Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
1.
J Med Chem ; 64(12): 8599-8606, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34096701

ABSTRACT

Glioblastoma multiforme (GBM) is a highly invasive and aggressive malignant glioma. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma, so more effective strategies of antitumor treatments are in urgent demand. Here, we found that lysosomal sulfatase expression was significantly correlated with poor prognosis of GBM. Hence, a new probe, MNG, was developed with a new protocol utilizing glucose groups to detect lysosomal sulfatase. It also exhibits potential for monitoring GBM cells, depending on the hyperactive lysosomal sulfatase expression of tumor cells. Meantime, we identified that sulbactam as the first reported lysosomal sulfatase inhibitor inhibits the tumor growth of GBM. Collectively, our work highlights that lysosomal sulfatase was detected using a new protocol and its potential as a therapeutic target in GBM and reveals a distinct mechanism that sulbactam inhibits cell proliferation related to lysosomal sulfatase, indicating that sulbactam could be a promising therapeutic agent against GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Sulbactam/pharmacology , Sulfatases/antagonists & inhibitors , Cell Line, Tumor , Fluorescent Dyes/chemistry , Glioblastoma/diagnosis , Glioblastoma/enzymology , Glucosides/chemistry , Humans , Lysosomes/enzymology , Naphthalimides/chemistry , Prognosis , Sulfatases/analysis , Sulfatases/chemistry
2.
Hepatology ; 74(3): 1411-1428, 2021 09.
Article in English | MEDLINE | ID: mdl-33735525

ABSTRACT

BACKGROUND AND AIMS: Existing therapeutic approaches to treat cholangiocarcinoma (CCA) have limited effectiveness, prompting further study to develop therapies for CCA. We report a mechanistic role for the heparan sulfate editing enzyme sulfatase 2 (SULF2) in CCA pathogenesis. APPROACH AND RESULTS: In silico analysis revealed elevated SULF2 expression in human CCA samples, occurring partly through gain of SULF2 copy number. We examined the effects of knockdown or overexpression of SULF2 on tumor growth, chemoresistance, and signaling pathway activity in human CCA cell lines in vitro. Up-regulation of SULF2 in CCA leads to increased platelet-derived growth factor receptor beta (PDGFRß)-Yes-associated protein (YAP) signaling activity, promoting tumor growth and chemotherapy resistance. To explore the utility of targeting SULF2 in the tumor microenvironment for CCA treatment, we tested an anti-SULF2 mouse monoclonal antibody, 5D5, in a mouse CCA xenograft model. Targeting SULF2 by monoclonal antibody 5D5 inhibited PDGFRß-YAP signaling and tumor growth in the mouse xenograft model. CONCLUSIONS: These results suggest that SULF2 monoclonal antibody 5D5 or related agents may be potentially promising therapeutic agents in CCA.


Subject(s)
Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Sulfatases/genetics , YAP-Signaling Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Bile Duct Neoplasms/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cholangiocarcinoma/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Knockdown Techniques , Humans , Mice , Neoplasm Transplantation , Receptor, Platelet-Derived Growth Factor beta/drug effects , Sulfatases/antagonists & inhibitors , Sulfatases/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays , YAP-Signaling Proteins/drug effects
3.
J Am Chem Soc ; 142(11): 5282-5292, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32083852

ABSTRACT

Human endo-O-sulfatases (Sulf-1 and Sulf-2) are extracellular heparan sulfate proteoglycan (HSPG)-specific 6-O-endosulfatases, which regulate a multitude of cell-signaling events through heparan sulfate (HS)-protein interactions and are associated with the onset of osteoarthritis. These endo-O-sulfatases are transported onto the cell surface to liberate the 6-sulfate groups from the internal d-glucosamine residues in the highly sulfated subdomains of HSPGs. In this study, a variety of HS oligosaccharides with different chain lengths and N- and O-sulfation patterns via chemical synthesis were systematically studied about the substrate specificity of human Sulf-1 employing the fluorogenic substrate 4-methylumbelliferyl sulfate (4-MUS) in a competition assay. The trisaccharide sulfate IdoA2S-GlcNS6S-IdoA2S was found to be the minimal-size substrate for Sulf-1, and substitution of the sulfate group at the 6-O position of the d-glucosamine unit with the sulfonamide motif effectively inhibited the Sulf-1 activity with IC50 = 0.53 µM, Ki = 0.36 µM, and KD = 12 nM.


