Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Anal Methods ; 15(11): 1461-1469, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36876452

ABSTRACT

The fine structure of heparan sulfate (HS), the glycosaminoglycan polysaccharide component of cell surface and extracellular matrix HS proteoglycans, coordinates the complex cell signalling processes that control homeostasis and drive development in multicellular animals. In addition, HS is involved in the infection of mammals by viruses, bacteria and parasites. The current detection limit for fluorescently labelled HS disaccharides (low femtomole; 10-15 mol), has effectively hampered investigations of HS composition in small, functionally-relevant populations of cells and tissues that may illuminate the structural requirements for infection and other biochemical processes. Here, an ultra-high sensitivity method is described that utilises a combination of reverse-phase HPLC, with tetraoctylammonium bromide (TOAB) as the ion-pairing reagent and laser-induced fluorescence detection of BODIPY-FL-labelled disaccharides. The method provides an unparalleled increase in the sensitivity of detection by ∼six orders of magnitude, enabling detection in the zeptomolar range (∼10-21 moles; <1000 labelled molecules). This facilitates determination of HS disaccharide compositional analysis from minute samples of selected tissues, as demonstrated by analysis of HS isolated from the midguts of Anopheles gambiae mosquitoes that was achieved without approaching the limit of detection.


Subject(s)
Culicidae , Disaccharides , Animals , Disaccharides/analysis , Disaccharides/chemistry , Chromatography, High Pressure Liquid/methods , Heparitin Sulfate/analysis , Heparitin Sulfate/chemistry , Mammals
2.
J Mol Med (Berl) ; 101(1-2): 65-81, 2023 02.
Article in English | MEDLINE | ID: mdl-36538060

ABSTRACT

Prenatal diagnosis of congenital disease improves clinical outcomes; however, as many as 50% of congenital heart disease cases are missed by current ultrasound screening methods. This indicates a need for improved screening technology. Extracellular vesicles (EVs) have attracted enormous interest in recent years for their potential in diagnostics. EVs mediate endocrine signalling in health and disease and are known to regulate aspects of embryonic development. Here, we critically evaluate recent evidence suggesting that EVs released from the foetus are able to cross the placenta and enter the maternal circulation. Furthermore, EVs from the mother appear to be transported in the reverse direction, whilst the placenta itself acts as a source of EVs. Experimental work utilising rodent models employing either transgenically encoded reporters or application of fluorescent tracking dyes provide convincing evidence of foetal-maternal crosstalk. This is supported by clinical data demonstrating expression of placental-origin EVs in maternal blood, as well as limited evidence for the presence of foetal-origin EVs. Together, this work raises the possibility that foetal EVs present in maternal blood could be used for the diagnosis of congenital disease. We discuss the challenges faced by researchers in translating these basic science findings into a clinical non-invasive prenatal test.


Subject(s)
Extracellular Vesicles , Placenta , Pregnancy , Female , Humans , Placenta/metabolism , Extracellular Vesicles/metabolism , Fetus , Biomarkers/metabolism
3.
iScience ; 25(1): 103571, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-34984327

ABSTRACT

Mesothelioma is a cancer that typically originates in the pleura of the lungs. It rapidly invades the surrounding tissues, causing pain and shortness of breath. We compared cell lines injected either subcutaneously or intrapleurally and found that only the latter resulted in invasive and rapid growth. Pleural tumors displayed a transcriptional signature consistent with increased activity of nuclear receptors PPARα and PPARγ and with an increased abundance of endogenous PPAR-activating ligands. We found that chemical probe GW6471 is a potent, dual PPARα/γ antagonist with anti-invasive and anti-proliferative activity in vitro. However, administration of GW6471 at doses that provided sustained plasma exposure levels sufficient for inhibition of PPARα/γ transcriptional activity did not result in significant anti-mesothelioma activity in mice. Lastly, we demonstrate that the in vitro anti-tumor effect of GW6471 is off-target. We conclude that dual PPARα/γ antagonism alone is not a viable treatment modality for mesothelioma.

