Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 15(726): eadg8105, 2023 12 13.
Article in English | MEDLINE | ID: mdl-38091410

ABSTRACT

Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, affects millions of people in the Americas and across the world, leading to considerable morbidity and mortality. Current treatment options, benznidazole (BNZ) and nifurtimox, offer limited efficacy and often lead to adverse side effects because of long treatment durations. Better treatment options are therefore urgently required. Here, we describe a pyrrolopyrimidine series, identified through phenotypic screening, that offers an opportunity to improve on current treatments. In vitro cell-based washout assays demonstrate that compounds in the series are incapable of killing all parasites; however, combining these pyrrolopyrimidines with a subefficacious dose of BNZ can clear all parasites in vitro after 5 days. These findings were replicated in a clinically predictive in vivo model of chronic Chagas disease, where 5 days of treatment with the combination was sufficient to prevent parasite relapse. Comprehensive mechanism of action studies, supported by ligand-structure modeling, show that compounds from this pyrrolopyrimidine series inhibit the Qi active site of T. cruzi cytochrome b, part of the cytochrome bc1 complex of the electron transport chain. Knowledge of the molecular target enabled a cascade of assays to be assembled to evaluate selectivity over the human cytochrome b homolog. As a result, a highly selective and efficacious lead compound was identified. The combination of our lead compound with BNZ rapidly clears T. cruzi parasites, both in vitro and in vivo, and shows great potential to overcome key issues associated with currently available treatments.


Subject(s)
Chagas Disease , Parasites , Trypanocidal Agents , Trypanosoma cruzi , Animals , Humans , Cytochromes b , Trypanocidal Agents/adverse effects , Chagas Disease/drug therapy , Chagas Disease/chemically induced , Chagas Disease/parasitology
2.
Physiol Genomics ; 41(3): 224-31, 2010 May.
Article in English | MEDLINE | ID: mdl-20197419

ABSTRACT

Failure to express or expression of dysfunctional low-density lipoprotein receptors (LDLR) causes familial hypercholesterolemia in humans, a disease characterized by elevated blood cholesterol concentrations, xanthomas, and coronary heart disease, providing compelling evidence that high blood cholesterol concentrations cause atherosclerosis. In this study, we used (1)H nuclear magnetic resonance spectroscopy to examine the metabolic profiles of plasma and urine from the LDLR knockout mice. Consistent with previous studies, these mice developed hypercholesterolemia and atherosclerosis when fed a high-fat/cholesterol/cholate-containing diet. In addition, multivariate statistical analysis of the metabolomic data highlighted significant differences in tricarboxylic acid cycle and fatty acid metabolism, as a result of high-fat/cholesterol diet feeding. Our metabolomic study also demonstrates that the effect of high-fat/cholesterol/cholate diet, LDLR gene deficiency, and the diet-genotype interaction caused a significant perturbation in choline metabolism, notably the choline oxidation pathway. Specifically, the loss in the LDLR caused a marked reduction in the urinary excretion of betaine and dimethylglycine, especially when the mice are fed a high-fat/cholesterol/cholate diet. Furthermore, as we demonstrate that these metabolic changes are comparable with those detected in ApoE knockout mice fed the same high-fat/cholesterol/cholate diet they may be useful for monitoring the onset of atherosclerosis across animal models.


Subject(s)
Apolipoproteins E/deficiency , Choline/metabolism , Diet, High-Fat , Metabolomics/methods , Receptors, LDL/deficiency , Animals , Apolipoproteins E/metabolism , Atherosclerosis/blood , Atherosclerosis/metabolism , Cholesterol/blood , Databases, Genetic , Diet , Discriminant Analysis , Female , Genotype , Hypercholesterolemia/blood , Hypercholesterolemia/metabolism , Least-Squares Analysis , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Proton Magnetic Resonance Spectroscopy , Receptors, LDL/metabolism
3.
Am J Pathol ; 170(1): 416-26, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200212

