Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Biofabrication ; 11(1): 015015, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30524110

ABSTRACT

Calcified cartilage regeneration plays an important role in successful osteochondral repair, since it provides a biological and mechanical transition from the unmineralized cartilage at the articulating surface to the underlying mineralized bone. To biomimic native calcified cartilage in engineered constructs, here we test the hypothesis that hydroxyapatite (HAP) stimulates chondrocytes to secrete the characteristic matrix of calcified cartilage. Sodium citrate (SC) was added as a dispersant of HAP within alginate (ALG), and homogeneous dispersal of HAP within ALG hydrogel was confirmed using sedimentation tests, electron microscopy, and energy dispersive spectroscopy. To examine the biological performance of ALG/HAP composites, chondrocyte survival and proliferation, extracellular matrix production, and mineralization potential were evaluated in the presence or absence of the HAP phase. Chondrocytes in ALG/HAP constructs survived well and proliferated, but also expressed higher levels of calcified cartilage markers compared to controls, including Collagen type X secretion, alkaline phosphatase (ALP) activity, and mineral deposition. Compared to controls, ALG/HAP constructs also showed an elevated level of mineralized matrix in vivo when implanted subcutaneously in mice. The printability of ALG/HAP composite hydrogel precursors was verified by 3D printing of ALG/HAP hydrogel scaffolds with a porous structure. In summary, these results confirm the hypothesis that HAP in ALG hydrogel stimulates chondrocytes to secrete calcified matrix in vitro and in vivo and reveal that ALG/HAP composites have the potential for 3D bioprinting and osteochondral regeneration.


Subject(s)
Alginates/chemistry , Bioprinting/instrumentation , Cartilage/cytology , Chondrocytes/cytology , Durapatite/chemistry , Hydrogels/chemistry , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry , Animals , Bioprinting/methods , Calcification, Physiologic , Cartilage/physiology , Cell Proliferation , Cells, Cultured , Chickens , Collagen Type X/chemistry , Extracellular Matrix/chemistry , Printing, Three-Dimensional/instrumentation , Tissue Engineering/methods
2.
Tissue Eng Part C Methods ; 23(3): 156-168, 2017 03.
Article in English | MEDLINE | ID: mdl-28106517

ABSTRACT

Three-dimensional (3D)-printed constructs made of polycaprolactone and chondrocyte-impregnated alginate hydrogel (hybrid cartilage constructs) can mimic the biphasic nature of articular cartilage, thus offering promise for cartilage tissue engineering applications. Notably, the regulatory pathway for medical device development requires validation of such constructs through in vitro bench tests and in vivo preclinical examinations for premarket approval. For this, noninvasive imaging techniques are required for effective evaluation of the progress of these cartilage constructs, especially when implanted in animal models or human subjects. However, characterization of the individual components of the hybrid cartilage constructs and their associated time-dependent structural changes by currently available noninvasive techniques is challenging as these constructs contain a combination of hydrophobic and hydrophilic biomaterials with different refractive indices. In this study, we report the use of a novel synchrotron radiation inline phase contrast imaging computed tomography (SR-inline-PCI-CT) approach for noninvasive (in situ) characterization of 3D-printed hybrid cartilage constructs that has been implanted subcutaneously in mice over a 21-day period. In parallel, traditional invasive assays were used to evaluate the in vivo performance of the implanted hybrid cartilage constructs with respect to their cell viability and secretion of cartilage-specific extracellular matrix over the 21-day period postimplantation in mice. SR-inline-PCI-CT allowed striking visualization of the individual components within the 3D-printed hybrid cartilage constructs, as well as characterization of the time-dependent structural changes after implantation. In addition, the relationship between the implanted constructs and the surrounding tissues was delineated. Furthermore, traditional assays showed that cell viability within the cartilage constructs was at least 70% at all three time points, and secretion of alcian blue- and collagen type 2-positive matrices increased progressively over the 21-day period postimplantation. Overall, these results demonstrate that the 3D-printed hybrid cartilage constructs have good in vivo performance and validate their potential for regeneration of articular cartilage in vivo. In addition, SR-inline-PCI-CT has demonstrated potential for longitudinal and noninvasive monitoring of the functionality of 3D-printed hybrid cartilage constructs in a way that is translatable to other soft tissue engineering applications.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Printing, Three-Dimensional/instrumentation , Regeneration/physiology , Synchrotrons/instrumentation , Tissue Engineering/methods , Animals , Biocompatible Materials , Bioprinting , Cell Survival , Cells, Cultured , Extracellular Matrix , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Male , Mice , Mice, Nude , Tissue Scaffolds
3.
Clin Sci (Lond) ; 131(4): 309-325, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28007970