4.
Bioorg Med Chem ; 37: 116116, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33799173

ABSTRACT

The K+-sparing diuretic amiloride elicits anticancer activities in multiple animal models. During our recent medicinal chemistry campaign aiming to identify amiloride analogs with improved properties for potential use in cancer, we discovered novel 6-(hetero)aryl-substituted amiloride and 5-(N,N-hexamethylene)amiloride (HMA) analogs with up to 100-fold higher potencies than the parent compounds against urokinase plasminogen activator (uPA), one of amiloride's putative anticancer targets, and no diuretic or antikaliuretic effects. Here, we report the systematic evaluation of structure-property relationships (lipophilicity, aqueous solubility and in vitro metabolic stability in human and mouse liver microsomes) in twelve matched pair analogs selected from our 6-substituted amiloride and HMA libraries. Mouse plasma stability, plasma protein binding, Caco-2 cell permeability, cardiac ion channel activity and pharmacokinetics in mice (PO and IV) and rats (IV) are described alongside amiloride and HMA comparators for a subset of the four most promising matched-pair analogs. The findings combined with earlier uPA activity/selectivity and other data ultimately drove selection of two analogs (AA1-39 and AA1-41) that showed efficacy in separate mouse cancer metastasis studies.


Subject(s)
Amiloride/analogs & derivatives , Amiloride/pharmacology , Antineoplastic Agents/pharmacology , Amiloride/pharmacokinetics , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Caco-2 Cells , Drug Screening Assays, Antitumor , Female , Humans , Male , Mice, Inbred BALB C , Microsomes, Liver/drug effects , Molecular Structure , Rats, Sprague-Dawley , Structure-Activity Relationship
5.
ACS Infect Dis ; 6(7): 1844-1854, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32375471

ABSTRACT

Pantothenamides (PanAms) are potent antiplasmodials with low human toxicity currently being investigated as antimalarials with a novel mode of action. These structural analogues of pantothenate, the vitamin precursor of the essential cofactor coenzyme A, are susceptible to degradation by pantetheinase enzymes present in serum. We previously discovered that α-methylation of the ß-alanine moiety of PanAms increases their stability in serum and identified N-phenethyl-α-methyl-pantothenamide as a pantetheinase-resistant PanAm with potent, on-target, and selective antiplasmodial activity. In this study, we performed structure-activity relationship investigations to establish whether stability and potency can be improved further through alternative modification of the scissile amide bond and through substitution/modification of the phenyl ring. Additionally, for the first time, the importance of the stereochemistry of the α-methyl group was evaluated in terms of stability versus potency. Our results demonstrate that α-methylation remains the superior choice for amide modification, and that while monofluoro-substitution of the phenyl ring (that often improves ADME properties) was tolerated, N-phenethyl-α-methyl-pantothenamide remains the most potent analogue. We show that the 2S,2'R-diastereomer is far more potent than the 2R,2'R-diastereomer and that this cannot be attributed to preferential metabolic activation by pantothenate kinase, the first enzyme of the coenzyme A biosynthesis pathway. Unexpectedly, the more potent 2S,2'R-diastereomer is also more prone to pantetheinase-mediated degradation. Finally, the results of in vitro studies to assess permeability and metabolic stability of the 2S,2'R-diastereomer suggested species-dependent degradation via amide hydrolysis. Our study provides important information for the continued development of PanAm-based antimalarials.


Subject(s)
Antimalarials , Antimalarials/pharmacology , Coenzyme A/metabolism , Humans , Pantothenic Acid/analogs & derivatives , Structure-Activity Relationship
6.
J Cell Physiol ; 235(10): 6854-6861, 2020 10.
Article in English | MEDLINE | ID: mdl-31985037