ABSTRACT

We explored the role of the classic complement pathway in atherogenesis by intercrossing C1q-deficient mice (C1qa-/-) with low-density lipoprotein receptor knockout mice (Ldlr-/-). Mice were fed a normal rodent diet until 22 weeks of age. Aortic root lesions were threefold larger in C1qa-/-/Ldlr-/- mice compared with Ldlr-/- mice (3.72 +/- 1.0% aortic root versus 1.1 +/- 0.4%; mean +/- SEM, P < 0.001). Furthermore, the cellular composition of lesions in C1qa-/-/Ldlr-/- was more complex, with an increase in vascular smooth muscle cells. The greater aortic root lesion size in C1qa-/-/Ldlr-/- mice occurred despite a significant reduction in C5b-9 deposition per lesion unit area, suggesting the critical importance of proximal pathway activity. Apoptotic cells were readily detectable by cleaved caspase-3 staining, terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, and electron microscopy in C1qa-/-/Ldlr-/-, whereas apoptotic cells were not detected in Ldlr-/- mice. This is the first direct demonstration of a role for the classic complement pathway in atherogenesis. The greater lesion size in C1qa-/-/Ldlr-/- mice is consistent with the emerging homeostatic role for C1q in the disposal of dying cells. This study suggests the importance of effective apoptotic cell removal for containing the size and complexity of early lesions in atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Complement C1q/physiology , Complement Pathway, Classical , Receptors, LDL/deficiency , Animals , Apoptosis/physiology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Mice , Mice, Knockout , Receptors, LDL/genetics
4.
Dis Markers ; 21(4): 181-90, 2005.
Article in English | MEDLINE | ID: mdl-16403953

ABSTRACT

Dietary antioxidants are reported to suppress cellular expression of chemokines and adhesion molecules that recruit monocytes to the artery wall during atherosclerosis. In the present study we measured the effect of feeding apoE*3 Leiden mice or their non-transgenic (C57BL) littermates with atherogenic diets either deficient in, or supplemented with, dietary antioxidants (vitamin E, vitamin C and beta-carotene) for 12 weeks, on serum levels of CC (JE/MCP-1) and CXC (KC) chemokines and soluble adhesion molecules (sVCAM-1, sICAM-1) and atherosclerotic lesion size. ApoE*3 Leiden mice developed gross hypercholesterolaemia, and markedly accelerated (10-20 fold; P < 0.0001) atherogenesis, compared with non-transgenic animals. Antioxidant consumption reduced lesion area in non-transgenic, but not apoE*3 Leiden, mice. Serum sVCAM-1 and sICAM-1 levels were significantly (P<0.0001) increased (sVCAM-1 up to 3.9 fold; sICAM-1 up to 2.4 fold) by 4-8 weeks in all groups, and then declined. The initial increase in the concentration of adhesion molecules was reduced by 38%-61% (P < 0.05) by antioxidant consumption, particularly in non-transgenic mice. By contrast, serum chemokine levels tended to increase more rapidly from baseline in apoE*3 Leiden mice, compared with non-transgenic animals, but were unaffected by dietary antioxidants. We conclude that dietary antioxidants reduce circulating soluble adhesion molecules and atherosclerosis in C57BL mice.


Subject(s)
Antioxidants/administration & dosage , Atherosclerosis/diet therapy , Chemokine CCL2/blood , Chemokine CXCL1/blood , Down-Regulation/physiology , Hypercholesterolemia/diet therapy , Intercellular Adhesion Molecule-1/blood , Vascular Cell Adhesion Molecule-1/blood , Animal Feed , Animals , Antioxidants/physiology , Apolipoproteins C/genetics , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/biosynthesis , Chemokine CXCL1/antagonists & inhibitors , Chemokine CXCL1/biosynthesis , Female , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , Intercellular Adhesion Molecule-1/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Random Allocation , Vascular Cell Adhesion Molecule-1/biosynthesis
5.
Pathol Oncol Res ; 10(3): 159-65, 2004.
Article in English | MEDLINE | ID: mdl-15448752

ABSTRACT

High-resolution, non-invasive imaging methods are required to monitor progression and regression of atherosclerotic plaques. We investigated the use of MRI to measure changes in plaque volume and vessel remodelling during progression and regression of atherosclerosis in New Zealand White rabbits. Atherosclerotic lesions were induced in the abdominal aorta by balloon injury and cholesterol feeding. MR images (2D) of the abdominal aorta were acquired with cardiac and respiratory gating using a fast spin echo sequence with and without fat-suppression. In an initial study on rabbits treated for 30 weeks we imaged the aortae with a spatial resolution of 250x250 micrometers with a slice thickness of 2 mm and achieved a close correlation between MRI-derived measurements and those made on perfusion pressure-fixed histological sections (r(1) = 0.83, slope p(1) < 0.01). We subsequently imaged 18 rabbits before and periodically during 12 weeks of cholesterol feeding (progression) followed by 12 weeks on normal diet (regression). Aortic wall (atherosclerotic lesion) volume increased significantly during progression and decreased during regression. In contrast, lumen volume increased during progression and did not change during regression. In conclusion, this study confirms that non-invasive, high-resolution MRI can be used to monitor progression and regression of atherosclerosis, each within 3 months and shows, for the first time in a short-term model, that positive remodelling occurs early during progression and persists through regression of atherosclerotic lesions.


Subject(s)
Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/pathology , Arteriosclerosis/diagnostic imaging , Arteriosclerosis/pathology , Cholesterol, Dietary , Animals , Aorta, Abdominal/metabolism , Arteriosclerosis/metabolism , Diet, Atherogenic , Disease Models, Animal , Female , Image Processing, Computer-Assisted , Immunohistochemistry , Magnetic Resonance Imaging , Rabbits , Radiography
6.
Arterioscler Thromb Vasc Biol ; 23(9): 1615-20, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12842836

ABSTRACT

OBJECTIVE: CC and CXC chemokines are implicated in leukocyte recruitment during development of atherosclerotic lesions, suggesting circulating levels of chemokines may be useful serum markers of atherogenesis. Serum chemokine concentrations were measured in apolipoprotein (apo) E*3 Leiden mice and their nontransgenic littermates and related to the differing rates of atherogenesis in these animals. METHODS AND RESULTS: Mice were fed a high-fat, high-cholesterol/cholate (HFC/C) diet for 18 weeks. Circulating levels of JE/monocyte chemotactic protein-1 increased (P<0.05) after 2 to 4 weeks, coincident with development of diet-induced hypercholesterolemia, and remained elevated throughout the study. Circulating KC concentrations increased (P<0.05) after consumption of HFC/C diet; however, unlike JE, serum KC concentrations increased more rapidly in apoE*3 Leiden mice than their nontransgenic littermates. Hepatic expression of JE and KC mRNA were detected by in situ hybridization in all mice fed HFC/C diet. Aortic expression of JE mRNA was seen only in apoE*3 Leiden mice within macrophage-rich atherosclerotic lesions. By contrast, no aortic expression of KC mRNA was detected by in situ hybridization. CONCLUSIONS: Increases in serum chemokine concentrations did not reflect temporal aortic production of these molecules and proved less predictive than serum cholesterol of the markedly different extent of atheroma in apoE*3 Leiden and nontransgenic mice.


Subject(s)
Arteriosclerosis/etiology , Chemokines, CC/blood , Chemokines, CXC/blood , Animals , Aorta/chemistry , Aorta/metabolism , Apolipoprotein E3 , Apolipoproteins E/genetics , Arteriosclerosis/blood , Arteriosclerosis/pathology , Chemokines, CC/biosynthesis , Chemokines, CXC/biosynthesis , Cholesterol/blood , Diet, Atherogenic , Lipids/blood , Lipoproteins/blood , Liver/chemistry , Liver/metabolism , Macrophages/metabolism , Mice
7.
Br J Nutr ; 89(3): 341-50, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12628029