ABSTRACT

Increased production of methylglyoxal (MG) in vascular tissues is one of the causative factors for vascular remodelling in different subtypes of metabolic syndrome, including hypertension and insulin resistance. Fructose-induced up-regulation of aldolase B (AldoB) contributes to increased vascular MG production but the underlying mechanisms are unclear. Serum levels of MG and fructose were determined in diabetic patients with hypertension. MG level had significant positive correlations with blood pressure and fructose level respectively. C57BL/6 mice were fed with control or fructose-enriched diet for 3 months and ultrasonographic and histologic analyses were performed to evaluate arterial structural changes. Fructose-fed mice exhibited hypertension and high levels of serum MG with normal glucose level. Fructose intake increased blood vessel wall thickness and vascular smooth muscle cell (VSMC) proliferation. Western blotting and real-time PCR analysis revealed that AldoB level was significantly increased in both the aorta of fructose-fed mice and the fructose-treated VSMCs, whereas aldolase A (AldoA) expression was not changed. The knockdown of AldoB expression prevented fructose-induced MG overproduction and VSMC proliferation. Moreover, fructose significantly increased carbohydrate-responsive element-binding protein (ChREBP), phosphorylated FoxO1/3α and Akt1 levels. Fructose induced translocation of ChREBP from the cytosol to nucleus and activated AldoB gene expression, which was inhibited by the knockdown of ChREBP. Meanwhile, fructose caused FoxO1/3α shuttling from the nucleus to cytosol and inhibited its binding to AldoB promoter region. Fructose-induced AldoB up-regulation was suppressed by Akt1 inhibitor but enhanced by FoxO1/3α siRNA. Collectively, fructose activates ChREBP and inactivates FoxO1/3α pathways to up-regulate AldoB expression and MG production, leading to vascular remodelling.


Subject(s)
Forkhead Box Protein O1/metabolism , Fructose-Bisphosphate Aldolase/biosynthesis , Fructose/pharmacology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Vascular Remodeling/drug effects , Adult , Aged , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Female , Fructose/blood , Fructose-Bisphosphate Aldolase/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Humans , Hypertension/blood , Hypertension/etiology , Male , Mice, Inbred C57BL , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Pyruvaldehyde/blood , Up-Regulation/drug effects , Vascular Remodeling/physiology
4.
J Synchrotron Radiat ; 23(Pt 3): 802-12, 2016 05.
Article in English | MEDLINE | ID: mdl-27140161