Subject(s)
Enzyme Inhibitors/chemistry , Sulfatases/antagonists & inhibitors , Sulfonamides/chemistry , Sulfotransferases/antagonists & inhibitors , Trisaccharides/chemistry , Enzyme Assays , Enzyme Inhibitors/chemical synthesis , Heparitin Sulfate/chemistry , Humans , Kinetics , Substrate Specificity , Sulfatases/chemistry , Sulfonamides/chemical synthesis , Sulfotransferases/chemistry , Trisaccharides/chemical synthesis
4.
Eur J Med Chem ; 177: 116-143, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31129450

ABSTRACT

Breast cancer, a most common malignancy in women, was known to be associated with steroid hormone estrogen. The discovery of estrogen receptor (ER) gave us not only a powerful predictive and prognostic marker, but also an efficient target for the treatment of hormone-dependent breast cancer with various estrogen ligands. ER consists of two subtypes i.e. ERα and ERß, that are mostly G-protein-coupled receptors and activated by estrogen, specially 17ß-estradiol. The activation is followed by translocation into the nucleus and binding with DNA to modulate activities of different genes. ERs can manage synthesis of RNA through genomic actions without directly binding to DNA. Receptors are tethered by protein-protein interactions to a transcription factor complex to communicate with DNA. Estrogens also exhibit nongenomic actions, a characteristic feature of steroid hormones, which are so rapid to be considered by the activation of RNA and translation. These are habitually related to stimulation of different protein kinase cascades. Majority of post-menopausal breast cancer is estrogen dependent, mostly potent biological estrogen (E2) for continuous growth and proliferation. Estrogen helps in regulating the differentiation and proliferation of normal breast epithelial cells. In this review we have investigated the important role of ER in development and progression of breast cancer, which is complicated by receptor's interaction with co-regulatory proteins, cross-talk with other signal transduction pathways and development of treatment strategies viz. selective estrogen receptor modulators (SERMs), selective estrogen receptor down regulators (SERDs), aromatase and sulphatase inhibitors.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Estrogens/therapeutic use , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Signal Transduction/drug effects , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Cell Line, Tumor , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Estrogens/chemistry , Estrogens/pharmacology , Female , Humans , Ligands , Men , Molecular Structure , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/physiology , Sulfatases/antagonists & inhibitors
5.
Exp Dermatol ; 28(1): 28-34, 2019 01.
Article in English | MEDLINE | ID: mdl-30315662

ABSTRACT

Although there are many treatment options for skin cancer, the chemotherapeutic agents for skin cancer are linked with many adverse effects as well as the development of multidrug resistance. Sulforaphane is an isothiocyanate, which is found in cruciferous vegetables. Consumption of sulforaphane-rich diet has been linked to inhibition of UV-exposed skin carcinogenesis. Therefore, the goal of this study was to determine the ability of sulforaphane to reduce skin cancer in mice through inhibition of sulfatase-2 enzyme. Epicutaneous application of 7,12-dimethylbenz (a) anthracene was performed on the shaved dorsal skin of mice followed by croton oil. Sulforaphane (9 µmol/mouse/day) was administered to mice orally. Skin was removed from the dorsal area for assessment of sulfatase-2, glypican-3, heparan sulphate proteoglycans (HSPGs), nuclear factor (NF)κB, nuclear factor E2-related factor 2 (Nrf2), tumor necrosis factor (TNF)-α, IL-1ß and caspase-3. In addition, skin sections were stained with haematoxylin/eosin, Mallory and cytokeratin immunostaining. We found that, sulforaphane blocked sulfatase-2 activity, leading to significant elevation in HSPGs as well as significant reduction in glypican-3. In addition, sulforaphane significantly activated Nrf2 and reduced both the gene and protein expression of NFκB, TNF-α, IL-1ß and caspase-3. In parallel, stained sections obtained from skin cancer mice treated with sulforaphane showed significant reduction in hyperkeratosis, acanthosis and epithelial dysplasia. The collective results indicate that sulforaphane suppresses skin cancer via blocking sulfatase-2 with subsequent elevation in HSPGs and reduction in glypican-3. Moreover, sulforaphane attenuated skin cancer-induced activation of inflammatory and apoptotic pathways.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Isothiocyanates/therapeutic use , Skin Neoplasms/drug therapy , Sulfatases/antagonists & inhibitors , Animals , Anthracenes , Antioxidants/metabolism , Apoptosis , Carcinogens , Caspase 3/metabolism , Disease Models, Animal , Glypicans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Interleukin-1beta/metabolism , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B p50 Subunit/metabolism , Sulfatases/metabolism , Sulfoxides , Tumor Necrosis Factor-alpha/metabolism
6.
FEMS Microbiol Lett ; 364(16)2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28859275