ABSTRACT

We have compared lipoprotein metabolism in, and susceptibility to atherosclerosis of, two strains of male Golden Syrian hamster, the Bio F(1)B hybrid and the dominant spot normal inbred (DSNI) strain. When fed a normal low-fat diet containing approximately 40 g fat and 0.3 g cholesterol/kg, triacylglycerol-rich lipoprotein (chylomicron+VLDL) and HDL-cholesterol were significantly higher (P<0.001) in Bio F(1)B hamsters than DSNI hamsters. When this diet was supplemented with 150 g coconut oil and either 0.5 or 5.0 g cholesterol/kg, significant differences were seen in response. In particular, the high-cholesterol diet produced significantly greater increases in plasma cholesterol and triacylglycerol in the Bio F(1)B compared with the DSNI animals (P=0.002 and P<0.001 for cholesterol and triacylglycerol, respectively). This was particularly dramatic in non-fasting animals, suggesting an accumulation of chylomicrons. In a second experiment, animals were fed 150 g coconut oil/kg and 5.0 g cholesterol/kg for 6 and 12 months. Again, the Bio F(1)B animals showed dramatic increases in plasma cholesterol and triacylglycerol, and this was confirmed as primarily due to a rise in chylomicron concentration. Post-heparin lipoprotein lipase activity was significantly reduced (P<0.001) in the Bio F(1)B compared with the DSNI animals at 6 months, and virtually absent at 12 months. Bio F(1)B animals were also shown to develop significantly more (P<0.001) atherosclerosis. These results indicate that, in the Bio F(1)B hybrid hamster, cholesterol feeding reduces lipoprotein lipase activity, thereby causing the accumulation of chylomicrons that may be associated with their increased susceptibility to atherosclerosis.


Subject(s)
Arteriosclerosis/enzymology , Cholesterol, Dietary/pharmacology , Disease Susceptibility , Lipoprotein Lipase/blood , Animals , Cholesterol, Dietary/administration & dosage , Chylomicrons/metabolism , Coconut Oil , Cricetinae , Diet , Dietary Fats/pharmacology , Lipoprotein Lipase/drug effects , Liver/metabolism , Liver/pathology , Male , Mesocricetus , Organ Size/drug effects , Plant Oils/pharmacology
8.
Atherosclerosis ; 163(1): 69-77, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12048123

ABSTRACT

Hyperlipidaemia may accelerate the development of atherosclerosis by enhancing the expression of chemokines by cells within the arterial wall. Chemokines of the CC subfamily are clearly implicated in atherogenesis; however, recent reports suggest that CXC chemokines may play a hitherto unrecognised role in monocyte recruitment into atheromatous lesions expressing these molecules. Here, we examine whether circulating levels of CXC chemokines may reflect the pathogenic changes occurring during early atherogenesis. ApoE*3 Leiden mice developed marked hypercholesterolaemia, and early Type I 'fatty streak' lesions, following consumption of an atherogenic diet high in saturated fat and cholesterol, and containing sodium cholate, for up to 4 weeks. By contrast, their non-transgenic littermates (C57BL/6J) exhibited a much less pronounced hypercholesterolaemia and did not develop fatty streak lesions, when fed the same diet. Under these conditions, serum concentrations of CXC chemokines, KC and Macrophage Inflammatory Protein-2 (MIP-2) were significantly (P

Subject(s)
Arteriosclerosis/pathology , Chemokines, CXC/blood , Diet, Atherogenic , Hypercholesterolemia/blood , Monokines/blood , RNA, Messenger/analysis , Animals , Aorta/pathology , Apolipoproteins E , Arteriosclerosis/physiopathology , Base Sequence , Chemokine CXCL2 , Chemokines, CXC/metabolism , Disease Models, Animal , Female , Hypercholesterolemia/physiopathology , Immunohistochemistry , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Monokines/metabolism , Polymerase Chain Reaction , Probability , Reference Values , Risk Factors , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...