Rats, Zucker
14.
Adv Genet ; 53PA: 189-214, 2005.
Article in English | MEDLINE | ID: mdl-16243064

ABSTRACT

As with any conventional drug, the body's response to cationic lipid-DNA complexes is highly dependent on both the dose administered and the route of delivery. At relatively low doses there is little to no effect on organ function or tissue architecture, but at higher doses, acute inflammation and tissue damage can occur that is sometimes quite profound. Of the two most common routes of delivery, intravenous (IV) or intrapulmonary, IV administration tends to cause more severe adverse effects and can be lethal at higher doses of complex. Both routes activate an innate immune response that includes the induction of proinflammatory cytokines and immune cell activation, a major portion of which has been attributed to the presence of immunostimulatory CpG motifs within the plasmid DNA vector. Removing CpGs from the plasmid vector reduces several, but not all of the acute inflammatory responses to cationic lipid-DNA complexes. Therefore, other strategies are required to improve the therapeutic potential of these vectors, such as transient immune suppression, aerosolization of the complex, and novel formulations that have increased efficiency of transduction and decreased interaction with immune cells.

15.
Adv Genet ; 53: 189-214, 2005.
Article in English | MEDLINE | ID: mdl-16240994

ABSTRACT

As with any conventional drug, the body's response to cationic lipid-DNA complexes is highly dependent on both the dose administered and the route of delivery. At relatively low doses there is little to no effect on organ function or tissue architecture, but at higher doses, acute inflammation and tissue damage can occur that is sometimes quite profound. Of the two most common routes of delivery, intravenous (i.v.) or intrapulmonary, i.v. administration tends to cause more severe adverse effects and can be lethal at higher doses of complex. Both routes activate an innate immune response that includes the induction of proinflammatory cytokines and immune cell activation, a major portion of which has been attributed to the presence of immunostimulatory CpG motifs within the plasmid DNA vector. Removing CpGs from the plasmid vector reduces several, but not all of the acute inflammatory responses to cationic lipid-DNA complexes. Therefore, other strategies are required to improve the therapeutic potential of these vectors, such as transient immune suppression, aerosolization of the complex, and novel formulations that have increased efficiency of transduction and decreased interaction with immune cells.


Subject(s)
DNA/toxicity , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/toxicity , Inflammation/immunology , Lipids/toxicity , Animals , Blood/drug effects , CpG Islands/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/pharmacokinetics , Humans , Inflammation/chemically induced , Injections, Intravenous , Liver/drug effects , Lung/drug effects , Mice , Plasmids/genetics , Sheep
16.
Mol Ther ; 12(2): 264-73, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15946902

ABSTRACT

Hepatocytes are an effective depot for protein production from gene therapy vectors. However, when gene transfer vectors or their delivery induces hepatic inflammation, adaptive immune responses against the transgene product can ensue. In BALB/c mice, hydrodynamic delivery of a CMV-driven plasmid DNA (pDNA) bearing human alpha-galactosidase A (alphagal) to the liver generated antibodies against alphagal. This humoral immune response was more robust in a transgenic knockout for alphagal, the Fabry mouse. The antibody response could be attenuated in both mouse strains by using a promoter more restricted to hepatocytes. In an attempt to reduce further the humoral responses to alphagal, expression from the transgene was attenuated by using siRNA during the period of initial delivery-associated liver inflammation. In both mouse models and with both promoters, codelivering an alphagal siRNA resulted in a 2 log decrease in initial expression that then increased over the next few weeks to levels generated by the pDNA alone. This strategy led to both attenuated antibodies and an immune status approximating "tolerance" to alphagal. Importantly, in the Fabry mouse, an alphagal siRNA together with a hepatocyte-restricted promoter gave minimal anti-alphagal antibodies and profound tolerance, suggesting that such an approach might have clinical utility for genetic diseases.


Subject(s)
Hepatocytes/metabolism , Immune Tolerance/drug effects , RNA, Small Interfering/pharmacology , alpha-Galactosidase/immunology , Animals , Cell Communication , Disease Models, Animal , Fabry Disease , Gene Expression/drug effects , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Hepatocytes/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Plasmids/genetics , Promoter Regions, Genetic , RNA, Small Interfering/therapeutic use , Transfection , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
17.
Adv Drug Deliv Rev ; 57(5): 769-80, 2005 Apr 05.
Article in English | MEDLINE | ID: mdl-15757760

ABSTRACT

While a vast array of liposomes, peptides, and molecular conjugates have been evaluated for nonviral gene transfer, the entity containing the actual gene itself is almost always a plasmid. The layout of most plasmid DNA (pDNA) vectors is usually quite simple, consisting of a promoter, transgene, polyadenylation signal, and a backbone that permits propagation of the plasmid in bacteria. Additional sequence elements and modifications can be incorporated to influence the stability of gene expression and retention of the pDNA molecule in a given tissue. This review describes the different choices that can be made when designing a pDNA vector for transient, sustained, or regulated expression. The choice of promoter is a major determinant governing the kinetics of expression, but other factors, such as CpG content and the topological form of the pDNA are also influential. Vectors can also be designed to respond to the local environment of a given cell or tissue, or engineered to respond to a small molecule drug.