ABSTRACT

Extracellular calcium (Ca2+ ) and store-operated Ca2+ entry (SOCE) govern homoeostasis in the mammalian epidermis. Multiple microRNAs (miRNA) also regulate epidermal differentiation, and raised external Ca2+ modulates the expression of several such miRNAs in keratinocytes. However, little is known about the regulation of miR-184 in keratinocytes or the roles of miR-184 in keratinocyte differentiation. Here we report that exogenous Ca2+ stimulates miR-184 expression in primary epidermal keratinocytes and that this occurs in a SOCE-dependent manner. Levels of miR-184 were raised by about 30-fold after exposure to 1.5 mM Ca2+ for 5 days. In contrast, neither phorbol ester nor 1,25-dihydroxyvitamin D3 had any effect on miR-184 levels. Pharmacologic and genetic inhibitors of SOCE abrogated Ca2+ -dependent miR-184 induction by 70% or more. Ectopic miR-184 inhibited keratinocyte proliferation and led to a fourfold increase in the expression of involucrin, a marker of early keratinocyte differentiation. Exogenous miR-184 also triggered a threefold rise in levels of cyclin E and doubled the levels of γH2AX, a marker of DNA double-strand breaks. The p21 cyclin-dependent kinase inhibitor, which supports keratinocyte growth arrest, was also induced by miR-184. Together our findings point to an SOCE:miR-184 pathway that targets a cyclin E/DNA damage regulatory node to facilitate keratinocyte differentiation.


Subject(s)
Calcium/metabolism , Cell Differentiation/physiology , Keratinocytes/metabolism , MicroRNAs/metabolism , Cell Proliferation/physiology , Cells, Cultured , DNA Damage/physiology , Epidermal Cells/metabolism , Epidermis/metabolism , Humans , Protein Precursors/metabolism , Signal Transduction/physiology , Vitamin D/analogs & derivatives , Vitamin D/metabolism
7.
Inorg Chem ; 58(5): 3382-3395, 2019 Mar 04.
Article in English | MEDLINE | ID: mdl-30785268

ABSTRACT

Amyloid-ß plaques, consisting of aggregated amyloid-ß peptides, are one of the pathological hallmarks of Alzheimer's disease. Copper complexes formed using positron-emitting copper radionuclides that cross the blood-brain barrier and bind to specific molecular targets offer the possibility of noninvasive diagnostic imaging using positron emission tomography. New thiosemicarbazone-pyridylhydrazone based ligands that incorporate pyridyl-benzofuran functional groups designed to bind amyloid-ß plaques have been synthesized. The ligands form stable complexes with copper(II) ( Kd = 10-18 M) and can be radiolabeled with copper-64 at room temperature. Subtle changes to the periphery of the ligand backbone alter the metabolic stability of the complexes in mouse and human liver microsomes, and influenced the ability of the complexes to cross the blood-brain barrier in mice. A lead complex was selected based on possessing the best metabolic stability and brain uptake in mice. Synthesis of this lead complex with isotopically enriched copper-65 allowed us to show that the complex bound to amyloid-ß plaques present in post-mortem human brain tissue using laser ablation-inductively coupled plasma-mass spectrometry. This work provides insight into strategies to target metal complexes to amyloid-ß plaques, and how small modifications to ligands can dramatically alter the metabolic stability of metal complexes as well as their ability to cross the blood-brain barrier.


Subject(s)
Alzheimer Disease/diagnostic imaging , Coordination Complexes/chemistry , Positron-Emission Tomography , Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Binding Sites/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Copper Radioisotopes , Humans , Ligands , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure
8.
Glycoconj J ; 34(3): 405-410, 2017 06.
Article in English | MEDLINE | ID: mdl-27523650

ABSTRACT

Studying polysaccharide-protein interactions under physiological conditions by conventional techniques is challenging. Ideally, macromolecules could be followed by both in vitro spectroscopy experiments as well as in tissues using microscopy, to enable a proper comparison of results over these different scales but, often, this is not feasible. The cell surface and extracellular matrix polysaccharides, glycosaminoglycans (GAGs) lack groups that can be detected selectively in the biological milieu. The introduction of 19F labels into GAG polysaccharides is explored and the interaction of a labelled GAG with the heparin-binding protein, antithrombin, employing 19F NMR spectroscopy is followed. Furthermore, the ability of 19F labelled GAGs to be imaged using CARS microscopy is demonstrated. 19F labelled GAGs enable both 19F NMR protein-GAG binding studies in solution at the molecular level and non-linear microscopy at a microscopic scale to be conducted on the same material, essentially free of background signals.


