Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Sci Rep ; 13(1): 22392, 2023 12 16.
Article in English | MEDLINE | ID: mdl-38104212

ABSTRACT

Cellular homeostasis of creatine (CT), integral part of the energy buffering and transducing system connecting intracellular sites of ATP production and utilization, comprises of mechanisms that increase CT, i.e., biosynthesis and cellular uptake, and CT-lowering processes, such as export and non-enzymatic conversion to creatinine. The biosynthesis of CT is controlled by negative feedback loop via suppression of the rate-limiting enzyme arginine:glycine amidinotransferase (AGAT). Although the regulatory mechanism involved is not well understood, AGAT suppression is successfully used in patients with guanidinoacetate methyltransferase (GAMT) deficiency to reduce the neurotoxic accumulation of the AGAT-mediated guanidinoacetate production by supplementing patients with CT. Utilizing the CT-dependent feedback loop for the upregulation of AGAT expression may well represent a therapeutic target for an additional CT deficiency syndrome, the CT transporter (CrT) defect, for which no effective treatment option is available so far. We have used CRISPR to tag the C-terminus of AGAT with a nanoluc luciferase (NLuc) reporter in HAP1 cells. A biphasic decay of AGAT-NLuc in response to increasing extracellular CT was observed, whereas the decrease in AGAT-NLuc expression was directly proportional to the rise in intracellular CT levels with an approximate IC50 of 1-2 mM. CRISPR generated HAP1 CrT null cells and HAP1 CrT null cells stably expressing a CrT-GFP fusion protein further demonstrated that the biphasic response to extracellular CT is mediated by a high-affinity (Km 9-10 µM) CrT dependent, saturable mechanism and a CrT independent, unsaturable uptake process. The direct response to intracellular CT suggests the existence of an intracellular CT sensing system enabling a dynamic cell response to changing CT concentration that is relevant for cellular CT homeostasis.


Subject(s)
Amidinotransferases , Language Development Disorders , Movement Disorders , Humans , Amidinotransferases/metabolism , Creatine/metabolism , Guanidinoacetate N-Methyltransferase/genetics
2.
Genet Med ; 24(11): 2399-2407, 2022 11.
Article in English | MEDLINE | ID: mdl-36083289

ABSTRACT

PURPOSE: RABGAP1 is a GTPase-activating protein implicated in a variety of cellular and molecular processes, including mitosis, cell migration, vesicular trafficking, and mTOR signaling. There are no known Mendelian diseases caused by variants in RABGAP1. METHODS: Through GeneMatcher, we identified 5 patients from 3 unrelated families with homozygous variants in the RABGAP1 gene found on exome sequencing. We established lymphoblastoid cells lines derived from an affected individual and her parents and performed RNA sequencing and functional studies. Rabgap1 knockout mice were generated and phenotyped. RESULTS: We report 5 patients presenting with a common constellation of features, including global developmental delay/intellectual disability, microcephaly, bilateral sensorineural hearing loss, and seizures, as well as overlapping dysmorphic features. Neuroimaging revealed common features, including delayed myelination, white matter volume loss, ventriculomegaly, and thinning of the corpus callosum. Functional analysis of patient cells revealed downregulated mTOR signaling and abnormal localization of early endosomes and lysosomes. Rabgap1 knockout mice exhibited several features in common with the patient cohort, including microcephaly, thinning of the corpus callosum, and ventriculomegaly. CONCLUSION: Collectively, our results provide evidence of a novel neurodevelopmental syndrome caused by biallelic loss-of-function variants in RABGAP1.