ABSTRACT

Synchrotron radiation inline phase-contrast imaging combined with computed tomography (SR-inline-PCI-CT) offers great potential for non-invasive characterization and three-dimensional visualization of fine features in weakly absorbing materials and tissues. For cartilage tissue engineering, the biomaterials and any associated cartilage extracellular matrix (ECM) that is secreted over time are difficult to image using conventional absorption-based imaging techniques. For example, three-dimensional printed polycaprolactone (PCL)/alginate/cell hybrid constructs have low, but different, refractive indices and thicknesses. This paper presents a study on the optimization and utilization of inline-PCI-CT for visualizing the components of three-dimensional printed PCL/alginate/cell hybrid constructs for cartilage tissue engineering. First, histological analysis using Alcian blue staining and immunofluorescent staining assessed the secretion of sulfated glycosaminoglycan (GAGs) and collagen type II (Col2) in the cell-laden hybrid constructs over time. Second, optimization of inline PCI-CT was performed by investigating three sample-to-detector distances (SDD): 0.25, 1 and 3 m. Then, the optimal SDD was utilized to visualize structural changes in the constructs over a 42-day culture period. The results showed that there was progressive secretion of cartilage-specific ECM by ATDC5 cells in the hybrid constructs over time. An SDD of 3 m provided edge-enhancement fringes that enabled simultaneous visualization of all components of hybrid constructs in aqueous solution. Structural changes that might reflect formation of ECM also were evident in SR-inline-PCI-CT images. Summarily, SR-inline-PCI-CT images captured at the optimized SDD enables visualization of the different components in hybrid cartilage constructs over a 42-day culture period.

5.
Tissue Eng Part C Methods ; 22(3): 173-88, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26592915

ABSTRACT

Three-dimensional (3D) bioprinting of hybrid constructs is a promising biofabrication method for cartilage tissue engineering because a synthetic polymer framework and cell-impregnated hydrogel provide structural and biological features of cartilage, respectively. During bioprinting, impregnated cells may be subjected to high temperatures (caused by the adjacent melted polymer) and process-induced mechanical forces, potentially compromising cell function. This study addresses these biofabrication issues, evaluating the heat distribution of printed polycaprolactone (PCL) strands and the rheological property and structural stability of alginate hydrogels at various temperatures and concentrations. The biocompatibility of parameters from these studies was tested by culturing 3D hybrid constructs bioprinted with primary cells from embryonic chick cartilage. During initial two-dimensional culture expansion of these primary cells, two morphologically and molecularly distinct cell populations ("rounded" and "fibroblastic") were isolated. The biological performance of each population was evaluated in 3D hybrid constructs separately. The cell viability, proliferation, and cartilage differentiation were observed at high levels in hybrid constructs of both cell populations, confirming the validity of these 3D bioprinting parameters for effective cartilage tissue engineering. Statistically significant performance variations were observed, however, between the rounded and fibroblastic cell populations. Molecular and morphological data support the notion that such performance differences may be attributed to the relative differentiation state of rounded versus fibroblastic cells (i.e., differentiated chondrocytes vs. chondroprogenitors, respectively), which is a relevant issue for cell-based tissue engineering strategies. Taken together, our study demonstrates that bioprinting 3D hybrid constructs of PCL and cell-impregnated alginate hydrogel is a promising approach for cartilage tissue engineering.


Subject(s)
Cartilage/physiology , Chondrocytes/cytology , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cartilage/cytology , Cell Proliferation/drug effects , Cell Separation , Cell Shape/drug effects , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Chondrocytes/drug effects , Collagen Type II/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fluorescent Antibody Technique , Humans , Imaging, Three-Dimensional , Polyesters/pharmacology , Temperature , Viscosity
6.
PLoS One ; 7(5): e36610, 2012.
Article in English | MEDLINE | ID: mdl-22606274

ABSTRACT

Methylglyoxal (MG) is a highly reactive metabolite physiologically presented in all biological systems. The effects of MG on diabetes and hypertension have been long recognized. In the present study, we investigated the potential role of MG in obesity, one of the most important factors to cause metabolic syndrome. An increased MG accumulation was observed in the adipose tissue of obese Zucker rats. Cell proliferation assay showed that 5-20 µM of MG stimulated the proliferation of 3T3-L1 cells. Further study suggested that accumulated-MG stimulated the phosphorylation of Akt1 and its targets including p21 and p27. The activated Akt1 then increased the activity of CDK2 and accelerated the cell cycle progression of 3T3-L1 cells. The effects of MG were efficiently reversed by advanced glycation end product (AGE) breaker alagebrium and Akt inhibitor SH-6. In summary, our study revealed a previously unrecognized effect of MG in stimulating adipogenesis by up-regulation of Akt signaling pathway and this mechanism might offer a new approach to explain the development of obesity.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyruvaldehyde/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Glutathione/metabolism , Lactoylglutathione Lyase/metabolism , Lipid Metabolism/drug effects , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Mice , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Phosphorylation , Pyruvaldehyde/metabolism , Rats , Rats, Zucker
7.
Expert Rev Clin Pharmacol ; 4(1): 63-73, 2011 Jan.
Article in English | MEDLINE | ID: mdl-22115349

