Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
Curr Drug Discov Technol ; 5(1): 39-48, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18537566

ABSTRACT

Site-specific drug delivery to bone is considered to be achievable by utilizing acidic amino acid homopeptides. We found that fluorescence-labeled acidic amino acid (L-Asp or L-Glu) homopeptides containing six or more residues bound strongly to hydroxyapatite, which is a major component of bone, and were selectively delivered to and retained in bone after systemic administration. We explored the applicability of this result for drug delivery by conjugation of estradiol and levofloxacin with an L-Asp hexapeptide. We also similarly tagged an enzyme, tissue-nonspecific alkaline phosphatase, to see whether this would improve the efficacy of enzyme replacement therapy. The L-Asp hexapeptide-tagged drugs, including the enzyme, were selectively delivered to bone in comparison with the untagged drugs. It was expected that the ester linkage to the hexapeptide would be susceptible to hydrolysis in situ, releasing the drug or enzyme from the acidic oligopeptide. An in vivo experiment confirmed the efficacy of L-Asp hexapeptide-tagged estradiol and levofloxacin, although there was some loss of bioactivity of estradiol and levofloxacin in vitro, suggesting that the acidic hexapeptide was partly removed by hydrolysis in the body after delivery to bone. The adverse effect of estradiol on the uterus was greatly reduced by conjugation to the hexapeptide. These results support the usefulness of acidic oligopeptides as bone-targeting carriers for therapeutic agents. We present some pharmacokinetic and pharmacological properties of the L-Asp hexapeptide-tagged drugs and enzyme.


Subject(s)
Bone and Bones/metabolism , Oligopeptides/pharmacology , Oligopeptides/pharmacokinetics , Pharmaceutical Preparations/administration & dosage , Acids , Alkaline Phosphatase/chemistry , Alkaline Phosphatase/pharmacokinetics , Alkaline Phosphatase/pharmacology , Animals , Bone and Bones/drug effects , Drug Delivery Systems , Estradiol/chemistry , Estradiol/pharmacokinetics , Estradiol/pharmacology , Humans , Levofloxacin , Ofloxacin/chemistry , Ofloxacin/pharmacokinetics , Ofloxacin/pharmacology
3.
Mol Genet Metab ; 94(2): 178-89, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18359257

ABSTRACT

We have tested an acidic oligopeptide-based targeting system for delivery of enzymes to tissues, especially bone and brain, in a murine mucopolysaccharidosis type VII (MPS VII) model. This strategy is based upon tagging a short peptide consisting of acidic amino acids (AAA) to N terminus of human beta-glucuronidase (GUS). The pharmacokinetics, biodistribution, and the pathological effect on MPS VII mouse after 12 weekly infusions were determined for recombinant human untagged and tagged GUS. The tagged GUS was taken up by MPS VII fibroblasts in a mannose 6-phosphate receptor-dependent manner. Intravenously injected AAA-tagged enzyme had five times more prolonged blood clearance compared with the untagged enzyme. The tagged enzyme was delivered effectively to bone, bone marrow, and brain in MPS VII mice and was effective in reversing the storage pathology. The storage in osteoblasts was cleared similarly with both enzyme types. However, cartilage showed a little response to any of the enzymes. The tagged enzyme reduced storage in cortical neurons, hippocampus, and glia cells. A highly sensitive method of tandem mass spectrometry on serum indicated that the concentration of serum dermatan sulfate and heparan sulfate in mice treated with the tagged enzyme decreased more than the untagged enzyme. These preclinical studies suggest that this AAA-based targeting system may enhance enzyme-replacement therapy.


Subject(s)
Amino Acids, Acidic/therapeutic use , Glucuronidase/metabolism , Mucopolysaccharidosis VII/drug therapy , Amino Acids, Acidic/genetics , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Disease Models, Animal , Gene Targeting , Glucuronidase/administration & dosage , Glucuronidase/genetics , Humans , Lysosomes/enzymology , Mice , Mice, Transgenic , Mucopolysaccharidosis VII/enzymology , Mucopolysaccharidosis VII/genetics , Mucopolysaccharidosis VII/metabolism , Peptides/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use
4.
Hum Mol Genet ; 17(6): 815-24, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18056156