Subject(s)
Hydrocephalus , Intellectual Disability , Microcephaly , Neurodevelopmental Disorders , Animals , Mice , Female , Humans , Microcephaly/genetics , Pedigree , Intellectual Disability/genetics , Syndrome , Mice, Knockout , TOR Serine-Threonine Kinases , Neurodevelopmental Disorders/genetics
3.
Glycobiology ; 31(9): 1093-1101, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34080004

ABSTRACT

N-Deacetylase/N-sulfotransferases (NDSTs) are critical enzymes in heparan sulfate (HS) biosynthesis. Radioactive labeling assays are the preferred methods to determine the N-sulfotransferase activity of NDST. In this study, we developed a fluorometric coupled enzyme assay that is suitable for the study of enzyme kinetics and inhibitory properties of drug candidates derived from a large-scale in silico screening targeting the sulfotransferase moiety of NDST1. The assay measures recombinant mouse NDST1 (mNDST1) sulfotransferase activity by employing its natural substrate adenosine 3'-phophoadenosine-5'-phosphosulfate (PAPS), a bacterial analog of desulphated human HS, Escherichia coli K5 capsular polysaccharide (K5), the fluorogenic substrate 4-methylumbelliferylsulfate and a double mutant of rat phenol sulfotransferase SULT1A1 K56ER68G. Enzyme kinetic analysis of mNDST1 performed with the coupled assay under steady state conditions at pH 6.8 and 37°C revealed Km (K5) 34.8 µM, Km (PAPS) 10.7 µM, Vmax (K5) 0.53 ± 0.13 nmol/min/µg enzyme, Vmax (PAPS) 0.69 ± 0.05 nmol/min/µg enzyme and the specific enzyme activity of 394 pmol/min/µg enzyme. The pH optimum of mNDST1 is pH 8.2. Our data indicate that mNDST1 is specific for K5 substrate. Finally, we showed that the mNDST1 coupled assay can be utilized to assess potential enzyme inhibitors for drug development.


Subject(s)
Heparitin Sulfate , Sulfotransferases , Animals , Enzyme Assays , Enzyme Inhibitors/pharmacology , Heparitin Sulfate/chemistry , Kinetics , Mice , Rats , Sulfotransferases/metabolism
4.
Hum Gene Ther ; 27(7): 509-21, 2016 07.
Article in English | MEDLINE | ID: mdl-27197548

ABSTRACT

GM2 gangliosidosis is a family of three genetic neurodegenerative disorders caused by the accumulation of GM2 ganglioside (GM2) in neuronal tissue. Two of these are due to the deficiency of the heterodimeric (α-ß), "A" isoenzyme of lysosomal ß-hexosaminidase (HexA). Mutations in the α-subunit (encoded by HEXA) lead to Tay-Sachs disease (TSD), whereas mutations in the ß-subunit (encoded by HEXB) lead to Sandhoff disease (SD). The third form results from a deficiency of the GM2 activator protein (GM2AP), a substrate-specific cofactor for HexA. In their infantile, acute forms, these diseases rapidly progress with mental and psychomotor deterioration resulting in death by approximately 4 years of age. After gene transfer that overexpresses one of the deficient subunits, the amount of HexA heterodimer formed would empirically be limited by the availability of the other endogenous Hex subunit. The present study used a new variant of the human HexA α-subunit, µ, incorporating critical sequences from the ß-subunit that produce a stable homodimer (HexM) and promote functional interactions with the GM2AP- GM2 complex. We report the design of a compact adeno-associated viral (AAV) genome using a synthetic promoter-intron combination to allow self-complementary (sc) packaging of the HEXM gene. Also, a previously published capsid mutant, AAV9.47, was used to deliver the gene to brain and spinal cord while having restricted biodistribution to the liver. The novel capsid and cassette design combination was characterized in vivo in TSD mice for its ability to efficiently transduce cells in the central nervous system when delivered intravenously in both adult and neonatal mice. This study demonstrates that the modified HexM is capable of degrading long-standing GM2 storage in mice, and it further demonstrates the potential of this novel scAAV vector design to facilitate widespread distribution of the HEXM gene or potentially other similar-sized genes to the nervous system.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Hexosaminidases/genetics , Mutation/genetics , Tay-Sachs Disease/therapy , Animals , Animals, Newborn , Disease Models, Animal , Female , G(M2) Ganglioside/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Tay-Sachs Disease/genetics
5.
Mol Ther Methods Clin Dev ; 3: 15057, 2016.
Article in English | MEDLINE | ID: mdl-26966698

ABSTRACT

Tay-Sachs or Sandhoff disease result from mutations in either the evolutionarily related HEXA or HEXB genes encoding respectively, the α- or ß-subunits of ß-hexosaminidase A (HexA). Of the three Hex isozymes, only HexA can interact with its cofactor, the GM2 activator protein (GM2AP), and hydrolyze GM2 ganglioside. A major impediment to establishing gene or enzyme replacement therapy based on HexA is the need to synthesize both subunits. Thus, we combined the critical features of both α- and ß-subunits into a single hybrid µ-subunit that contains the α-subunit active site, the stable ß-subunit interface and unique areas in each subunit needed to interact with GM2AP. To facilitate intracellular analysis and the purification of the µ-homodimer (HexM), CRISPR-based genome editing was used to disrupt the HEXA and HEXB genes in a Human Embryonic Kidney 293 cell line stably expressing the µ-subunit. In association with GM2AP, HexM was shown to hydrolyze a fluorescent GM2 ganglioside derivative both in cellulo and in vitro. Gene transfer studies in both Tay-Sachs and Sandhoff mouse models demonstrated that HexM expression reduced brain GM2 ganglioside levels.

6.
J Med Chem ; 58(11): 4483-93, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25984755

ABSTRACT

In order to identify structural features of pyrimethamine (5-(4-chlorophenyl)-6-ethylpyrimidine-2,4-diamine) that contribute to its inhibitory activity (IC50 value) and chaperoning efficacy toward ß-N-acetylhexosaminidase, derivatives of the compound were synthesized that differ at the positions bearing the amino, ethyl, and chloro groups. Whereas the amino groups proved to be critical to its inhibitory activity, a variety of substitutions at the chloro position only increased its IC50 by 2-3-fold. Replacing the ethyl group at the 6-position with butyl or methyl groups increased IC50 more than 10-fold. Surprisingly, despite its higher IC50, a derivative lacking the chlorine atom in the para-position was found to enhance enzyme activity in live patient cells a further 25% at concentrations >100 µM, while showing less toxicity. These findings demonstrate the importance of the phenyl group in modulating the chaperoning efficacy and toxicity profile of the derivatives.


Subject(s)
Mutant Proteins/metabolism , Mutation/genetics , Pyrimethamine/chemistry , Pyrimethamine/metabolism , beta-N-Acetylhexosaminidases/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Models, Molecular , Molecular Structure , Mutant Proteins/genetics , Structure-Activity Relationship
7.
PLoS One ; 9(10): e109128, 2014.
Article in English | MEDLINE | ID: mdl-25343256

ABSTRACT

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a myocardial disease characterized by fibro-fatty replacement of myocardium in the right ventricular free wall and frequently results in life-threatening ventricular arrhythmias and sudden cardiac death. A heterozygous missense mutation in the transmembrane protein 43 (TMEM43) gene, p.S358L, has been genetically identified to cause autosomal dominant ARVC type 5 in a founder population from the island of Newfoundland, Canada. Little is known about the function of the TMEM43 protein or how it leads to the pathogenesis of ARVC. We sought to determine the distribution of TMEM43 and the effect of the p.S358L mutation on the expression and distribution of various intercalated (IC) disc proteins as well as functional effects on IC disc gap junction dye transfer and conduction velocity in cell culture. Through Western blot analysis, transmission electron microscopy (TEM), immunofluorescence (IF), and electrophysiological analysis, our results showed that the stable expression of p.S358L mutation in the HL-1 cardiac cell line resulted in decreased Zonula Occludens (ZO-1) expression and the loss of ZO-1 localization to cell-cell junctions. Junctional Plakoglobin (JUP) and α-catenin proteins were redistributed to the cytoplasm with decreased localization to cell-cell junctions. Connexin-43 (Cx43) phosphorylation was altered, and there was reduced gap junction dye transfer and conduction velocity in mutant TMEM43-transfected cells. These observations suggest that expression of the p.S358L mutant of TMEM43 found in ARVC type 5 may affect localization of proteins involved in conduction, alter gap junction function and reduce conduction velocity in cardiac tissue.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Endoplasmic Reticulum/genetics , Membrane Proteins/biosynthesis , Arrhythmogenic Right Ventricular Dysplasia/physiopathology , Cytoplasm , Desmosomes/metabolism , Gap Junctions/genetics , Gene Expression Regulation , Humans , Membrane Proteins/genetics , Mutation, Missense , Myocardium/metabolism , Myocardium/pathology , Phosphorylation
8.
J Org Chem ; 79(10): 4398-404, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24735108

ABSTRACT

We report herein a newly developed domino reaction that facilitates the synthesis of new 1,5-dideoxy-1,5-iminoribitol iminosugar C-glycosides 7a-e and 8. The key intermediate in this approach is a six-membered cyclic sugar nitrone that is generated in situ and trapped by an alkene dipolarophile via a [2 + 3] cycloaddition reaction to give the corresponding isooxazolidines 10a-e in a "one-pot" protocol. The iminoribitol C-glycosides 7a-e and 8 were found to be modest ß-galactosidase (bGal) inhibitors. However, compounds 7c and 7e showed "pharmacological chaperone" activity for mutant lysosomal bGal activity and facilitated its recovery in GM1 gangliosidosis patient fibroblasts by 2-6-fold.


Subject(s)
Alkenes/chemistry , Fibroblasts/chemistry , Gangliosidosis, GM1/drug therapy , Lysosomes/chemistry , Molecular Chaperones/pharmacology , Molecular Chaperones/therapeutic use , Monosaccharides/chemical synthesis , Nitrogen Oxides/chemistry , beta-Galactosidase/antagonists & inhibitors , beta-Galactosidase/chemistry , Cycloaddition Reaction , Gangliosidosis, GM1/enzymology , Gangliosidosis, GM1/metabolism , Glycosides , Humans , Lysosomes/metabolism , Monosaccharides/chemistry
9.
Basic Res Cardiol ; 109(3): 406, 2014 May.
Article in English | MEDLINE | ID: mdl-24604614

ABSTRACT

Remote ischemic preconditioning (rIPC) induced by transient limb ischemia (li-rIPC) leads to neurally dependent release of blood-borne factors that provide potent cardioprotection. We hypothesized that transcutaneous electrical nerve stimulation (TENS) is a clinically relevant stimulus of rIPC. Study 1: seven rabbits were subjected to lower limb TENS; six to li-rIPC, and six to sham intervention. Blood was drawn and used to prepare a dialysate for subsequent analysis of cardioprotection in rabbit Langendorff preparation. Study 2: 14 healthy adults underwent upper limb TENS stimulation on one study day, 10 of whom also underwent li-rIPC on another study day. Blood was drawn before and after each stimulus, dialysate prepared, and cardioprotective activity assessed in mouse Langendorff preparation. The infarct size and myocardial recovery were measured after 30 min of global ischemia and 60 or 120 min of reperfusion. Animal validation: compared to control, TENS induced marked cardioprotection with significantly reduced infarct size (TENS vs. sham p < 0.01, rIPC vs. sham p < 0.01, TENS vs. rIPC p = ns) and improved functional recovery during reperfusion. Human study: compared to baseline, dialysate after rIPC (pre-rIPC vs. post-rIPC, p < 0.001) and TENS provided potent cardioprotection (pre-TENS vs. post-TENS p < 0.001) and improved myocardial recovery during reperfusion. The cardioprotective effects of TENS dialysates were blocked by pretreatment of the receptor heart with the opioid antagonist naloxone. TENS is a novel method for inducing cardioprotection and may provide an alternative to the limb ischemia stimulus for induction of rIPC clinically.


Subject(s)
Hindlimb/blood supply , Ischemic Preconditioning/methods , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Transcutaneous Electric Nerve Stimulation , Upper Extremity/blood supply , Adult , Animals , Biomarkers/blood , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/blood , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Narcotic Antagonists/pharmacology , Rabbits , Regional Blood Flow , Time Factors , Ventricular Function, Left , Ventricular Pressure
10.
Cardiovasc Drugs Ther ; 28(1): 7-17, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24018748

ABSTRACT

BACKGROUND: Coronary effluent from an isolated perfused heart undergoing ischemic preconditioning can be transferred to precondition another naïve isolated heart. We investigated the effects of this effluent on mitochondrial integrity and function following a global infarct model of ischemia/reperfusion and the role of adenosine in this model of remote preconditioning. METHODS AND RESULTS: Coronary effluent from isolated perfused rabbit hearts was collected prior to (control effluent) and during three cycles of 5-min ischemia and 10-min reperfusion (IPC effluent). Adenosine concentration was significantly increased in IPC effluent (2.6 ± 1.1 µM) versus control effluent (0.21 ± 0.06 µM, P < 0.01). Infarct size (% necrotic LV mass) after 30-min global ischemia and 90-min reperfusion was significantly reduced in hearts preconditioned with IPC effluent (IPC(eff), 23 ± 7 %) and control effluent supplemented with 2.5 µM exogenous adenosine (C(eff)+ 2.5 µM ADO, 25 ± 10 %) when compared to control effluent perfused hearts (C(eff), 41 ± 8 %, P < 0.05). Compared to C(eff) mitochondria, IPC(eff) mitochondria had preserved complex I/State3 and complex IV/State 3 respiration and outer membrane integrity, and reduced cytochrome c release. In contrast, C(eff) + 2.5 µM ADO mitochondria had improved state 2 respiration and coupling to oxidative phosphorylation, reduced reactive oxygen species production and preserved outer membrane integrity. Administration of adenosine receptor blocker 8-(p-sulfophenyl)theophylline abolished the infarct limiting effect (46 ± 7 %) and the mitochondrial integrity and function preservation of IPC effluent. CONCLUSION: Remote cardioprotection by IPC effluent preserves mitochondrial integrity and function in an adenosine receptor dependent mechanism, and although infarct size reduction can be mimicked by adenosine, IPC effluent contains additional factor(s) contributing to modulation of the mitochondrial response to ischemia/reperfusion injury.


Subject(s)
Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/physiopathology , Receptors, Purinergic P1/metabolism , Adenosine/metabolism , Animals , Disease Models, Animal , Male , Mitochondria/metabolism , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Rabbits , Reactive Oxygen Species/metabolism , Theophylline/analogs & derivatives , Theophylline/pharmacology
11.
J Mol Cell Cardiol ; 60: 142-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23608604

ABSTRACT

Multiple initiatives are underway to harness the clinical benefits of remote ischemic preconditioning (rIPC) based on applying non-invasive, brief, intermittent limb ischemia/reperfusion using an external occluder. However, little is known about how rIPC induces protection in cardiomyocytes, particularly through G-protein coupled receptors. In these studies, we determined the role of opioid and adenosine receptors and their functional interactions in rIPC cardioprotection. In freshly isolated cardiomyocytes subjected to 45-min simulated ischemia followed by 60-min simulated reperfusion, we examined the ability of plasma dialysate (derived from blood obtained from rabbits remotely preconditioned by application of brief cycles of hind limb ischemia/reperfusion, rIPC dialysate) to protect cells against necrosis. rIPC dialysate and selective activation of either δ-opioid receptors or κ-opioid receptors significantly reduced the % of dead cells after simulated ischemia and simulated reperfusion. Inhibition of adenosine A1 receptors, but not adenosine A3 receptors, blocked the protection by rIPC dialysate, δ-opioid receptor and κ-opioid receptor activation. In HEK293 cells expressing either hemagglutinin A-tagged δ-opioid receptors or hemagglutinin A-tagged κ-opioid receptors, selective immunoprecipitation of adenosine A1 receptors pulled down both δ-opioid and κ-opioid receptors. This molecular association of adenosine A1 receptors with δ-opioid and κ-opioid receptors was confirmed by reverse pull-down assays. These findings strongly suggest that rIPC cardioprotection requires the activation of δ-opioid and κ-opioid receptors and relies on these receptors functionally interacting with adenosine A1 receptors.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocytes, Cardiac/metabolism , Receptor, Adenosine A1/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Animals , HEK293 Cells , Humans , Myocytes, Cardiac/pathology , Rabbits
12.
Mol Genet Metab ; 107(1-2): 203-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22784478

ABSTRACT

Deficiencies of lysosomal ß-D-galactosidase can result in GM1 gangliosidosis, a severe neurodegenerative disease characterized by massive neuronal storage of GM1 ganglioside in the brain. Currently there are no available therapies that can even slow the progression of this disease. Enzyme enhancement therapy utilizes small molecules that can often cross the blood brain barrier, but are also often competitive inhibitors of their target enzyme. It is a promising new approach for treating diseases, often caused by missense mutations, associated with dramatically reduced levels of functionally folded enzyme. Despite a number of positive reports based on assays performed with patient cells, skepticism persists that an inhibitor-based treatment can increase mutant enzyme activity in vivo. To date no appropriate animal model, i.e., one that recapitulates a responsive human genotype and clinical phenotype, has been reported that could be used to validate enzyme enhancement therapy. In this report, we identify a novel enzyme enhancement-agent, N-nonyl-deoxygalactonojirimycin, that enhances the mutant ß-galactosidase activity in the lysosomes of a number of patient cell lines containing a variety of missense mutations. We then demonstrate that treatment of cells from a previously described, naturally occurring feline model (that biochemically, clinically and molecularly closely mimics GM1 gangliosidosis in humans) with this molecule, results in a robust enhancement of their mutant lysosomal ß-galactosidase activity. These data indicate that the feline model could be used to validate this therapeutic approach and determine the relationship between the disease stage at which this therapy is initiated and the maximum clinical benefits obtainable.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enzyme Replacement Therapy , Gangliosidosis, GM1/metabolism , Mutant Proteins/metabolism , beta-Galactosidase/metabolism , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/pharmacology , Animals , Cats , Cell Line, Tumor , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Gangliosidosis, GM1/drug therapy , Gangliosidosis, GM1/genetics , Hot Temperature , Humans , Hydrogen-Ion Concentration , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutation , Protein Denaturation/drug effects , Treatment Outcome , beta-Galactosidase/antagonists & inhibitors , beta-Galactosidase/chemistry
13.
Mol Genet Metab ; 106(3): 323-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22592100

ABSTRACT

Gaucher disease is a prevalent lysosomal storage disease in which affected individuals inherit mutations in the gene (GBA1) encoding lysosomal acid ß-glucosidase (glucocerebrosidase, GCase, EC 3.2.1.45). One of the most prevalent disease-causing mutations in humans is a N370S missense mutation in the GCase protein. As part of a larger endeavor to study the fate of mutant human proteins expressed in plant cells, the N370S mutant protein along with the wild-type- (WT)-GCase, both equipped with a signal peptide, were synthesized in transgenic tobacco BY2 cells, which do not possess lysosomes. The enzymatic activity of plant-recombinant N370S GCase lines was significantly lower (by 81-95%) than that of the WT-GCase lines. In contrast to the WT-GCase protein, which was efficiently secreted from tobacco BY2 cells, and detected in large amounts in the culture medium, only a small proportion of the N370S GCase was secreted. Pharmacological chaperones such as N-(n-nonyl) deoxynojirimycin and ambroxol increased the steady-state mutant protein levels both inside the plant cells and in the culture medium. These findings contradict the assertion that small molecule chaperones increase N370S GCase activity (as assayed in treated patient cell lysates) by stabilizing the enzyme in the lysosome, and suggest that the mutant protein is impaired in its ability to obtain its functional folded conformation, which is a requirement for exiting the lumen of the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Glucosylceramidase/genetics , Molecular Chaperones/metabolism , Biological Transport , Catalytic Domain , Cells, Cultured , Gaucher Disease/enzymology , Gaucher Disease/genetics , Glucosylceramidase/metabolism , Humans , Molecular Chaperones/genetics , Mutation , Plant Cells/metabolism , Plants, Genetically Modified , Protein Folding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
J Med Chem ; 55(6): 2737-45, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22360565

ABSTRACT

A highly divergent route to lipophilic iminosugars that utilizes the thiol-ene reaction was developed to enable the rapid synthesis of a collection of 16 dideoxyiminoxylitols bearing various different lipophilic substituents. Enzyme kinetic analyses revealed that a number of these products are potent, low-nanomolar inhibitors of human glucocerebrosidase that stabilize the enzyme to thermal denaturation by up to 20 K. Cell based assays conducted on Gaucher disease patient derived fibroblasts demonstrated that administration of the compounds can increase lysosomal glucocerebrosidase activity levels by therapeutically relevant amounts, as much as 3.2-fold in cells homozygous for the p.N370S mutation and 1.4-fold in cells homozygous for the p.L444P mutation. Several compounds elicited this increase in enzyme activity over a relatively wide dosage range. The data assembled here illustrate how the lipophilic moiety common to many glucocerebrosidase inhibitors might be used to optimize a lead compound's ability to chaperone the protein in cellulo. The flexibility of this synthetic strategy makes it an attractive approach to the rapid optimization of glycosidase inhibitor potency and pharmacokinetic behavior.


Subject(s)
Allylamine/analogs & derivatives , Allylamine/chemical synthesis , Carbohydrates/chemical synthesis , Gaucher Disease/drug therapy , Glucosylceramidase/antagonists & inhibitors , Imines/chemical synthesis , Xylitol/analogs & derivatives , Xylitol/chemical synthesis , Allylamine/pharmacology , Carbohydrates/pharmacology , Enzyme Assays , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Gaucher Disease/enzymology , Gaucher Disease/pathology , Glucosylceramidase/genetics , Humans , Imines/pharmacology , Isomerism , Lysosomes/drug effects , Lysosomes/enzymology , Mutation , Small Molecule Libraries , Structure-Activity Relationship , Xylitol/pharmacology
15.
Basic Res Cardiol ; 107(2): 241, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22231674

ABSTRACT

We have previously shown that remote ischemic preconditioning by limb ischemia (rIPC) or intra-arterial adenosine releases a dialyzable cardioprotective circulating factor(s), the release of which requires an intact neural connection to the limb and is blocked by pretreatment with S-nitroso-N-acetylpenicillamine (SNAP). Remote cardioprotection can be induced by other forms of peripheral stimulation including topical capsaicin, but the mechanisms of their signal transduction are incompletely understood. Rabbits were anesthetized by intravenous pentobarbital, intubated and ventilated, then randomized (4-7 animals in each group) to receive sham procedure, rIPC (4 cycles of 5 min lower limb ischemia, 5 min reperfusion), direct femoral nerve stimulation, topical capsaicin, pretreatment with intra-arterial SNAP + capsaicin, pretreatment with topical DMSO (a sensory nerve blocker) + topical capsaicin, or pretreatment with intra-arterial SNAP + femoral nerve stimulation, topical DMSO alone, or intra-arterial SNAP alone. Blood was then rapidly drawn from the carotid artery to produce the plasma dialysate which was used to perfuse a naïve heart from an untreated donor rabbit. The infarct size and recovery of LV-developed pressure and end-diastolic pressure were measured after 30 min of global ischemia and 120 min of reperfusion. Compared to sham, dialysate from rIPC, femoral nerve stimulation, and topical capsaicin groups all produced significant cardioprotection with significantly reduced infarct size, and improved the post-ischemic cardiac performance. Cardioprotection was not seen in the topical DMSO-capsaicin, SNAP + capsaicin, and SNAP + FNS groups. These results confirm the central role of peripheral nerves in the local signal transduction of remote cardioprotection. Direct electrical or peripheral neural stimulation evokes the release of cardioprotective substances into the bloodstream, with comparable effects to that of rIPC induced by limb ischemia.