ABSTRACT

The metabolic syndrome is a group of abnormalities including obesity, high blood pressure, hyperinsulinemia, high blood glucose levels and hyperlipidemia that together greatly increase the risk of developing cardiovascular disease and Type 2 diabetes. Hydrogen sulfide (H(2)S) is a vasodilatory gasotransmitter mediator in the cardiovascular system, proposed as an endothelium-derived relaxing factor. A lack of H(2)S and its synthesizing enzyme, cystathionine γ-lyase, in the vasculature causes hypertension, whereas an increase in the pancreas reduces insulin secretion. Thus, research is making inroads to determine whether H(2)S is involved in the pathogenesis of the metabolic syndrome. Several laboratories are synthesizing and testing clinically used drugs that release H(2)S. Some of these compounds are being tested for effectiveness in the metabolic syndrome.


Subject(s)
Hydrogen Sulfide/metabolism , Metabolic Syndrome/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Humans , Hydrogen Sulfide/therapeutic use , Hypertension/drug therapy , Hypertension/metabolism , Metabolic Syndrome/drug therapy , Obesity/drug therapy , Obesity/metabolism
8.
Proc Natl Acad Sci U S A ; 108(33): 13829-34, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21808008

ABSTRACT

The goal of the present studies was to investigate the role of changes in hydrogen sulfide (H(2)S) homeostasis in the pathogenesis of hyperglycemic endothelial dysfunction. Exposure of bEnd3 microvascular endothelial cells to elevated extracellular glucose (in vitro "hyperglycemia") induced the mitochondrial formation of reactive oxygen species (ROS), which resulted in an increased consumption of endogenous and exogenous H(2)S. Replacement of H(2)S or overexpression of the H(2)S-producing enzyme cystathionine-γ-lyase (CSE) attenuated the hyperglycemia-induced enhancement of ROS formation, attenuated nuclear DNA injury, reduced the activation of the nuclear enzyme poly(ADP-ribose) polymerase, and improved cellular viability. In vitro hyperglycemia resulted in a switch from oxidative phosphorylation to glycolysis, an effect that was partially corrected by H(2)S supplementation. Exposure of isolated vascular rings to high glucose in vitro induced an impairment of endothelium-dependent relaxations, which was prevented by CSE overexpression or H(2)S supplementation. siRNA silencing of CSE exacerbated ROS production in hyperglycemic endothelial cells. Vascular rings from CSE(-/-) mice exhibited an accelerated impairment of endothelium-dependent relaxations in response to in vitro hyperglycemia, compared with wild-type controls. Streptozotocin-induced diabetes in rats resulted in a decrease in the circulating level of H(2)S; replacement of H(2)S protected from the development of endothelial dysfunction ex vivo. In conclusion, endogenously produced H(2)S protects against the development of hyperglycemia-induced endothelial dysfunction. We hypothesize that, in hyperglycemic endothelial cells, mitochondrial ROS production and increased H(2)S catabolism form a positive feed-forward cycle. H(2)S replacement protects against these alterations, resulting in reduced ROS formation, improved endothelial metabolic state, and maintenance of normal endothelial function.


Subject(s)
Endothelium, Vascular/drug effects , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hyperglycemia/drug therapy , Mitochondria/drug effects , Animals , Cell Line , Diabetes Mellitus, Experimental , Endothelial Cells , Glucose/pharmacology , Homeostasis , Hydrogen Sulfide/metabolism , Hyperglycemia/pathology , Mitochondria/metabolism , