Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
FASEB J ; 27(9): 3805-17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23752203

ABSTRACT

We examined the effects of a natural secondary bile acid, hyodeoxycholic acid (HDCA), on lipid metabolism and atherosclerosis in LDL receptor-null (LDLRKO) mice. Female LDLRKO mice were maintained on a Western diet for 8 wk and then divided into 2 groups that received chow, or chow + 1.25% HDCA, diets for 15 wk. We observed that mice fed the HDCA diet were leaner and exhibited a 37% (P<0.05) decrease in fasting plasma glucose level. HDCA supplementation significantly decreased atherosclerotic lesion size at the aortic root region, the entire aorta, and the innominate artery by 44% (P<0.0001), 48% (P<0.01), and 94% (P<0.01), respectively, as compared with the chow group. Plasma VLDL/IDL/LDL cholesterol levels were significantly decreased, by 61% (P<0.05), in the HDCA group as compared with the chow diet group. HDCA supplementation decreased intestinal cholesterol absorption by 76% (P<0.0001) as compared with the chow group. Furthermore, HDL isolated from the HDCA group exhibited significantly increased ability to mediate cholesterol efflux ex vivo as compared with HDL of the chow diet group. In addition, HDCA significantly increased the expression of genes involved in cholesterol efflux, such as Abca1, Abcg1, and Apoe, in a macrophage cell line. Thus, HDCA is a candidate for antiatherosclerotic drug therapy.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Deoxycholic Acid/therapeutic use , Lipoproteins, HDL/blood , Receptors, LDL/deficiency , Animals , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Female , Intestinal Absorption/drug effects , Lipoproteins, LDL/blood , Mice , Mice, Knockout , Receptors, LDL/genetics
2.
J Biomed Mater Res A ; 100(12): 3416-21, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22767395

ABSTRACT

Porous silicon nanoparticles (PSiNPs) are attractive carriers for targeted drug delivery in nanomedicine. For in vivo applications, the biodegradation property of PSiNPs provides a pathway for their safe clearance from the body. Particles sizes of 80-120 nm are of particular interest as they are important for cellular applications, such as drug delivery for cancer therapy, because these nanoparticles can take advantage of the enhanced permeability and retention effect to deliver drug preferentially to tumors with leaky vasculature, yet large enough to avoid renal clearance. However, the biodegradability rate of such particles is often too fast, which limits particle half-life and potentially reduces their in vivo delivery efficiency. In this work, we focus on the degradation of nanoscale particles and study the effect of both thermal oxidation and silica coating on the stability of PSiNPs in phosphate buffered saline solution (a close mimic of a basic biological fluid). Using thermal oxidation, the half-life of PSiNPs can be varied from 10 min up to 3 h. Using silica coating, the half-life can be extended further to 8 h. The particles produced using both these techniques can be functionalized using standard silica surface chemistries developed for applications in drug delivery.


Subject(s)
Nanoparticles/chemistry , Silicon/chemistry , Biodegradation, Environmental , Kinetics , Nanoparticles/ultrastructure , Oxidation-Reduction , Porosity , Silicon Dioxide/chemistry , Temperature
3.
J Am Chem Soc ; 133(23): 8798-801, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21595466

ABSTRACT

Porous silicon nanoparticles (PSiNPs) were synthesized by silver-assisted electroless chemical etching of silicon nanowires generated on a silicon wafer. The rod-shaped particles (200-400 nm long and 100-200 nm in diameter) were derivatized with a cyclodextrin-based nanovalve that was closed at the physiological pH of 7.4 but open at pH <6. Release profiles in water and tissue culture media showed that no cargo leaked when the valves were closed and that release occurred immediately after acidification. In vitro studies using human pancreatic carcinoma PANC-1 cells proved that these PSiNPs were endocytosed and carried cargo molecules into the cells and released them in response to lysosomal acidity. These studies show that PSiNPs can serve as an autonomously functioning delivery platform in biological systems and open new possibilities for drug delivery.


Subject(s)
Nanoparticles/chemistry , Silicon/chemistry , Biological Transport , Catalysis , Cell Line, Tumor , Cyclodextrins/chemistry , Humans , Hydrogen-Ion Concentration , Porosity , Silicon/metabolism
4.
J Lipid Res ; 51(7): 1962-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20194110

ABSTRACT

Previous studies have shown that colony stimulating factor-1 (CSF-1) deficiency dramatically reduced atherogenesis in mice. In this report we investigate this mechanism and explore a therapeutic avenue based on inhibition of CSF-1 signaling. Lesions from macrophage colony stimulating factor-1 (Csf1)+/- mice showed increased numbers of apoptotic macrophages, decreased overall macrophage content, and inflammation. In vitro studies indicated that CSF-1 is chemotactic for monocytes. Bone marrow transplantation studies suggested that vascular cell-derived, rather than macrophage-derived, CSF-1 is responsible for the effect on atherosclerosis. Consistent with previous studies, CSF-1 affected lesion development in a dose-dependent manner, suggesting that pharmacological inhibition of CSF-1 might achieve similar results. Indeed, we observed that treatment of hyperlipidemic mice with a CSF-1 receptor kinase inhibitor inhibited plaque progression. This observation was accompanied by a reduction in the expression of adhesion factors (ICAM-1), macrophage markers (F4/80), inflammatory cytokines (Il-6, Il-1beta), and macrophage matrix degradation enzymes (MMP-9). We conclude that the M-CSF pathway contributes to monocyte recruitment and macrophage survival and that this pathway is a potential target for therapeutic intervention.


Subject(s)
Apoptosis/drug effects , Arteries , Atherosclerosis/pathology , Cell Movement/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Monocytes/drug effects , Monocytes/physiology , Animals , Arteries/drug effects , Arteries/metabolism , Arteries/pathology , Atherosclerosis/metabolism , Bone Marrow Transplantation , Dose-Response Relationship, Drug , Female , Gene Expression , Macrophage Colony-Stimulating Factor/genetics , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects
5.
Blood ; 115(7): 1461-71, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-20008303

ABSTRACT

Tumor-infiltrating myeloid cells (TIMs) support tumor growth by promoting angiogenesis and suppressing antitumor immune responses. CSF-1 receptor (CSF1R) signaling is important for the recruitment of CD11b(+)F4/80(+) tumor-associated macrophages (TAMs) and contributes to myeloid cell-mediated angiogenesis. However, the impact of the CSF1R signaling pathway on other TIM subsets, including CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSCs), is unknown. Tumor-infiltrating MDSCs have also been shown to contribute to tumor angiogenesis and have recently been implicated in tumor resistance to antiangiogenic therapy, yet their precise involvement in these processes is not well understood. Here, we use the selective pharmacologic inhibitor of CSF1R signaling, GW2580, to demonstrate that CSF-1 regulates the tumor recruitment of CD11b(+)Gr-1(lo)Ly6C(hi) mononuclear MDSCs. Targeting these TIM subsets inhibits tumor angiogenesis associated with reduced expression of proangiogenic and immunosuppressive genes. Combination therapy using GW2580 with an anti-VEGFR-2 antibody synergistically suppresses tumor growth and severely impairs tumor angiogenesis along with reverting at least one TIM-mediated antiangiogenic compensatory mechanism involving MMP-9. These data highlight the importance of CSF1R signaling in the recruitment and function of distinct TIM subsets, including MDSCs, and validate the benefits of targeting CSF1R signaling in combination with antiangiogenic drugs for the treatment of solid cancers.


Subject(s)
Anisoles/pharmacology , Carcinoma, Lewis Lung/drug therapy , Cell Movement/drug effects , Lung Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Pyrimidines/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Lung Neoplasms/pathology , Macrophages/cytology , Male , Matrix Metalloproteinase 9/metabolism , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/pathology , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rats , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
J Lipid Res ; 50(4): 623-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19029066

ABSTRACT

Secretory phospholipase A2 (sPLA2) activity promotes foam cell formation, increases proinflammatory bioactive lipid levels, decreases HDL levels, increases atherosclerosis in transgenic mice, and is an independent marker of cardiovascular disease. The effects of the sPLA2 inhibitor A-002 (varespladib) and pravastatin as monotherapies and in combination on atherosclerosis, lipids, and paraoxonase (PON) activity in apoE(-/-) mice were investigated. Male apoE(-/-) mice were placed on a 12-week high-fat diet supplemented with A-002 alone or combined with pravastatin. Atherosclerotic lesions were examined for size and composition using en face analysis, Movat staining, anti-CD68, and anti-alpha actin antibodies. Plasma lipids and PON activity were measured. A-002 decreased atherosclerotic lesion area by approximately 75% while increasing fibrous cap size by over 200%. HDL levels increased 40% and plasma PON activity increased 80%. Pravastatin monotherapy had no effect on lesion size but when combined with A-002, decreased lesion area 50% and total cholesterol levels 18% more than A-002 alone. A-002, a sPLA2 inhibitor, acts synergistically with pravastatin to decrease atherosclerosis, possibly through decreased levels of systemic inflammation or decreased lipid levels. A-002 treatment also resulted in a profound increase in plasma PON activity and significantly larger fibrous caps, suggesting the formation of more stable plaque architecture.


Subject(s)
Acetates/administration & dosage , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Enzyme Inhibitors/administration & dosage , Indoles/administration & dosage , Phospholipases A2, Secretory/antagonists & inhibitors , Pravastatin/administration & dosage , Animals , Apolipoproteins E/genetics , Aryldialkylphosphatase/blood , Atherosclerosis/blood , Atherosclerosis/pathology , Drug Synergism , Keto Acids , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prodrugs/administration & dosage
7.
J Immunol ; 181(10): 7176-85, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981139

ABSTRACT

Experimental and clinical studies link Chlamydia pneumoniae infection to atherogenesis and atherothrombotic events, but the underlying mechanisms are unclear. We tested the hypothesis that C. pneumoniae-induced acceleration of atherosclerosis in apolipoprotein E (ApoE)(-/-) mice is reciprocally modulated by activation of TLR-mediated innate immune and liver X receptor alpha (LXRalpha) signaling pathways. We infected ApoE(-/-) mice and ApoE(-/-) mice that also lacked TLR2, TLR4, MyD88, or LXRalpha intranasally with C. pneumoniae followed by feeding of a high fat diet for 4 mo. Mock-infected littermates served as controls. Atherosclerosis was assessed in aortic sinuses and in en face preparation of whole aorta. The numbers of activated dendritic cells (DCs) within plaques and the serum levels of cholesterol and proinflammatory cytokines were also measured. C. pneumoniae infection markedly accelerated atherosclerosis in ApoE-deficient mice that was associated with increased numbers of activated DCs in aortic sinus plaques and higher circulating levels of MCP-1, IL-12p40, IL-6, and TNF-alpha. In contrast, C. pneumoniae infection had only a minimal effect on atherosclerosis, accumulation of activated DCs in the sinus plaques, or circulating cytokine increases in ApoE(-/-) mice that were also deficient in TLR2, TLR4, or MyD88. However, C. pneumoniae-induced acceleration of atherosclerosis in ApoE(-/-) mice was further enhanced in ApoE(-/-)LXRalpha(-/-) double knockout mice and was accompanied by higher serum levels of IL-6 and TNF-alpha. We conclude that C. pneumoniae infection accelerates atherosclerosis in hypercholesterolemic mice predominantly through a TLR/MyD88-dependent mechanism and that LXRalpha appears to reciprocally modulate and reduce the proatherogenic effects of C. pneumoniae infection.


Subject(s)
Atherosclerosis/microbiology , Chlamydia Infections/complications , DNA-Binding Proteins/metabolism , Myeloid Differentiation Factor 88/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Animals , Aorta/immunology , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Chlamydia Infections/metabolism , Chlamydophila pneumoniae , Cytokines/blood , Cytokines/immunology , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fluorescent Antibody Technique , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Hypercholesterolemia/complications , Immunohistochemistry , Liver X Receptors , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Oligonucleotide Array Sequence Analysis , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/genetics
8.
Circ Res ; 100(12): 1703-11, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17495224

ABSTRACT

Heme oxygenase (HO-1) is the rate-limiting enzyme in the catabolism of heme, which leads to the generation of biliverdin, iron, and carbon monoxide. It has been shown to have important antioxidant and antiinflammatory properties that result in a vascular antiatherogenic effect. To determine whether HO-1 expression in macrophages constitutes a significant component of the protective role in atherosclerosis, we evaluated the effect of decreased or absent HO-1 expression in peritoneal macrophages on oxidative stress and inflammation in vitro, and the effect of complete deficiency of HO-1 expression in macrophages in atherosclerotic lesion formation in vivo. We found that compared with HO-1(+/+) controls, peritoneal macrophages from HO-1(-/-) and HO-1(+/-) mice exhibited (1) increased reactive oxygen species (ROS) generation, (2) increased proinflammatory cytokines such as monocyte chemotactic protein 1 (MCP-1) and interleukin 6 (IL-6), and (3) increased foam cell formation when treated with oxLDL, attributable in part to increased expression of scavenger receptor A (SR-A). Bone marrow transplantation experiments performed in lethally irradiated LDL-R null female mice, reconstituted with bone marrow from HO-1(-/-) versus HO-1(+/+) mice, revealed that HO-1(-/-) reconstituted animals exhibited atherosclerotic lesions with a greater macrophage content as evaluated by immunohistochemistry and planimetric assessment. We conclude that HO-1 expression in macrophages constitutes an important component of the antiatherogenic effect by increasing antioxidant protection and decreasing the inflammatory component of atherosclerotic lesions.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Heme Oxygenase-1/metabolism , Macrophages/enzymology , Animals , Antioxidants/metabolism , Atherosclerosis/pathology , Bone Marrow Transplantation/pathology , Chemokine CCL2/metabolism , Foam Cells/pathology , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Interleukin-6/metabolism , Macrophages/pathology , Mice , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Receptors, LDL/genetics , Receptors, LDL/physiology , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/physiology
9.
Arterioscler Thromb Vasc Biol ; 27(3): 621-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17158354

ABSTRACT

OBJECTIVE: Recent evidence suggests that dendritic cells may play an important role in atherosclerosis. Based primarily on previous in vitro studies, we hypothesized that granulocyte macrophage colony-stimulating factor (GM-CSF)-deficient mice would have decreased dendritic cells in lesions. METHODS AND RESULTS: To test this, we characterized gene targeted GM-CSF(-/-) mice crossed to hypercholesterolemic low-density lipoprotein receptor null mice. Our results provide conclusive evidence that GM-CSF is a major regulator of dendritic cell formation in vivo. Aortic lesion sections in GM-CSF(-/-) low-density lipoprotein receptor null animals showed a dramatic 60% decrease in the content of dendritic cells as judged by CD11c staining but no change in the overall content of monocyte-derived cells. The GM-CSF-deficient mice exhibited a significant 20% to 50% decrease in the size of aortic lesions, depending on the location of the lesions. Other prominent changes in GM-CSF(-/-) mice were decreased lesional T cell content, decreased autoantibodies to oxidized lipids, and striking disruptions of the elastin fibers adjacent to the lesion. CONCLUSION: Given that GM-CSF is dramatically induced by oxidized lipids in endothelial cells, our data suggest that GM-CSF serves to regulate dendritic cell formation in lesions and that this, in turn, influences inflammation, plaque growth and possibly plaque stability.


Subject(s)
Atherosclerosis/physiopathology , Dendritic Cells/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Cell Count , Cells, Cultured , Dendritic Cells/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Lipid Peroxidation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reference Values , Sensitivity and Specificity
10.
Ann Med ; 37(3): 173-8, 2005.
Article in English | MEDLINE | ID: mdl-16019715

ABSTRACT

The ability of high-density lipoprotein (HDL) to promote cholesterol efflux is thought to be important in its protection against cardiovascular disease. Anti-inflammatory properties of HDL have emerged as additional properties that may also be important. HDL appears to have evolved as part of the innate immune system functioning to inhibit inflammation in the absence of an acute phase response (APR) but functioning to increase inflammation in the presence of an APR. Inbred strains of mice that are genetically susceptible to atherosclerosis have pro-inflammatory HDL, while inbred strains that are resistant to atherosclerosis have anti-inflammatory HDL. In one small study, humans with coronary heart disease (CHD) or CHD equivalents had pro-inflammatory HDL prior to statin therapy and about half continued to have pro-inflammatory HDL after statin therapy despite a profound decrease in plasma lipids. Pro-inflammatory HDL was relatively weak in its ability to promote cholesterol efflux while anti-inflammatory HDL was better in promoting cholesterol efflux. In other studies, oxidative alterations of the major protein of HDL, apolipoprotein A-I (apoA-I), impaired the ability of the apoA-I to promote cholesterol efflux. Thus, HDL structure and function may be more important than HDL-cholesterol levels in predicting risk for cardiovascular disease.


Subject(s)
Cholesterol, HDL/physiology , Coronary Disease/physiopathology , Inflammation/prevention & control , Inflammation/physiopathology , Animals , Apolipoprotein A-I/blood , Coronary Disease/blood , Humans
11.
Circ Res ; 91(2): 120-6, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-12142344

ABSTRACT

We previously reported the identification of a locus on mouse chromosome 6 that confers almost total resistance to atherogenesis, even on a hypercholesterolemic (LDL receptor-null) background. 5-Lipoxygenase (5-LO) is the rate-limiting enzyme in leukotriene synthesis and was among the chromosome 6 locus candidate genes that we examined. The levels of 5-LO mRNA were reduced about 5-fold in a congenic strain, designated CON6, containing the resistant chromosome 6 region derived from the CAST/Ei strain (CAST), as compared with the background C57BL/6J (B6) strain. 5-LO protein levels were similarly reduced in the CON6 mice. Sequencing of the 5-LO cDNA revealed several differences between CON6 and the B6 strain. To test the whether 5-LO is responsible for the resistant phenotype, we bred a 5-LO knockout allele onto an LDL receptor-null (LDLR(-/-)) background. On this background, the mice bred poorly and only heterozygous 5-LO knockout mice were obtained. These mice showed a dramatic decrease (>26-fold; P<0.0005) in aortic lesion development, similar to the CON6 mice. Immunohistochemistry revealed that 5-LO was abundantly expressed in atherosclerotic lesions of apoE(-/-) and LDLR(-/-) deficient mice, appearing to colocalize with a subset of macrophages but not with all macrophage-staining regions. When bone marrow from 5-LO(+/-) mice was transplanted into LDLR(-/-), there was a significant reduction in atherogenesis, suggesting that macrophage 5-LO is responsible, at least in part, for the effect on atherosclerosis. These results indicate that 5-LO contributes importantly to the atherogenic process and they provide strong presumptive evidence that reduced 5-LO expression is partly responsible for the resistance to atherosclerosis in CON6 mice.


Subject(s)
Arachidonate 5-Lipoxygenase/genetics , Arteriosclerosis/genetics , Alleles , Amino Acid Substitution , Animals , Arachidonate 5-Lipoxygenase/physiology , Arteriosclerosis/enzymology , Arteriosclerosis/pathology , Bone Marrow Transplantation , Genetic Predisposition to Disease , Insulin/blood , Macrophages/enzymology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , Receptors, LDL/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...