Subject(s)
Capsaicin/administration & dosage , Ischemic Preconditioning/methods , Myocardial Reperfusion Injury/prevention & control , Peripheral Nervous System/drug effects , Animals , Femoral Nerve/physiology , Hemodynamics , Ischemic Preconditioning, Myocardial/methods , Peripheral Nervous System/physiology , Rabbits
16.
Glycobiology ; 22(4): 492-503, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22061999

ABSTRACT

There is a clear need for efficient methods to produce protein therapeutics requiring mannose-termination for therapeutic efficacy. Here we report on a unique system for production of active human lysosomal acid ß-glucosidase (glucocerebrosidase, GCase, EC 3.2.1.45) using seeds of the Arabidopsis thaliana complex-glycan-deficient (cgl) mutant, which are deficient in the activity of N-acetylglucosaminyl transferase I (EC 2.4.1.101). Gaucher disease is a prevalent lysosomal storage disease in which affected individuals inherit mutations in the gene (GBA1) encoding GCase. A gene cassette optimized for seed expression was used to generate the human enzyme in seeds of the cgl (C5) mutant, and the recombinant GCase was mainly accumulated in the apoplast. Importantly, the enzymatic properties including kinetic parameters, half-maximal inhibitory concentration of isofagomine and thermal stability of the cgl-derived GCase were comparable with those of imiglucerase, a commercially available recombinant human GCase used for enzyme replacement therapy in Gaucher patients. N-glycan structural analyses of recombinant cgl-GCase showed that the majority of the N-glycans (97%) were mannose terminated. Additional purification was required to remove ∼15% of the plant-derived recombinant GCase that possessed potentially immunogenic (xylose- and/or fucose-containing) N-glycans. Uptake of cgl-derived GCase by mouse macrophages was similar to that of imiglucerase. The cgl seed system requires no addition of foreign (non-native) amino acids to the mature recombinant GCase protein, and the dry transgenic seeds represent a stable repository of the therapeutic protein. Other strategies that may completely prevent plant-like complex N-glycans are discussed, including the use of a null cgl mutant.


Subject(s)
Arabidopsis/genetics , Glucosylceramidase/biosynthesis , Plants, Genetically Modified , Seeds/genetics , Animals , Arabidopsis/cytology , Carbohydrate Conformation , Carbohydrate Sequence , Cells, Cultured , Enzyme Stability , Glucosylceramidase/isolation & purification , Glucosylceramidase/metabolism , Glycosylation , Humans , Kinetics , Macrophages/metabolism , Mannose , Mice , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Seeds/cytology
19.
Carbohydr Res ; 346(12): 1592-8, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21645885

ABSTRACT

Two simple and reliably accessible intermediates, N-carboxypentyl- and N-aminohexyl-1-deoxy-D-galactonojirimycin were employed for the synthesis of a set of terminally N-dansyl substituted derivatives. Reaction of the terminal carboxylic acid of N-carboxypentyl-1-deoxy-D-galactonojirimycin with N-dansyl-1,6-diaminohexane provided the chain-extended fluorescent derivative. Employing bis(6-dansylaminohexyl)amine, the corresponding branched di-N-dansyl compound was obtained. Partially protected N-aminohexyl-1-deoxy-D-galactonojirimycin served as intermediate for two additional chain-extended fluorescent 1-deoxy-D-galactonojirimycin (1-DGJ) derivatives featuring terminal dansyl groups in the N-alkyl substituent. These new compounds are strong inhibitors of d-galactosidases and may serve as leads en route to pharmacological chaperones for GM1-gangliosidosis.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Bacterial Proteins/metabolism , Dansyl Compounds/chemistry , Enzyme Inhibitors/pharmacology , Gangliosidosis, GM1/enzymology , Phosphatidylcholines/chemistry , Plant Proteins/metabolism , beta-Galactosidase , 1-Deoxynojirimycin/chemical synthesis , 1-Deoxynojirimycin/pharmacology , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Cats , Cell Line , Diamines/chemistry , Enzyme Inhibitors/chemical synthesis , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/pharmacology , Gangliosidosis, GM1/drug therapy , Gangliosidosis, GM1/physiopathology , Humans , Imines/chemistry , Kinetics , Lysosomes/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/pharmacology , Molecular Probes/chemical synthesis , Molecular Probes/pharmacology , Molecular Targeted Therapy , Plant Proteins/antagonists & inhibitors , Plant Proteins/chemistry , Sugar Alcohols/chemistry , beta-Galactosidase/antagonists & inhibitors , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...