ABSTRACT

Triclosan (TCS) is an antimicrobial compound found in personal care products, and consequently in greywater. After its release to the environment, it continues its antimicrobial action on indigenous microbial communities. Little is known about the environmental impacts of high levels of TCS, which may occur due to accumulation following long-term greywater application to soil. Soil microcosms were established using a silty clay loam and augmented with a range of TCS concentrations ranging from 500 to 7500 mg kg-1. Samples were analysed for substrate-induced respiration, microbial biomass and sulphatase activity. The soil augmented with the lowest concentration of TCS (500 mg kg-1) significantly decreased microbial biomass, with a calculated EC20 of 195 mg kg-1. Substrate-induced respiration indicated that the soil microbial community was impacted for all TCS concentrations; however, the community showed potential to recover over time. Sulphatase activity was less sensitive to TCS and was significantly impacted at high concentrations of TCS (>2500 mg kg-1). It is likely that TCS has selective toxicity for more susceptible microbes when introduced into the soil environment. At high levels, TCS could overwhelm TCS-degrading soil microbes.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Microbiota/drug effects , Soil Microbiology , Soil Pollutants/pharmacology , Triclosan/pharmacology , Wastewater/chemistry , Water Microbiology , Water Pollutants/pharmacology , Anti-Infective Agents, Local/analysis , Microbiota/physiology , Soil Pollutants/isolation & purification , Sulfatases/analysis , Sulfatases/antagonists & inhibitors , Time Factors , Triclosan/analysis , Wastewater/analysis , Water Pollutants/isolation & purification
7.
Bioorg Chem ; 71: 257-274, 2017 04.
Article in English | MEDLINE | ID: mdl-28274582

ABSTRACT

Estrogens display intriguing tissue selective action that is of great biomedical importance in the development of optimal therapeutics for the prevention and treatment of breast cancer. There are also strong evidences to show that both endogenous and exogenous estrogens are involved in the pathogenesis of breast cancer. Tamoxifen has been the only drug of choice for more than 30years to treat patients with estrogen related (ER) positive breast tumors. There is a need therefore, for identifying newer, potential and novel candidates for breast cancer. Keeping this in view, the present review focuses on selective estrogen receptor modulators and estrogen antagonists such as sulfatase and aromatase inhibitors involved in breast cancer therapy. A succinct and critical overview of the structure of estrogen receptors, their signaling and involvement in breast carcinogenesis are herein described.


Subject(s)
Breast Neoplasms/drug therapy , Breast/drug effects , Estrogen Antagonists/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Animals , Aromatase/metabolism , Breast/enzymology , Breast/metabolism , Breast/pathology , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Coumarins/chemistry , Coumarins/pharmacology , Coumarins/therapeutic use , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Female , Humans , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacology , Sulfatases/antagonists & inhibitors , Sulfatases/metabolism
8.
Eur J Med Chem ; 114: 170-90, 2016 May 23.
Article in English | MEDLINE | ID: mdl-26974384

ABSTRACT

Steroid sulfatase (STS) plays a momentous role in the conversion of sulfated steroids, which are biologically inactive, into biologically active un-sulfated steroid hormones, which support the development and growth of a number of hormone-dependent cancers, including breast cancer. Therefore, inhibitors of STS are supposed to be potential drugs for the treatment of breast and other steroid-dependent cancers. The present review concentrates on broad chemical classification of steroid sulfatase inhibitors. The inhibitors reviewed are classified into four main categories: Steroid sulfamate based inhibitors; Steroid non-sulfamate based inhibitors; Non-steroidal sulfamate based inhibitors; Non-steroidal non-sulfamate based inhibitors. A succinct overview of current treatment of cancer, estradiol precursors, STS enzyme and its role in breast cancer is herein described.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/enzymology , Sulfatases/antagonists & inhibitors , Sulfonic Acids/pharmacology , Breast Neoplasms/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Estrogens/metabolism , Female , Humans , Neoplasm Recurrence, Local/metabolism , Sulfatases/metabolism , Sulfonic Acids/chemical synthesis , Sulfonic Acids/chemistry
9.
Org Biomol Chem ; 13(18): 5279-84, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25858034

ABSTRACT

Regioselective sulfamoylation of primary hydroxyl groups enabled a 5-step synthesis (overall yield 17%) of the first reported small molecule inhibitor of sulfatase-1 and 2, ((2S,3R,4R,5S,6R)-4,5-dihydroxy-2-methoxy-6-((sulfamoyloxy)methyl)tetrahydro-2H-pyran-3-yl)sulfamic acid, which obviated the use of hydroxyl protecting groups and is a marked improvement on the reported 9-step synthesis (overall yield 9%) employing hazardous trifluoromethylsulfonyl azide. The sulfamoylation methodology was used to prepare a range of derivatives of 1, and inhibition data was generated for Sulf-2, ARSA and ARSB.


Subject(s)
Cold Temperature , Enzyme Inhibitors/chemical synthesis , Sulfatases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Stereoisomerism
10.
Angiogenesis ; 17(1): 77-91, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23959107

ABSTRACT

Arterial and venous specification is critical for establishing and maintaining a functioning vascular system, and defects in key arteriovenous signaling pathways including VEGF (vascular endothelial growth factor) lead to congenital arteriopathies. The activities of VEGF, are in part controlled by heparan sulfate (HS) proteoglycans, significant components of the endothelial glycocalyx. The level of 6-O sulfation on HS polysaccharide chains, that mediate the interaction between HS and VEGFA, is edited at the cell surface by the enzyme SULF1. We investigated the role of sulf1 in vascular development. In zebrafish sulf1 is expressed in the head and tail vasculature, corresponding spatially and temporally with vascular development. Targeted knockdown of sulf1 by antisense morpholinos resulted in severe vascular patterning and maturation defects. 93 % of sulf1 morphants show dysmorphogenesis in arterial development leading to occlusion of the distal aorta and lack of axial and cranial circulation. Co-injection of vegfa165 mRNA rescued circulatory defects. While the genes affecting haematopoiesis are unchanged, expression of several arterial markers downstream of VegfA signalling such as notch and ephrinB2 are severely reduced in the dorsal aorta, with a concomitant increase in expression of the venous markers flt4 in the dorsal aorta of the morphants. Furthermore, in vitro, lack of SULF1 expression downregulates VEGFA-mediated arterial marker expression, confirming that Sulf1 mediates arterial specification by regulating VegfA165 activity. This study provides the first in vivo evidence for the integral role of the endothelial glycocalyx in specifying arterial-venous identity, vascular patterning and arterial integrity, and will help to better understand congenital arteriopathies.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Signal Transduction/physiology , Sulfatases/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Zebrafish Proteins/biosynthesis , Zebrafish/metabolism , Animals , Arteries/embryology , Arteries/metabolism , Ephrin-B2/immunology , Ephrin-B2/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glycocalyx/genetics , Glycocalyx/metabolism , Morpholinos/pharmacology , Oligonucleotides, Antisense/pharmacology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/drug effects , Sulfatases/antagonists & inhibitors , Sulfatases/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , Veins/embryology , Veins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
11.
J Biol Chem ; 288(8): 5828-39, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23288839

ABSTRACT

Formylglycine-generating enzyme (FGE) post-translationally converts a specific cysteine in newly synthesized sulfatases to formylglycine (FGly). FGly is the key catalytic residue of the sulfatase family, comprising 17 nonredundant enzymes in human that play essential roles in development and homeostasis. FGE, a resident protein of the endoplasmic reticulum, is also secreted. A major fraction of secreted FGE is N-terminally truncated, lacking residues 34-72. Here we demonstrate that this truncated form is generated intracellularly by limited proteolysis mediated by proprotein convertase(s) (PCs) along the secretory pathway. The cleavage site is represented by the sequence RYSR(72)↓, a motif that is conserved in higher eukaryotic FGEs, implying important functionality. Residues Arg-69 and Arg-72 are critical because their mutation abolishes FGE processing. Furthermore, residues Tyr-70 and Ser-71 confer an unusual property to the cleavage motif such that endogenous as well as overexpressed FGE is only partially processed. FGE is cleaved by furin, PACE4, and PC5a. Processing is disabled in furin-deficient cells but fully restored upon transient furin expression, indicating that furin is the major protease cleaving FGE. Processing by endogenous furin occurs mostly intracellularly, although also extracellular processing is observed in HEK293 cells. Interestingly, the truncated form of secreted FGE no longer possesses FGly-generating activity, whereas the unprocessed form of secreted FGE is active. As always both forms are secreted, we postulate that furin-mediated processing of FGE during secretion is a physiological means of higher eukaryotic cells to regulate FGE activity upon exit from the endoplasmic reticulum.


Subject(s)
Glycine/analogs & derivatives , Proprotein Convertases/metabolism , Sulfatases/antagonists & inhibitors , Amino Acid Motifs , Animals , Arginine/chemistry , Binding Sites , CHO Cells , Cell Line, Tumor , Cricetinae , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Furin/chemistry , Glycine/chemistry , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Oxidoreductases Acting on Sulfur Group Donors , Plasmids/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proteolysis , Tyrosine/chemistry
12.
Biochimie ; 94(6): 1309-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22410212

ABSTRACT

The commonly used food additive carrageenan, including lambda (λ), kappa (κ) and iota (ι) forms, is composed of galactose disaccharides linked in alpha-1,3 and beta-1,4 glycosidic bonds with up to three sulfate groups per disaccharide residue. Carrageenan closely resembles the endogenous galactose or N-acetylgalactosamine-containing glycosaminoglycans (GAGs), chondroitin sulfate (CS), dermatan sulfate (DS), and keratan sulfate. However, these GAGs have beta-1,3 and beta-1,4 glycosidic bonds, in contrast to the unusual alpha-1,3 glycosidic bond in carrageenan. Since sulfatase activity is inhibited by sulfate, and carrageenan is so highly sulfated, we tested the effect of carrageenan exposure on sulfatase activity in human intestinal and mammary epithelial cell lines and found that carrageenan exposure significantly reduced the activity of sulfatases, including N-acetylgalactosamine-4-sulfatase, galactose-6-sulfatase, iduronate sulfatase, steroid sulfatase, arylsulfatase A, SULF-1,2, and heparan sulfamidase. Consistent with the inhibition of sulfatase activity, following exposure to carrageenan, GAG content increased significantly and showed marked differences in disaccharide composition. Specific changes in CS disaccharides included increases in di-sulfated disaccharide components of CSD (2S6S) and CS-E (4S6S), with declines in CS-A (4S) and CS-C (6S). Specific changes in heparin-heparan sulfate disaccharides included increases in 6S disaccharides, as well as increases in NS and 2S6S disaccharides. Study results suggest that carrageenan inhibition of sulfatase activity leads to re-distribution of the cellular GAG composition with increase in di-sulfated CS and with potential consequences for cell structure and function.


Subject(s)
Carrageenan/adverse effects , Food Additives/adverse effects , Sulfatases/antagonists & inhibitors , Animals , Breast/cytology , Cell Line , Cerebroside-Sulfatase/antagonists & inhibitors , Chondroitinsulfatases/antagonists & inhibitors , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Female , Humans , Hydrolases/antagonists & inhibitors , Iduronate Sulfatase/antagonists & inhibitors , Intestinal Mucosa/cytology , N-Acetylgalactosamine-4-Sulfatase/antagonists & inhibitors , Steryl-Sulfatase/antagonists & inhibitors , Sulfotransferases/antagonists & inhibitors
13.
Hepatology ; 55(6): 1746-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22234891

ABSTRACT

UNLABELLED: Type 2 diabetes mellitus (T2DM) impairs hepatic clearance of atherogenic postprandial triglyceride-rich lipoproteins (TRLs). We recently reported that livers from T2DM db/db mice markedly overexpress the heparan sulfate glucosamine-6-O-endosulfatase-2 (SULF2), an enzyme that removes 6-O sulfate groups from heparan sulfate proteoglycans (HSPGs) and suppresses uptake of TRLs by cultured hepatocytes. In the present study, we evaluated whether Sulf2 inhibition in T2DM mice in vivo could correct their postprandial dyslipidemia. Selective second-generation antisense oligonucleotides (ASOs) targeting Sulf2 were identified. Db/db mice were treated for 5 weeks with Sulf2 ASO (20 or 50 mg/kg per week), nontarget (NT) ASO, or phosphate-buffered saline (PBS). Administration of Sulf2 ASO to db/db mice suppressed hepatic Sulf2 messenger RNA expression by 70%-80% (i.e., down to levels in nondiabetic db/m mice) and increased the ratio of tri- to disulfated disaccharides in hepatic HSPGs (P < 0.05). Hepatocytes isolated from db/db mice on NT ASO exhibited a significant impairment in very-low-density lipoprotein (VLDL) binding that was entirely corrected in db/db mice on Sulf2 ASO. Sulf2 ASO lowered the random, nonfasting plasma triglyceride (TG) levels by 50%, achieving nondiabetic values. Most important, Sulf2 ASO treatment flattened the plasma TG excursions in db/db mice after corn-oil gavage (iAUC, 1,500 ± 470 mg/dL·h for NT ASO versus 160 ± 40 mg/dL · h for Sulf2 ASO\P < 0.01). CONCLUSIONS: Despite extensive metabolic derangements in T2DM mice, inhibition of a single dys-regulated molecule, SULF2, normalizes the VLDL-binding capacity of their hepatocytes and abolishes postprandial hypertriglyceridemia. These findings provide a key proof of concept in vivo to support Sulf2 inhibition as an attractive strategy to improve metabolic dyslipidemia.


Subject(s)
Diabetes Mellitus, Type 2/complications , Dyslipidemias/drug therapy , Liver/enzymology , Oligonucleotides, Antisense/therapeutic use , Sulfatases/antagonists & inhibitors , Animals , Diabetes Mellitus, Type 2/metabolism , Dyslipidemias/metabolism , Hepatocytes/metabolism , Humans , Lipoproteins, VLDL/metabolism , Male , Mice , Triglycerides/metabolism
14.
Bioorg Med Chem ; 20(2): 622-7, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21570853

ABSTRACT

Sulfatases hydrolytically cleave sulfate esters through a unique catalytic aldehyde, which is introduced by a posttranslational oxidation. To profile active sulfatases in health and disease, activity-based proteomic tools are needed. Herein, quinone methide (QM) traps directed against sulfatases are evaluated as activity-based proteomic probes (ABPPs). Starting from a p-fluoromethylphenyl sulfate scaffold, enzymatically generated QM-traps can inactivate bacterial aryl sulfatases from Pseudomonas aeruginosa and Klebsiella pneumoniae, and human steroid sulfatase. However, multiple enzyme-generated QMs form, diffuse, and non-specifically label purified enzyme. In complex proteomes, QM labeling is sulfatase-dependent but also non-specific. Thus, fluoromethylphenyl sulfates are poor ABPPs for sulfatases.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Indolequinones/chemistry , Proteomics , Sulfatases/antagonists & inhibitors , Bacterial Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Fluorescent Dyes/chemistry , Humans , Klebsiella pneumoniae/enzymology , Pseudomonas aeruginosa/enzymology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Steryl-Sulfatase/antagonists & inhibitors , Steryl-Sulfatase/metabolism , Sulfatases/metabolism
15.
Mol Cell Endocrinol ; 340(2): 120-6, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-20888390

ABSTRACT

Aromatase, estrone sulfatase, and 17ß-hydroxysteroid dehydrogenase type 1 are involved in the key steps of 17ß-estradiol biosynthesis. Structure-function studies of aromatase, estrone sulfatase and 17ß-hydroxysteroid dehydrogenase type 1 are important to evaluate the molecular basis of the interaction between these enzymes and their inhibitors. Selective and potent inhibitors of the three enzymes have been developed as antiproliferative agents in hormone-dependent breast carcinoma. New treatment strategies for hormone-dependent breast cancer are discussed.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Aromatase/metabolism , Drug Design , Enzyme Inhibitors/pharmacology , Sulfatases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/chemistry , Animals , Aromatase/chemistry , Enzyme Inhibitors/chemistry , Humans , Structure-Activity Relationship , Sulfatases/chemistry , Sulfatases/metabolism
16.
Steroids ; 75(12): 891-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20546769

ABSTRACT

4-(p-Sulphamoylphenyl)androstenedione (3) and 6alpha-p-sulphamoylphenyl analogues 12-14 were synthesised and tested as aromatase inhibitors as well as oestrone sulphatase inhibitors in human placental microsomes. All of the p-sulphamoylphenyl compounds synthesised were powerful inhibitors of aromatase with apparent K(i) values ranging between 30 and 97nM. In addition, the aromatase inhibitory activities of 6alpha-p-hydroxyphenyl compounds 9-11, which may be produced from their respective sulphamoylphenyl compounds by action of oestrone sulphatase, were also high in a range of 23 and 75nM of the K(i) values. On the other hand, all of the sulphamoylphenyl compounds were poor inhibitors of oestrone sulphatase with more than about 200microM of IC(25) values. Although the present findings of the oestrone sulphatase inhibition are disappointing, such attempts may be valuable to develop a new class of drugs having a dual function, aromatase inhibitor and oestrone sulphatase inhibitor, for the treatment of oestrogen-dependent breast cancer.


Subject(s)
Androstenedione/chemistry , Androstenedione/pharmacology , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/pharmacology , Aromatase/metabolism , Sulfatases/antagonists & inhibitors , Androstenedione/chemical synthesis , Aromatase Inhibitors/chemical synthesis , Humans , Inhibitory Concentration 50
17.
Proc Natl Acad Sci U S A ; 107(22): 10202-7, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20479257

ABSTRACT

The balance between anabolic and catabolic signaling pathways is critical in maintaining cartilage homeostasis and its disturbance contributes to joint diseases such as osteoarthritis (OA). A unique mechanism that modulates the activity of cell signaling pathways is controlled by extracellular heparan endosulfatases Sulf-1 and Sulf-2 (Sulfs) that are overexpressed in OA cartilage. This study addressed the role of Sulfs in cartilage homeostasis and in regulating bone morphogenetic protein (BMP)/Smad and fibroblast growth factor (FGF)/Erk signaling in articular cartilage. Spontaneous cartilage degeneration and surgically induced OA were significantly more severe in Sulf-1(-/-) and Sulf-2(-/-) mice compared with wild-type mice. MMP-13, ADAMTS-5, and the BMP antagonist noggin were elevated whereas col2a1 and aggrecan were reduced in cartilage and chondrocytes from Sulf(-/-) mice. Articular cartilage and cultured chondrocytes from Sulf(-/-) mice showed reduced Smad1 protein expression and Smad1/5 phosphorylation, whereas Erk1/2 phosphorylation was increased. In human chondrocytes, Sulfs siRNA reduced Smad phosphorylation but enhanced FGF-2-induced Erk1/2 signaling. These findings suggest that Sulfs simultaneously enhance BMP but inhibit FGF signaling in chondrocytes and maintain cartilage homeostasis. Approaches to correct abnormal Sulf expression have the potential to protect against cartilage degradation and promote cartilage repair in OA.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cartilage, Articular/metabolism , Fibroblast Growth Factors/metabolism , Sulfatases/metabolism , ADAM Proteins/genetics , ADAMTS5 Protein , Animals , Bone Morphogenetic Protein 7/metabolism , Carrier Proteins/genetics , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/metabolism , Homeostasis , Humans , Matrix Metalloproteinase 13/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis/etiology , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Sulfatases/antagonists & inhibitors , Sulfatases/deficiency , Sulfatases/genetics , Sulfotransferases/deficiency , Sulfotransferases/genetics , Sulfotransferases/metabolism
18.
Curr Cancer Drug Targets ; 10(3): 279-86, 2010 May.
Article in English | MEDLINE | ID: mdl-20370689