Subject(s)
Plasmids/chemical synthesis , Plasmids/pharmacokinetics , Transgenes/physiology , Animals , Gene Expression Regulation/genetics , Humans
18.
Mol Ther ; 9(2): 241-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14759808

ABSTRACT

Immunostimulatory CpG motifs have been implicated as a major contributor to the acute inflammatory response associated with nonviral vectors, most prominently seen after systemic delivery of cationic lipid-plasmid DNA (pDNA) complexes. We have shown previously that complexes containing pDNA vectors that have been largely depleted of CpG motifs have significantly reduced acute toxicity when delivered systemically. However, several CpGs remain in these vectors and the toxicity is not negligible, especially at higher doses of complex. To determine the maximal reduction in the acute toxic response that could be achieved by eliminating CpG signaling, we injected cationic lipid-pDNA complexes into transgenic mice that are deficient in Toll-like receptor 9 (TLR9), which is the receptor that recognizes immunostimulatory CpG motifs. We observed significantly decreased adverse hematological changes and liver damage in TLR9(-/-) mice compared to normal mice and increased survival at higher doses of complex. However, a pronounced loss of lymphocytes and platelets was still observed in the TLR9(-/-) mice at higher doses. We also measured the toxicity in normal mice of systemically delivered complexes containing non-CpG oligonucleotides. Although serum transaminase levels were reduced, a loss of lymphocytes and platelets akin to that seen in the TLR9(-/-) mice was observed. Taken together, these findings suggest that signaling through TLR9 contributes to the majority but not all of the toxic responses associated with systemic delivery of cationic lipid-pDNA complexes.


Subject(s)
DNA-Binding Proteins/metabolism , Genetic Vectors/toxicity , Inflammation/chemically induced , Inflammation/metabolism , Receptors, Cell Surface/metabolism , Acute Disease , Animals , Cations/administration & dosage , Cations/metabolism , CpG Islands/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/metabolism , Inflammation/genetics , Lipid Metabolism , Lipids/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Oligonucleotides/administration & dosage , Oligonucleotides/genetics , Oligonucleotides/metabolism , Oligonucleotides/toxicity , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Sequence Deletion , Signal Transduction/drug effects , Survival Rate , Toll-Like Receptor 9
19.
J Gene Med ; 6(1): 85-92, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14716680

ABSTRACT

BACKGROUND: Fabry disease is a recessive, X-linked disorder caused by a deficiency of the lysosomal enzyme alpha-galactosidase A, leading to an accumulation of the glycosphingolipid globotriaosylceramide (GL-3) in most tissues of the body. The goal of this study was to determine if systemic delivery of a nonviral vector could correct the enzyme deficiency and reduce the levels of GL-3 in different tissues of a transgenic knockout mouse model of the disease. METHODS: Cationic lipid was complexed with a CpG-depleted plasmid DNA vector and then injected intravenously into Fabry mice. The levels of alpha-galactosidase A and GL-3 in different tissues were assayed at various time points after injection. RESULTS: Expression of alpha-galactosidase A was detected in the different tissues of Fabry mice for up to 3 months after complex administration, but resulted in minimal reductions in GL-3 levels. However, the use of the anti-inflammatory drug dexamethasone and multiple dosing increased alpha-galactosidase A expression and resulted in significant reductions of GL-3 in all the organs with the exception of the kidney. In addition, injecting complex into young Fabry mice partially prevented the normal accumulation of GL-3 in the heart, lung, and liver. CONCLUSIONS: Systemic delivery of a cationic lipid-pDNA complex partially corrected the enzyme deficiency and reduced glycolipid storage in a mouse model of Fabry disease. The results are one of the few demonstrations of long-term efficacy in a genetic disease model using nonviral vectors. However, substantial improvements in expression, especially in critical organs such as the kidney, are required before these vectors can become a viable approach to treat Fabry disease and other lysosomal storage disorders.


Subject(s)
Fabry Disease/genetics , Fabry Disease/therapy , Genetic Therapy , Genetic Vectors , Trihexosylceramides/metabolism , alpha-Galactosidase/genetics , Animals , Disease Models, Animal , Female , Gene Expression , Lipids , Mice , Mice, Inbred BALB C , Plasmids/genetics , alpha-Galactosidase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...