Subject(s)
Fluorine-19 Magnetic Resonance Imaging/methods , Fluorine/chemistry , Glycosaminoglycans/chemistry , Molecular Probes/chemistry , Staining and Labeling/methods , Acetylation , Antithrombins/chemistry , Glycosaminoglycans/analysis , Halogenation , Magnetic Resonance Spectroscopy/methods , Molecular Probes/analysis , Protein Binding , Solutions , Spectrum Analysis, Raman/methods
9.
Mol Biosyst ; 12(10): 3166-75, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27502551

ABSTRACT

Heparin/heparan sulfate (HS) glycosaminoglycans are required for Slit-Robo cellular responses. Evidence exists for interactions between each combination of Slit, Robo and heparin/HS and for formation of a ternary complex. Heparin/HS are complex mixtures displaying extensive structural diversity. The relevance of this diversity has been studied to a limited extent using a few select chemically-modified heparins as models of HS diversity. Here we extend these studies by parallel screening of structurally diverse panels of eight chemically-modified heparin polysaccharides and numerous natural HS oligosaccharide chromatographic fractions for binding to both Drosophila Slit and Robo N-terminal domains and for activation of a chick retina axon response to the Slit fragment. Both the polysaccharides and oligosaccharide fractions displayed variability in binding and cellular activity that could not be attributed solely to increasing sulfation, extending evidence for the importance of structural diversity to natural HS as well as model modified heparins. They also displayed differences in their interactions with Slit compared to Robo, with Robo preferring compounds with higher sulfation. Furthermore, the patterns of cellular activity across compounds were different to those for binding to each protein, suggesting that biological outcomes are selectively determined in a subtle manner that does not simply reflect the sum of the separate interactions of heparin/HS with Slit and Robo.


Subject(s)
Drosophila Proteins/chemistry , Heparin/chemistry , Heparitin Sulfate/chemistry , Nerve Tissue Proteins/chemistry , Receptors, Immunologic/chemistry , Animals , Axons/metabolism , Chick Embryo , Chromatography , Drosophila , Drosophila Proteins/metabolism , Heparin/metabolism , Heparitin Sulfate/metabolism , Molecular Structure , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Immunologic/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Roundabout Proteins
10.
Eur J Med Chem ; 110: 43-64, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26807544

ABSTRACT

Malaria remains a global health problem, and though international efforts for treatment and eradication have made some headway, the emergence of drug-resistant parasites threatens this progress. Antimalarial therapeutics acting via novel mechanisms are urgently required. Plasmodium falciparum M1 and M17 are neutral aminopeptidases which are essential for parasite growth and development. Previous work in our group has identified inhibitors capable of dual inhibition of PfA-M1 and PfA-M17, and revealed further regions within the protease S1 pockets that could be exploited in the development of ligands with improved inhibitory activity. Herein, we report the structure-based design and synthesis of novel hydroxamic acid analogues that are capable of potent inhibition of both PfA-M1 and PfA-M17. Furthermore, the developed compounds potently inhibit Pf growth in culture, including the multi-drug resistant strain Dd2. The ongoing development of dual PfA-M1/PfA-M17 inhibitors continues to be an attractive strategy for the design of novel antimalarial therapeutics.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Antimalarials/pharmacology , Hydroxamic Acids/pharmacology , Plasmodium falciparum/drug effects , Protease Inhibitors/pharmacology , Protozoan Proteins/antagonists & inhibitors , Aminopeptidases/chemistry , Aminopeptidases/metabolism , Antimalarials/chemistry , HEK293 Cells , Humans , Hydroxamic Acids/chemistry , Malaria, Falciparum/drug therapy , Models, Molecular , Protease Inhibitors/chemistry , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Structure-Activity Relationship , Zinc/metabolism
11.
J R Soc Interface ; 12(110): 0589, 2015 Sep 06.
Article in English | MEDLINE | ID: mdl-26289657