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA) is an autosomal recessive disorder caused by a deficiency of N-acetylgalactosamine-6-sulfate sulfatase (GALNS), leading to accumulation of keratan sulfate (KS) and chrondroitin-6-sulfate. The pharmacokinetics and biodistributions were determined for two recombinant human GALNSs produced in CHO cell lines: native GALNS and sulfatase-modifier-factor 1 (SUMF1) modified GALNS. Preclinical studies of enzyme replacement therapy (ERT) by using two GALNS enzymes were performed on MPS IVA mice. The half-lives in blood circulation of two phosphorylated GALNS enzymes were similar (native, 2.4 min; SUMF1, 3.3 min). After intravenous doses of 250 units/g body weight were administered, each enzyme was primarily recovered in liver and spleen, with detectable activity in other tissues including bone and bone marrow. At 4 h post-injection, enzyme activity was retained in the liver, spleen, bone and bone marrow at levels that were 20-850% of enzyme activity in the wild-type mice. After intravenous doses of 250 units/g of native GALNS, and 250, 600 or 1000 units/g of SUMF1-GALNS were administered weekly for 12 weeks, MPS IVA mice showed marked reduction of storage in visceral organs, sinus lining cells in bone marrow, heart valves, ligaments and connective tissues. A dose-dependent clearance of storage material was observed in brain. The blood KS level assayed by tandem mass spectrometry was reduced nearly to normal level. These preclinical studies demonstrate the clearance of tissue and blood KS by administered GALNS, providing the in vivo rationale for the design of ERT trials in MPS IVA.


Subject(s)
Chondroitinsulfatases/therapeutic use , Disease Models, Animal , Mucopolysaccharidosis IV/drug therapy , Animals , Chondroitinsulfatases/pharmacokinetics , Humans , Mice , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Tissue Distribution
5.
Biol Pharm Bull ; 30(11): 2079-83, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17978479

ABSTRACT

The anti-hyperglycemic action of Stephania tetrandra Radix (Stephania) is potentiated by Astragalus membranaceus BUNGE Radix (Astragali) in streptozotocin (STZ)-diabetic ddY mice (Tsutsumi et al., Biol. Pharm. Bull., 26, 313 (2003)). Fangchinoline (0.3-3 mg/kg), a main constituent of Stephania, decreased the high level of blood glucose and increased the low level of blood insulin in STZ-diabetic mice. Here, we investigated the combined effects of fangchinoline with isoflavone or isoflavonoid components (formononetin, calycosin and ononin) of Astragali on the hyperglycemia and hypoinsulinemia of STZ-diabetic mice. Formononetin, calycosin and ononin (0.03-0.1 mg/kg) alone did not affect the blood glucose or blood insulin level of the diabetic mice. Formononetin and calycosin (0.03-0.1 mg/kg) potentiated the anti-hyperglycemic action of fangchinoline (0.3 mg/kg), but ononin did not. Formononetin (0.1 mg/kg) facilitated the fangchinoline-induced insulin release, and calycosin (0.1 mg/kg) also facilitated it, though without statistical significance. In conclusion, the combined effect of fangchinoline with formononetin and calycosin on hyperglycemia in the diabetic mice accounted well for the therapeutic effect of the combination of Stephania with Astragali in Boi-ogi-to. The anti-hyperglycemic action of formononetin appeared to be due to its potentiating action on insulin release. Our strategy for studying combinations of crude drugs and their components in Kampo medicine has uncovered new potentiating effects of formononetin and calycosin on the anti-hyperglycemic action of fangchinoline in STZ-diabetic mice.


Subject(s)
Alkaloids/therapeutic use , Astragalus propinquus/chemistry , Benzylisoquinolines/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Hyperglycemia/drug therapy , Isoflavones/therapeutic use , Plant Roots/chemistry , Stephania tetrandra/chemistry , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Drug Therapy, Combination , Hyperglycemia/blood , Hyperglycemia/etiology , Insulin/blood , Mice , Molecular Structure
6.
Anticancer Res ; 27(5A): 3187-93, 2007.
Article in English | MEDLINE | ID: mdl-17970060

ABSTRACT

BACKGROUND: The mouse epidermal JB6 cell system is a model for studying tumor promotion. We used the JB6 Cl 41 cell line to examine the mechanism of the anti-tumor-promoting effect of tetrandrine, an alkaloid isolated from Stephania tetrandra S Moore. MATERIALS AND METHODS: The anti-tumor-promoting effect of tetrandrine was evaluated by assay of inhibition of epidermal growth factor (EGF)-induced transformation of JB6 Cl 41 cells in soft agar. The activity of activator protein-1 (AP-1), a transcription factor, was analyzed using the AP-1-dependent reporter assay. Phosphorylation of extracellular-signal regulated kinases (ERKs) and Akt, a pivotal effector of phosphatidylinositol 3-kinase (P13K), was detected by Western blotting. RESULTS: Tetrandrine significantly blocked EGF-induced cell transformation, attenuated EGF-induced AP-1 activation, and inhibited phosphorylation of ERKs, which regulates AP-1 activation. It also tended to suppress EGF-induced Akt phosphorylation. CONCLUSION: Our results indicate that tetrandrine inhibits EGF-induced transformation of JB6 cells by blocking the activation of ERKs, AP-1 and Akt.


Subject(s)
Alkaloids/pharmacology , Benzylisoquinolines/pharmacology , Cell Transformation, Neoplastic/drug effects , Epidermal Growth Factor/antagonists & inhibitors , Animals , Cell Line , Cell Line, Transformed , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Epidermal Growth Factor/pharmacology , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism
7.
Mol Genet Metab ; 91(1): 69-78, 2007 May.
Article in English | MEDLINE | ID: mdl-17336563

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA) is an autosomal recessive disorder caused by a deficiency of N-acetylgalactosamine-6-sulfate sulfatase (GALNS). The aims of this study were to establish Chinese hamster ovary (CHO) cells overexpressing recombinant human GALNS (rhGALNS) and to assess pharmacokinetics and tissue distribution of purified enzymes by using MPS IVA knock-out mouse (Galns(-/-)). The CHO-cell derived rhGALNS was purified from the media by a two-step affinity chromatography procedure. The rhGALNS was administered intravenously to 3-month-old Galns(-/-) mice at a single dose of 250U/g of body weight. The treated mice were examined by assaying the GALNS activity at baseline and up to 240min to assess clearance of the enzyme from blood circulation. The mice were sacrificed 4h after infusion of the enzyme to study the enzyme distribution in tissues. The rhGALNS was purified 1317-fold with 71% yield. The enzyme was taken up by Galns(-/-) chondrocytes (150U/mg/15h). The uptake was inhibited by mannose-6-phosphate. The enzyme activity disappeared from circulation with a half-life of 2.9min. After enzyme infusion, the enzyme was taken up and detected in multiple tissues (40.7% of total infused enzymes in liver). Twenty-four hours after a single infusion of the fluorescence-labeled enzymes into MPS IVA mice, biodistribution pattern showed the amount of tagged enzyme retained in bone, bone marrow, liver, spleen, kidney, and heart. In conclusion, we have shown that the phosphorylated rhGALNS is delivered to multiple tissues, including bone, and that it functions bioactively in Galns(-/-) chondrocytes implying a potential enzyme replacement treatment.


Subject(s)
Chondroitinsulfatases/pharmacokinetics , Recombinant Proteins/pharmacokinetics , Animals , CHO Cells , Chondroitinsulfatases/genetics , Chondroitinsulfatases/isolation & purification , Chondroitinsulfatases/metabolism , Cricetinae , Cricetulus , Disease Models, Animal , Enzyme Stability , Humans , Mice , Mice, Knockout , Mice, Transgenic , Mucopolysaccharidosis IV/drug therapy , Mucopolysaccharidosis IV/enzymology , Recombinant Proteins/isolation & purification , Time Factors , Tissue Distribution
8.
Biol Pharm Bull ; 30(1): 133-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17202673

ABSTRACT

N(epsilon)-(carboxymethyl)lysine (CML) adduct, a major structure of advanced glycation end product, facilitated production of immature microvessels from choroidal explant cultured in fibrin gel. The present study was investigated an action of endogenous CML adduct on neovascularization of cultured choroidal explants of aged Wistar rats with 9 months of age. The number of microvessels budded from explants was counted under optical microscope and used as an index of in vitro neovascularization. Aged choroidal explants increased the neovascularization in an age-dependent manner. Anti-CML antibody decreased age-facilitated neovascularization as well as CML-human serum albumin (HSA)-facilitated neovascularization. Both the aged explant and CML-HSA-treated explant significantly released vascular endothelial growth factor (VEGF), tumor necrosis factor (TNF) alpha and platelet-derived growth factor (PDGF)-B during the culture period. The release of TNF alpha and PDGF-B was earlier than that of VEGF from the aged explants. The antibodies against these factors decreased the CML-facilitated and age-facilitated neovascularization in the choroidal explants. The inhibitory capacity of anti-TNF alpha antibody was greater than those of anti-VEGF and anti-PDGF-B antibodies. In conclusion, endogenous CML adduct overproduced the neovascularization of the aged choroidal explant. The CML adduct releases TNF alpha which might induce the production and release of VEGF for the abnormal choroidal neovascularization in the patients of age-related macular degeneration.


Subject(s)
Aging/metabolism , Capillaries/metabolism , Choroid/blood supply , Choroidal Neovascularization/metabolism , Glycation End Products, Advanced/metabolism , Lysine/analogs & derivatives , Age Factors , Animals , Antibodies/pharmacology , Capillaries/drug effects , Glycation End Products, Advanced/immunology , Lysine/immunology , Lysine/metabolism , Macular Degeneration/metabolism , Macular Degeneration/physiopathology , Male , Organ Culture Techniques , Platelet-Derived Growth Factor/immunology , Platelet-Derived Growth Factor/metabolism , Rats , Rats, Wistar , Time Factors , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
9.
Mol Carcinog ; 46(6): 436-45, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17219438

ABSTRACT

We found that quercetin, myricetin, quercetagetin, fisetin, (-)-epigallocatechin gallate (EGCG), and theaflavins, among 24 flavonoids examined, markedly inhibited epidermal growth factor (EGF)-induced cell transformation of mouse epidermal JB6 Cl 41 cells. The six flavonoids suppressed the EGF-induced activation of activator protein 1 (AP-1). In addition, myricetin, quercetagetin, EGCG, and theaflavins directly inhibited EGF-induced phosphatidylinositol 3-kinase (PI3K) activation. The important structural features of flavonoids for cell transformation-inhibitory activity are 3'- and 4'-OH on the B-ring, 3-OH on the C-ring, C2=C3 double bond in the C-ring, and the phenylchromone (C6-C5-C6) skeleton in the flavonols, and the galloyl group in EGCG and theaflavins. Our results provide new insight into possible mechanisms of the anti-carcinogenic effects of flavonoids, and could help to provide a basis for the design of novel cancer chemopreventive agents.


Subject(s)
Cell Transformation, Neoplastic , Epidermal Growth Factor/metabolism , Flavonoids/therapeutic use , Animals , Apoptosis , Cell Adhesion , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Mice , Models, Chemical , Protein Binding , Signal Transduction , Structure-Activity Relationship , Transcription Factor AP-1/biosynthesis
10.
Proc Natl Acad Sci U S A ; 103(41): 15172-7, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-17015822

ABSTRACT

Enzyme replacement therapy (ERT) is available for several lysosomal storage diseases. Except for Gaucher disease, for which an enzyme with exposed mannosyl residues targets mannose receptors (MR) on macrophages, ERT targets primarily the mannose 6-phosphate receptor (MPR). Most recombinant lysosomal enzymes contain oligosaccharides with both terminal mannosyl and mannose 6-phosphate residues. Effective MPR-mediated delivery may be compromised by rapid clearance of infused enzyme by the MR on fixed tissue macrophages, especially Kupffer cells. To evaluate the impact of this obstacle to ERT, we introduced the MR-null mutation onto the mucopolysaccharidosis type VII (MPS VII) background and produced doubly deficient MR-/- MPS VII mice. The availability of both MR+/+ and MR-/- mice allowed us to study the effects of eliminating the MR on MR- and MPR-mediated plasma clearance and tissue distribution of infused phosphorylated (P) and nonphosphorylated (NP) forms of human beta-glucuronidase (GUS). In MR+/+ MPS VII mice, the MR clearance system predominated at doses up to 6.4 mg/kg P-GUS. Genetically eliminating the MR slowed plasma clearance of both P- and NP-GUS and enhanced the effectiveness of P-GUS in clearing storage in kidney, bone, and retina. Saturating the MR clearance system by high doses of enzyme also improved targeting to MPR-containing tissues such as muscle, kidney, heart, and hepatocytes. Although ablating the MR clearance system genetically is not practical clinically, blocking the MR-mediated clearance system with high doses of enzyme is feasible. This approach delivers a larger fraction of enzyme to MPR-expressing tissues, thus enhancing the effectiveness of MPR-targeted ERT.


Subject(s)
Glucuronidase/therapeutic use , Lectins, C-Type/deficiency , Lectins, C-Type/physiology , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/physiology , Mannosephosphates/metabolism , Mucopolysaccharidosis VII/drug therapy , Mucopolysaccharidosis VII/genetics , Receptor, IGF Type 2/physiology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/physiology , Animals , Glucuronidase/deficiency , Glucuronidase/genetics , Glucuronidase/metabolism , Lectins, C-Type/genetics , Mannose Receptor , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucopolysaccharidosis VII/enzymology , Phosphorylation , Receptors, Cell Surface/genetics
11.
J Bone Miner Metab ; 24(4): 260-5, 2006.
Article in English | MEDLINE | ID: mdl-16816919

ABSTRACT

To assess the combination effect of calcitonin and the phosphodiesterase 4 inhibitor rolipram on osteoclastogenesis, adherent cell-depleted bone marrow cells from mouse tibia and femur (ACD-BMCs), which were cultured in the presence of 25 ng/ml colony-stimulating factor 1 (CSF-1) and 100 ng/ml soluble receptor activator of NF-kappaB ligand (sRANKL), were utilized. Calcitonin inhibited formation of tartrate-resistant acid phosphatase-positive multinucleated cells, as mature osteoclasts, by 70% even at 20 pM, whereas rolipram (10 microM) scarcely affected osteoclast formation; in contrast, the combination of both agents led to significant inhibition of multinucleation and pit formation ability of osteoclasts. The combined administration of calcitonin and rolipram attenuated calcitonin receptor mRNA expression in comparison to treatment with either agent alone, whereas expression of RANK and CSF-1 receptor mRNAs was unchanged. Alone, these agents scarcely elevated intracellular cyclic AMP (cAMP) concentration; however, combination treatment with both agents significantly increased cAMP concentration in osteoclast progenitors and osteoclasts. The combination effect was abolished by H-89, an inhibitor of protein kinase A. It appears that rolipram inhibited hydrolysis of cAMP formed by calcitonin in cells and potentiated the inhibitory effect of calcitonin on osteoclastogenesis. The escape phenomenon following calcitonin treatment may also be prevented by concomitant treatment with the phosphodiesterase 4 inhibitor.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Calcitonin/pharmacology , Osteoclasts/cytology , Osteoclasts/drug effects , Rolipram/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Calcitonin/agonists , Cyclic Nucleotide Phosphodiesterases, Type 4 , Drug Synergism , Enzyme Inhibitors/pharmacology , Mice , RNA, Messenger
12.
Mol Genet Metab ; 89(1-2): 139-49, 2006.
Article in English | MEDLINE | ID: mdl-16837223

ABSTRACT

Design of efficient treatment strategies for diseases requires clarification of the nature of each mutation causing the disease. In this study, we have investigated three factors to correctly predict the correlation between genotype and phenotype on N-acetylgalactosamine-6-sulfate sulfatase (GALNS) gene responsible for one of lysosomal storage diseases, known as mucopolysaccharidosis IVA (MPS IVA); (i) evolutionary conservation of amino acid residues among family proteins, (ii) conservativeness of amino acid changes in GALNS, and (iii) structural conservation of amino acid residue. The results showed that (i) the likelihood of a missense variant causing MPS IVA was directly correlated with the level of evolutionary conservation and inversely correlated with conservativeness but not correlated with the structural conservation, (ii) the disease-causative mutations were 9 times more likely to be located on the 'highly conserved' residues than the polymorphisms, (iii) the likelihood of 'non-conservative' amino acid changes in missense mutations was 6.8 times higher than those in the polymorphisms, (iv) the degree of evolutionary conservation was nearly as predictive in phenotype as that of conservativeness of amino acid changes, and (v) the combination of the two factors, evolutionary conservation and conservativeness, provides a better association between missense variants and clinical severity with higher sensitivity (83.5-88.9%) and specificity (71.4-88.3%), than that obtained by either factor alone. These findings suggest that the combination of evolutionary conservation and conservativeness is a useful tool to predict the effect of each mutation on the clinical phenotype and can be applied to the analysis of phenotype/genotype relation in other genetic diseases.


Subject(s)
Chondroitinsulfatases/genetics , Mucopolysaccharidosis IV/diagnosis , Polymorphism, Genetic , Severity of Illness Index , Amino Acid Sequence , Amino Acid Substitution , Conserved Sequence , DNA Mutational Analysis , Evolution, Molecular , Genetic Markers , Humans , Molecular Sequence Data , Mutation, Missense
13.
Biol Pharm Bull ; 29(6): 1229-33, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16755022

ABSTRACT

We examined the usefulness of intranasal (i.n.) administration of a novel osteotropic prodrug of estradiol, estradiol-17beta-succinate-(L-aspartate)6 (E2.17D6), for selective drug delivery to bone. E2.17D6 alone or with 5% 2,6-di-O-methyl-beta-cyclodextrin (DMbetaCD), 5% beta-cyclodextrin (betaCD), or 10% hydroxypropyl cellulose (HPC) as an absorption enhancer was administered to ovariectomized (OVX) mice via the i.n. route. The oral and nasal bioavailability after p.o. or i.n. administration of E2.17D6 (3.7 micromol/kg) in mice amounted to 9.9 and 23.0% of the dose, respectively. The values of nasal bioavailability of E2.17D6 administered with DMbetaCD, betaCD, and HPC were 74.9, 55.8, and 49.1%, respectively. The plasma concentration of E2.17D6 after i.n. administration of E2.17D6-DMbetaCD decreased rapidly to the endogenous level by 6 h, but the concentration in the bone was about 200 times higher than that in plasma, and decreased slowly over a period of about a week. When E2 (total dose 4.4 micromol/kg, i.n., every 3rd day) was administered to OVX mice for 35 d, bone mineral density (BMD), liver weight, and uterus weight increased, whereas E2.17D6-DMbetaCD (total dose 0.44 to 8.8 micromol/kg, i.n., every 7th day) increased only BMD in a dose-dependent manner. In conclusion, intranasally administered E2.17D6-DMbetaCD has a potent antiosteoporotic effect without side effects, and has potential to provide an improved quality of life for patients with osteoporosis.


Subject(s)
Aspartic Acid/analogs & derivatives , Bone and Bones/metabolism , Estradiol/analogs & derivatives , Osteoporosis/drug therapy , Prodrugs/pharmacokinetics , Administration, Intranasal , Animals , Aspartic Acid/administration & dosage , Aspartic Acid/blood , Aspartic Acid/pharmacokinetics , Aspartic Acid/therapeutic use , Dose-Response Relationship, Drug , Drug Carriers , Estradiol/administration & dosage , Estradiol/blood , Estradiol/pharmacokinetics , Estradiol/therapeutic use , Female , Gas Chromatography-Mass Spectrometry , Mice , Mice, Inbred Strains , Molecular Structure , Ovariectomy , Prodrugs/administration & dosage , Prodrugs/therapeutic use , Time Factors , Tissue Distribution , beta-Cyclodextrins
14.
Eur J Hum Genet ; 14(7): 838-45, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16617305

ABSTRACT

Hunter syndrome, an X-linked disorder, results from deficiency of iduronate-2-sulfatase (IDS). Around 40% of independent point mutations at IDS were found at CpG sites as transitional events. The 15 CpG sites in the coding sequences of exons 1 and 2, which are normally hypomethylated, account for very few of transitional mutations. By contrast, the CpG sites in the coding sequences of exon 3, though also normally hypomethylated, account for much higher fraction of transitional mutations. To better understand relationship between methylation status and CpG transitional mutations in this region, the methylation patterns of 11 Hunter patients with transitional mutations at CpG sites were investigated using bisulfite genomic sequencing. The patient cohort mutation spectrum is composed of one mutation in exon 1 (one patient) and three different mutations in exon 3 (10 patients). We confirmed that in normal males, cytosines at the CpG sites from the promoter region to a portion of intron 3 were hypomethylated. However, specific CpG sites in this area were more highly methylated in patients. The patients with p.R8X (exon 1), p.P86L (exon 3), and p.R88H (exon 3) mutations had a hypermethylated condition in exon 2 to intron 3 but retained hypomethylation in exon 1. The same trend was found in four patients with p.A85T (exon 3), although the degree of hypermethylation was less. These findings suggest methylation patterns in the beginning of IDS genomic region are polymorphic in humans and that hypermethylation in this region in some individuals predisposes them to CpG mutations resulting in Hunter syndrome.


Subject(s)
CpG Islands/genetics , DNA Methylation , Glycoproteins/genetics , Mucopolysaccharidosis II/genetics , Adolescent , Adult , Amino Acid Sequence , Case-Control Studies , Child , Child, Preschool , Exons/genetics , Humans , Infant , Male , Molecular Sequence Data , Point Mutation/genetics , Promoter Regions, Genetic , Sequence Analysis, DNA
15.
Mol Genet Metab ; 88(3): 244-55, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16616566

ABSTRACT

Hypophosphatasia is caused by deficiency of activity of the tissue-nonspecific alkaline phosphatase (TNSALP), resulting in a defect of bone mineralization. Enzyme replacement therapy (ERT) with partially purified plasma enzyme was attempted but with little clinical improvement. Attaining clinical effectiveness with ERT for hypophosphatasia may require delivering functional TNSALP enzyme to bone. We tagged the C-terminal-anchorless TNSALP enzyme with an acidic oligopeptide (a six or eight residue stretch of L-Asp), and compared the biochemical properties of the purified tagged and untagged enzymes derived from Chinese hamster ovary cell lines. The specific activities of the purified enzymes tagged with the acidic oligopeptide were the same as the untagged enzyme. In vitro affinity experiments showed the tagged enzymes had 30-fold higher affinity for hydroxyapatite than the untagged enzyme. Lectin affinity chromatography for carbohydrate structure showed little difference among the three enzymes. Biodistribution pattern from single infusion of the fluorescence-labeled enzymes into mice showed delayed clearance from the plasma up to 18 h post infusion and the amount of tagged enzyme retained in bone was 4-fold greater than that of the untagged enzyme. In vitro mineralization assays with the bone marrow from a hypophosphatasia patient using each of the three enzymes in the presence of high concentrations of pyrophosphate provided evidence of bone mineralization. These results show the anchorless enzymes tagged with an acidic oligopeptide are delivered efficiently to bone and function bioactively in bone mineralization, at least in vitro. They suggest potential advantages for use of these tagged enzymes in ERT for hypophosphatasia, which should be explored.


Subject(s)
Alkaline Phosphatase/pharmacokinetics , Bone Marrow Cells/drug effects , Calcification, Physiologic/drug effects , Oligopeptides/chemistry , Alkaline Phosphatase/chemistry , Animals , Asparagine/chemistry , Bone Marrow Cells/physiology , Calcification, Physiologic/physiology , Cells, Cultured , Cricetinae , Cricetulus , Durapatite/chemistry , Humans , Hypophosphatasia/pathology , Infant , Liver/enzymology , Mice , Tissue Distribution
16.
Hum Mutat ; 26(6): 500-12, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16287098

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA; Morquio A disease) is an autosomal-recessive disorder caused by a deficiency of lysosomal N-acetylgalactosamine-6-sulfate sulfatase (GALNS; E.C.3.1.6.4). GALNS is required to degrade glycosaminoglycans, keratan sulfate (KS), and chondroitin-6-sulfate. Accumulation of undegraded substrates in lysosomes of the affected tissues leads to a systemic bone dysplasia. We summarize information on 148 unique mutations determined to date in the GALNS gene, including 26 novel mutations (19 missense, four small deletions, one splice-site, and two insertions). This heterogeneity in GALNS gene mutations accounts for an extensive clinical variability within MPS IVA. Seven polymorphisms that cause an amino acid change, and nine silent variants in the coding region are also described. Of the analyzed mutant alleles, missense mutations accounted for 78.4%; small deletions, 9.2%; nonsense mutation, 5.0%; large deletion, 2.4%; and insertions, 1.6%. Transitional mutations at CpG dinucleotides accounted for 26.4% of all the described mutations. The importance of the relationship between methylation status and distribution of transitional mutations at CpG sites at the GALNS gene locus was elucidated. The three most frequent mutations (over 5% of all mutations) were represented by missense mutations (p.R386C, p.G301C, and p.I113F). A genotype/phenotype correlation was defined in some mutations. Missense mutations associated with a certain phenotype were studied for their effects on enzyme activity and stability, the levels of blood and urine KS, the location of mutations with regard to the tertiary structure, and the loci of the altered amino acid residues among sulfatase proteins.


Subject(s)
Chondroitinsulfatases/genetics , Mucopolysaccharidosis IV/genetics , Mutation , Polymorphism, Genetic , Animals , Chondroitinsulfatases/chemistry , CpG Islands , DNA Methylation , Genotype , Humans , Keratan Sulfate/metabolism , Mice , Mucopolysaccharidosis IV/diagnosis , Mucopolysaccharidosis IV/epidemiology , Protein Structure, Tertiary
17.
Hum Mol Genet ; 14(22): 3321-35, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16219627

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA) is an autosomal recessive disease caused by N-acetylgalactosamine-6-sulfate sulfatase (GALNS) deficiency. In recent studies of enzyme replacement therapy for animal models with lysosomal storage diseases, cellular and humoral immune responses to the injected enzymes have been recognized as major impediments to effective treatment. To study the long-term effectiveness and side effects of therapies in the absence of immune responses, we have developed an MPS IVA mouse model, which has many similarities to human MPS IVA and is tolerant to human GALNS protein. We used a construct containing both a transgene (cDNA) expressing inactive human GALNS in intron 1 and an active site mutation (C76S) in adjacent exon 2 and thereby introduced both the inactive cDNA and the C76S mutation into the murine Galns by targeted mutagenesis. Affected homozygous mice have no detectable GALNS enzyme activity and accumulate glycosaminoglycans in multiple tissues including visceral organs, brain, cornea, bone, ligament and bone marrow. At 3 months, lysosomal storage is marked within hepatocytes, reticuloendothelial Kupffer cells, and cells of the sinusoidal lining of the spleen, neurons and meningeal cells. The bone storage is also obvious, with lysosomal distention in osteoblasts and osteocytes lining the cortical bone, in chondrocytes and in the sinus lining cells in bone marrow. Ubiquitous expression of the inactive human GALNS was also confirmed by western blot using the anti-GALNS monoclonal antibodies newly produced, which resulted in tolerance to immune challenge with human enzyme. The newly generated MPS IVA mouse model should provide a good model to evaluate long-term administration of enzyme replacement.


Subject(s)
Chondroitinsulfatases/genetics , Mucopolysaccharidosis IV/enzymology , Mucopolysaccharidosis IV/genetics , Animals , Chondroitinsulfatases/administration & dosage , Chondroitinsulfatases/biosynthesis , Chondroitinsulfatases/deficiency , Chondroitinsulfatases/immunology , Disease Models, Animal , Female , Heart Valves/pathology , Humans , Immune Tolerance/genetics , Liver/pathology , Male , Meninges/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucopolysaccharidosis IV/pathology , Organ Specificity/genetics , Organ Specificity/immunology , RNA, Messenger
18.
Biochem Pharmacol ; 68(10): 1929-37, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15476664

ABSTRACT

Caffeine inhibits insulin-induced glucose uptake in rat adipocytes and also decreases insulin sensitivity, including whole-body glucose disposal and glucose uptake in skeletal muscle, during a euglycemic-hyperinsulinemic clamp in human. However, the mechanism by which caffeine decreases the insulin sensitivity is not still clear. We found that pre-treatment with caffeine inhibited the insulin-induced 2-deoxy-D-[1-(3)H]glucose uptake in a concentration-dependent manner in mouse preadipose MC-3T3-G2/PA6 cells differentiated into mature adipose cells. Caffeine also suppressed insulin-induced GLUT4 translocation in the differentiated cells. Although caffeine did not alter insulin-induced activation of PI3K and protein kinase C-zeta (PKCzeta), an isoform of atypical PKC, which is reported to have an important role in insulin-induced GLUT4 translocation, we found that insulin-induced phosphorylation and activation of Akt were blocked by pre-treatment with caffeine. Inhibition of insulin-induced 2-deoxy-D-[1-(3)H]glucose uptake by caffeine was also observed in primary cultured brown adipocytes in a concentration-dependent manner. These results may, in part, explain the ability of caffeine to decrease insulin sensitivity.


Subject(s)
Adipocytes/drug effects , Caffeine/pharmacology , Glucose/metabolism , Insulin/pharmacology , Adipocytes/metabolism , Animals , Biological Transport , Cells, Cultured , Cyclic AMP/metabolism , Deoxyglucose/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Purinergic P1/metabolism , Tritium
19.
J Hum Genet ; 49(9): 490-494, 2004.
Article in English | MEDLINE | ID: mdl-15309681

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA) is a lysosomal storage disorder caused by the deficiency of N-acetylgalactosamine-6-sulfate sulfatase (GALNS). Mutation screening of the GALNS was performed by genomic PCR and direct sequence analyses in 20 MPS IVA patients from Latin America. In this study, 12 different gene mutations including nine unreported ones were identified in 16 severe and four attenuated patients and accounted for 90.0% of the unrelated mutant alleles. The gene alterations were missense mutations except one insertion. Six recurrent mutations, p.A75G, p.G116S, p.G139S, p.N164T, p.R380S, and p.R386C, accounted for 5.0, 10.0, 5.0, 7.5, 5.0, and 32.5% of the unrelated mutant alleles, respectively. The p.R386C mutation was identified in all Latin American populations studied. Eleven mutations correlated with a severe form, while one mutation, p.R380S, was associated with an attenuated form. MPS IVA patients had an elevation of urine and plasma keratan sulfate (KS) concentrations compared with those of the age-matched control. KS concentrations in severe patients were higher than those in attenuated patients. These data provide evidence for extensive allelic heterogeneity and presence of a common mutation in Latin American patients. Accumulation of mutations with clinical description and KS concentration will lead us to predict clinical severity of the patient more precisely.


Subject(s)
Chondroitinsulfatases/genetics , Mutation, Missense/genetics , Phenotype , Adolescent , Adult , Child , Child, Preschool , Chondroitinsulfatases/metabolism , DNA Primers , Female , Gene Frequency , Genetic Testing , Genotype , Humans , Keratan Sulfate/blood , Keratan Sulfate/urine , Male , Sequence Analysis, DNA , South America
20.
Hum Mutat ; 24(2): 187-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15241807

ABSTRACT

Mucopolysaccharidosis IVA (MPS IVA) is a lysosomal storage disorder caused by the deficiency of N-acetylgalactosamine-6-sulfate sulfatase (GALNS). Mutation screening of the GALNS gene was performed by RT-PCR with one amplicon and direct sequence analyses using cDNA samples from 15 Italian MPS IVA patients. Each mutation was confirmed at the genomic level. In this study, 13 different gene mutations with four common mutations (over 10% of mutant alleles) were identified in 12 severe and three milder (attenuated) MPS IVA patients. The gene alterations in 12 out of 13 were found to be point mutations and only one mutation was deletion. Ten of 13 mutations were novel. The c.1070C>T (p.Pro357Leu) mutation coexisted with c.1156C>T (p.Arg386Cys) mutation on the same allele. Together they accounted for 100% of the 30 disease alleles of the patients investigated. Four common mutations accounted for 70% of mutant alleles investigated. Urine keratan sulfate (KS) concentrations were elevated in all patients investigated. These data provide further evidence for extensive allelic heterogeneity and importance of relation among genotype, phenotype, and urine KS excretion as a biomarker in MPS IVA.


Subject(s)
Chondroitinsulfatases/genetics , Mucopolysaccharidosis IV/enzymology , Mucopolysaccharidosis IV/genetics , Mutation/genetics , Adolescent , Adult , Biomarkers/analysis , Blotting, Western/methods , Child , Chondroitinsulfatases/immunology , Female , Genotype , Humans , Italy , Male , Molecular Diagnostic Techniques/methods , Mucopolysaccharidosis IV/diagnosis , Phenotype , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Analysis, DNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...