ABSTRACT

Melatonin inhibits the growth of different kinds of neoplasias, especially breast cancer, by interacting with estrogen-responsive pathways, thus behaving as an antiestrogenic hormone. Recently, we described that melatonin reduces sulfatase expression and activity in MCF-7 human breast cancer cells, thus modulating the local estrogen biosynthesis. In this study, to investigate the in vivo sulfatase-inhibitory properties of melatonin, this indoleamine was administered to ovariectomized rats bearing DMBA-induced mammary tumors, and treated with estrone sulfate. In castrated animals, the growth of estrogen-sensitive mammary tumors depends on the local conversion of biologically inactive estrogens to bioactive unconjugated estrogens. Ovariectomy significantly reduced the size and the number of the tumors while the administration of estrone sulfate to ovariectomized animals stimulated tumor growth, an effect which was suppressed by melatonin. The uterine weight of ovariectomized rats, which depends on the local synthesis of estrogens, was increased by estrone sulfate, except in those animals which were also treated with melatonin. The growth-stimulatory effects of estrone sulfate on the uterus and tumors depend exclusively on locally formed estrogens, since no changes in serum estradiol were appreciated in estrone sulfate-treated rats. Melatonin counteracted the stimulatory effects of estrone sulfate on sulfatase activity and expression and incubation with melatonin decreased the sulfatase activity of tumors from control animals. Animals treated with melatonin had the same survival probability as the castrated animals and significantly higher than the uncastrated. We conclude that melatonin could exert its antitumoral effects on hormone-dependent mammary tumors by down-regulating the sulfatase pathway of the tumoral tissue.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Melatonin/pharmacology , Sulfatases/antagonists & inhibitors , 9,10-Dimethyl-1,2-benzanthracene , Animals , Dose-Response Relationship, Drug , Estrone/analogs & derivatives , Estrone/pharmacology , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Ovariectomy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sulfatases/genetics , Sulfatases/metabolism , Time Factors , Tumor Burden , Uterus/drug effects , Uterus/pathology
19.
Anticancer Agents Med Chem ; 9(6): 599-612, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19601744

ABSTRACT

A high proportion (approximately 40%) of breast cancers are hormone-dependent and it is the female hormone estradiol (E2) that is believed to play a key role in the initiation, promotion and progression of this disease. In the fight against this disease, compounds which are potent inhibitors of the cytochrome P-450 enzyme aromatase (AR) (which catalyses the conversion of the C19 androgens to the C18 estrogens) have been the major target. However, the administration of AR inhibitors alone does not prevent the localised biosynthesis of estrone (E1) (and therefore the subsequent synthesis of E2) within breast tumour cells via alternative non-AR routes. This has therefore been the major impetus for the development of steroid sulfatase (E1STS) inhibitors. The E1STS enzyme regulates the formation of E1 from estrone sulfate (E1S), a steroid conjugate present in high concentrations in tissue and blood in women with breast cancer. The STS enzyme has also been shown to catalyse the formation of dehydroepiandrosterone (DHEA) from DHEA-sulfate (DHEAS). This is important since DHEA can be converted to 5-androstene-3beta,17beta-diol, which has been shown to possess weak estrogenic properties, however, due to the high concentration of this steroid, it is able to stimulate the growth of breast cancer cells in vitro and in vivo. Considerable progress has been made in recent years in the development of a number of potent E1STS inhibitors, as such both steroidal and non-steroidal compounds have been considered and a number of highly potent inhibitors have been produced and evaluated against what is now considered a crucial enzyme in the fight against hormone-dependent breast cancer. The review therefore summarises the work that has been undertaken todate.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Sulfatases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Female , Humans , Molecular Conformation , Structure-Activity Relationship
20.
Ann N Y Acad Sci ; 1155: 76-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19250194

ABSTRACT

Steroid sulfatase (STS) hydrolyses biologically inactive estrogen sulfates to active estrogens, while estrogen sulfotransferase (EST) sulfonates estrogens to estrogen sulfates. Information regarding the expression of STS in human breast carcinoma tissues is still very limited compared to that of aromatase or 17beta-hydroxysteroid dehydrogenases (17beta-HSDs). In our study, EST and STS immunoreactivity was detected in carcinoma cells in 50 and 84 out of 113 breast carcinomas (44.2% and 74.3%, respectively), which was also associated with mRNA levels determined by RT/real-time PCR. Using microdissection/RT-PCR analyses, EST mRNA was localized to both carcinoma and intratumoral stromal cells, whereas STS was detected only in carcinoma or parenchymal cells. STS immunoreactivity was positively associated with tumor size. EST immunoreactivity was inversely correlated with tumor size or lymph node status and was significantly associated with a decreased risk of recurrence and improved prognosis. These data suggest that both EST and STS play important roles in the regulation of in situ estrogen production in human breast cancer. In addition, EST is an independent prognostic factor in human breast carcinoma and loss of EST may result in altered estrogen metabolism in hormone-dependent breast cancer cells. An inhibition of intratumoral STS in the patients with estrogen-dependent breast carcinoma is also considered to provide more clinical benefits, especially to the patients in which primary source of an availability of intratumoral estrogen is through STS rather than aromatase.


Subject(s)
Breast Neoplasms/enzymology , Sulfatases/metabolism , Sulfotransferases/metabolism , Breast Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Humans , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sulfatases/antagonists & inhibitors , Sulfotransferases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...