ABSTRACT

Heparan sulfate (HS) polysaccharides are ubiquitous components of the cell surface and extracellular matrix of all multicellular animals, whereas heparin is present within mast cells and can be viewed as a more sulfated, tissue-specific, HS variant. HS and heparin regulate biological processes through interactions with a large repertoire of proteins. Owing to these interactions and diverse effects observed during in vitro, ex vivo and in vivo experiments, manifold biological/pharmacological activities have been attributed to them. The properties that have been thought to bestow protein binding and biological activity upon HS and heparin vary from high levels of sequence specificity to a dependence on charge. In contrast to these opposing opinions, we will argue that the evidence supports both a level of redundancy and a degree of selectivity in the structure-activity relationship. The relationship between this apparent redundancy, the multi-dentate nature of heparin and HS polysaccharide chains, their involvement in protein networks and the multiple binding sites on proteins, each possessing different properties, will also be considered. Finally, the role of cations in modulating HS/heparin activity will be reviewed and some of the implications for structure-activity relationships and regulation will be discussed.


Subject(s)
Heparin , Heparitin Sulfate , Mast Cells , Proteins , Animals , Binding Sites , Heparin/chemistry , Heparin/metabolism , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Humans , Mast Cells/chemistry , Mast Cells/metabolism , Proteins/chemistry , Proteins/metabolism , Structure-Activity Relationship
12.
Org Biomol Chem ; 13(21): 6066-72, 2015 Jun 07.
Article in English | MEDLINE | ID: mdl-25952831

ABSTRACT

Phage display antibodies are widely used to follow heparan sulfate (HS) expression in tissues and cells. We demonstrate by ELISA, that cations alter phage display antibody binding profiles to HS and this is mediated by changes in polysaccharide conformation, demonstrated by circular dichroism spectroscopy. Native HS structures, expressed on the cell surfaces of neuroblastoma and fibroblast cells, also exhibited altered antibody binding profiles following exposure to low mM concentrations of these cations. Phage display antibodies recognise conformationally-defined HS epitopes, rather than sequence alone, as has been assumed, and resemble proteins in being sensitive to changes in both charge distribution and conformation following binding of cations to HS polysaccharides.


Subject(s)
Antibodies/immunology , Epitopes/immunology , Heparitin Sulfate/immunology , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Cations/immunology , Cell Line , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Heparitin Sulfate/chemistry , Humans , Mice , Peptide Library
13.
ChemMedChem ; 9(7): 1556-66, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24677741

ABSTRACT

The complex natural products silvestrol (1) and episilvestrol (2) are inhibitors of translation initiation through binding to the DEAD-box helicase eukaryotic initiation factor 4A (eIF4A). Both compounds are potently cytotoxic to cancer cells in vitro, and 1 has demonstrated efficacy in vivo in several xenograft cancer models. Here we show that 2 has limited plasma membrane permeability and is metabolized in liver microsomes in a manner consistent with that reported for 1. In addition, we have prepared a series of analogues of these compounds where the complex pseudo-sugar at C6 has been replaced with chemically simpler moieties to improve drug-likeness. Selected compounds from this work possess excellent activity in biochemical and cellular translation assays with potent activity against leukemia cell lines.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Triterpenes/chemistry , Triterpenes/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Eukaryotic Initiation Factor-4A/chemistry , Eukaryotic Initiation Factor-4A/metabolism , Humans , Microsomes, Liver/metabolism , Molecular Conformation , Protein Binding , Triterpenes/metabolism
14.
Bioorg Med Chem Lett ; 23(24): 6868-73, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24169234

ABSTRACT

Cinnamoylanthranilates including tranilast have been identified as promising antifibrotics that can reduce fibrosis occurring in the kidney during diabetes, thereby delaying and/or preventing kidney dysfunction. Structure-activity relationships aimed at improving potency and metabolic stability have led to the discovery of FT061. This compound, which bears a bis-difluoromethoxy catechol, attenuates TGF-ß-stimulated production of collagen in cultured renal mesangial cells (approx 50% at 3 µM). When dosed orally at 20mg/kg to male Sprague Dawley rats, FT061 exhibited a high bioavailability (73%), Cmax of 200 µM and Tmax of 150 min, and a half-life of 5.4h. FT061 reduced albuminuria when orally dosed in rats at 200 mg kg/day in a late intervention study of a rat model of progressive diabetic nephropathy.


Subject(s)
Albuminuria/drug therapy , Antifibrinolytic Agents/therapeutic use , Caffeic Acids/chemistry , ortho-Aminobenzoates/chemistry , Administration, Oral , Albuminuria/complications , Albuminuria/metabolism , Animals , Antifibrinolytic Agents/chemistry , Antifibrinolytic Agents/pharmacokinetics , Caffeic Acids/pharmacokinetics , Caffeic Acids/therapeutic use , Cells, Cultured , Collagen/metabolism , Diabetic Nephropathies/complications , Diabetic Nephropathies/metabolism , Disease Models, Animal , Half-Life , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , ortho-Aminobenzoates/pharmacokinetics , ortho-Aminobenzoates/pharmacology , ortho-Aminobenzoates/therapeutic use
15.
Chem Biol ; 19(5): 553-8, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22633407

ABSTRACT

Array methodologies have become powerful tools for interrogation of glycan-protein interactions but have critically lacked the ability to generate cell response data. Here, we report the development of a slide-based array method exemplified by measurement of activation of fibroblast growth factor signaling by heparin saccharides. Heparan sulfate-deficient Swiss 3T3 cells were overlaid onto an aminosilane-coated slide surface onto which heparin saccharides had been spotted and immobilized. The cells were transiently stimulated with FGF2 and immunofluorescence measured to assess downstream ERK1/2 phosphorylation. Activation of this signaling pathway response was restricted to cells exposed to heparin saccharides competent to activate FGF2 signaling. Differential activation of the overlaid cells by different-sized heparin saccharides was demonstrated by quantitative measurement of fluorescence intensity. This "glycobioarray" platform has significant potential as a generic tool for functional glycomics screening.


Subject(s)
Fibroblast Growth Factors/metabolism , Heparin/metabolism , Microarray Analysis/methods , 3T3 Cells , Animals , Fibroblast Growth Factor 2/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Signal Transduction
16.
J Exp Med ; 209(4): 837-54, 2012 Apr 09.
Article in English | MEDLINE | ID: mdl-22473957

ABSTRACT

Parkinson's disease (PD) is a progressive, chronic disease characterized by dyskinesia, rigidity, instability, and tremors. The disease is defined by the presence of Lewy bodies, which primarily consist of aggregated α-synuclein protein, and is accompanied by the loss of monoaminergic neurons. Current therapeutic strategies only give symptomatic relief of motor impairment and do not address the underlying neurodegeneration. Hence, we have identified Cu(II)(atsm) as a potential therapeutic for PD. Drug administration to four different animal models of PD resulted in improved motor and cognition function, rescued nigral cell loss, and improved dopamine metabolism. In vitro, this compound is able to inhibit the effects of peroxynitrite-driven toxicity, including the formation of nitrated α-synuclein oligomers. Our results show that Cu(II)(atsm) is effective in reversing parkinsonian defects in animal models and has the potential to be a successful treatment of PD.


Subject(s)
Cognition/drug effects , Motor Activity/drug effects , Neuroprotective Agents/therapeutic use , Organometallic Compounds/therapeutic use , Parkinson Disease/drug therapy , Radiopharmaceuticals/therapeutic use , Thiosemicarbazones/therapeutic use , Animals , Cell Line, Tumor , Coordination Complexes , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Neuroprotective Agents/pharmacology , Organometallic Compounds/pharmacology , Parkinson Disease/psychology , Positron-Emission Tomography , Radiopharmaceuticals/pharmacology , Rats , Rats, Sprague-Dawley , Thiosemicarbazones/pharmacology , alpha-Synuclein/chemistry
17.
Eur J Heart Fail ; 14(5): 549-62, 2012 May.
Article in English | MEDLINE | ID: mdl-22417655

ABSTRACT

AIMS: Cardiac remodelling in diabetes includes pathological accumulation of extracellular matrix and myocyte hypertrophy that contribute to heart dysfunction. Attenuation of remodelling represents a potential therapeutic target. We tested this hypothesis using a new anti-fibrotic drug, FT011 (Fibrotech Therapeutics Pty Ltd), on diabetic Ren-2 rats, a model which replicates many of the structural and functional manifestations of diabetic cardiomyopathy in humans. METHODS AND RESULTS: Homozygous Ren-2 rats were randomized to receive streptozotocin or vehicle then further randomized to FT011 (200 mg/kg/day) or vehicle treatment for 6 weeks. Prior to tissue collection, cardiac function was assessed via echocardiography and cardiac catheterization. Total collagen deposition and cardiomyocyte hypertrophy were assessed by picrosirius red and haematoxylin and eosin staining, respectively. Macrophage interstitial infiltration and type I and III collagen were quantitated by immunostaining. Without affecting blood pressure or hyperglycaemia, treatment of diabetic rats with FT011 significantly attenuated interstitial fibrosis (total collagen, 5.09 ±1.28 vs, 2.42 ±0.43%/area; type I collagen, 4.09 ±1.16 vs. 1.42 ±0.38%/area; type III collagen, 1.52 ±0.33 vs. 0.71 ±0.14 %/area; P < 0.05), cardiomyocyte hypertrophy (882 ±38 vs. 659 ±28 µm(2); P < 0.05), and interstitial macrophage influx (66 ±5.3 vs, 44 ±7.9 number/section; P < 0.05). Cardiac myopathic dilatation was normalized, as evidenced by reduced left ventricular inner diameter at diastole (0.642 ±0.016 vs. 0.577 ±0.024 cm), increased ejection fraction (75 ±1.1 vs. 83 ±1.2%) and preload recruitable stroke work relationship (44 ±6.7 vs. 77 ±6.3 slope-mmHg; P < 0.05), and reduced end-diastolic pressure-volume relationship (0.059 ±0.011 vs. 0.02 ±0.003 slope-mmHg/µL; P < 0.05). CONCLUSIONS: A direct anti-fibrotic agent, FT011, attenuates cardiac remodelling and dysfunction in experimental diabetic cardiomyopathy. This represents a novel therapy for the treatment of diabetic cardiomyopathy associated with cardiac fibrosis and hypertrophy.


Subject(s)
Antifibrinolytic Agents/therapeutic use , Caffeic Acids/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetic Cardiomyopathies/drug therapy , Heart Failure/drug therapy , Myocardium/pathology , Myocytes, Cardiac/pathology , ortho-Aminobenzoates/therapeutic use , Animals , Cardiac Catheterization , Cardiomegaly/etiology , Cardiomegaly/pathology , Chronic Disease , Collagen/metabolism , Fibrosis/drug therapy , Heart Failure/complications , Heart Failure/metabolism , Immunohistochemistry , Male , Rats , Rats, Inbred Strains , Rats, Sprague-Dawley , Streptozocin
18.
J Pharmacol Exp Ther ; 339(3): 799-806, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21917561

ABSTRACT

The orally active microtubule-disrupting agent (S)-1-ethyl-3-(2-methoxy-4-(5-methyl-4-((1-(pyridin-3-yl)butyl)amino)pyrimidin-2-yl)phenyl)urea (CYT997), reported previously by us (Bioorg Med Chem Lett 19:4639-4642, 2009; Mol Cancer Ther 8:3036-3045, 2009), is potently cytotoxic to a variety of cancer cell lines in vitro and shows antitumor activity in vivo. In addition to its cytotoxic activity, CYT997 possesses antivascular effects on tumor vasculature. To further characterize the vascular disrupting activity of CYT997 in terms of dose and temporal effects, we studied the activity of the compound on endothelial cells in vitro and on tumor blood flow in vivo by using a variety of techniques. In vitro, CYT997 is shown to potently inhibit the proliferation of vascular endothelial growth factor-stimulated human umbilical vein endothelial cells (IC(50) 3.7 ± 1.8 nM) and cause significant morphological changes at 100 nM, including membrane blebbing. Using the method of corrosion casting visualized with scanning electron microscopy, a single dose of CYT997 (7.5 mg/kg i.p.) in a metastatic cancer model was shown to cause destruction of tumor microvasculature in metastatic lesions. Furthermore, repeat dosing of CYT997 at 10 mg/kg and above (intraperitoneally, b.i.d.) was shown to effectively inhibit development of liver metastases. The time and dose dependence of the antivascular effects were studied in a DLD-1 colon adenocarcinoma xenograft model using the fluorescent dye Hoechst 33342. CYT997 demonstrated rapid and dose-dependent vascular shutdown, which persists for more than 24 h after a single oral dose. Together, the data demonstrate that CYT997 possesses potent antivascular activity and support continuing development of this promising compound.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Pyridines/pharmacology , Pyrimidines/pharmacology , Tubulin Modulators/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Male , Mice , Mice, Nude , Time Factors , Xenograft Model Antitumor Assays
19.
Anal Biochem ; 411(2): 261-70, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21211507

ABSTRACT

Here we describe a versatile high-throughput expression system that permits genome-wide screening of type 1 membrane and secreted proteins for interactions with glycans and proteins using both cell-expressed and soluble forms of the expressed proteins. Based on Gateway cloning methodology, we have engineered a destination vector that directs expression of enhanced green fluorescent protein (EGFP)-tagged proteins at the cell surface via a glycosylphosphatidylinositol tail. The EGFP fusion proteins can then be cleaved with PreScission protease to release soluble forms of proteins that can be optionally biotinylated. We demonstrate the utility of this cloning and expression system for selected low-affinity membrane lectins from the siglec family of sialic acid-binding immunoglobulin-like lectins, for the glycosaminoglycan-binding proteins FGF-1 and BACE, and for the heterotypic adhesion molecules JAM-B and JAM-C. Cell-expressed proteins can be evaluated for glycan interactions using polyvalent soluble glycan probes and for protein interactions using either cells or soluble proteins. Following cleavage from the cell surface, proteins were complexed in solution and sufficient avidity was achieved to measure weak protein-glycan and weak protein-protein interactions using glycan arrays and surface plasmon resonance, respectively.


Subject(s)
Membrane Proteins/chemistry , Polysaccharides/chemistry , Protein Array Analysis/methods , Surface Plasmon Resonance/methods , Aspartic Acid Endopeptidases/chemistry , Carbohydrate Sequence , Cell Adhesion Molecules/chemistry , Fibroblast Growth Factor 1/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lectins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins
20.
Nat Protoc ; 5(5): 821-33, 2010 May.
Article in English | MEDLINE | ID: mdl-20379137

ABSTRACT

Natural and semi-synthetic heparan sulfate (HS) saccharide libraries are a valuable resource for investigating HS structure-function relationships, enabling high-throughput glycomics studies. Owing to the difficulty of chemical or in vitro enzymatic synthesis of HS saccharides, the structural diversity displayed in saccharides from tissue or cell sources cannot be readily accessed. In contrast, saccharide libraries can be generated by partial digestion of tissue-derived HS polysaccharide chains and chromatographic fractionation of the resulting saccharide mixtures. Fractionation is initially on the basis of hydrodynamic volume, using size exclusion chromatography. Further fractionation, on the basis of charge using strong anion exchange, can subsequently be applied. Desalting and sample concentration follows each fractionation step. Chromatographic fractions are generated that contain purified, or partially purified, saccharides. Here we describe a comprehensive protocol for generation of structurally diverse natural saccharide libraries from HS variants that is fast (approximately 3 weeks) and reproducible.


Subject(s)
Glycomics/methods , Heparitin Sulfate/chemistry , Chromatography , Heparin Lyase , Heparitin Sulfate/isolation & purification , Molecular Structure , Monosaccharides/chemistry , Monosaccharides/isolation & purification , Oligosaccharides/chemistry , Oligosaccharides/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...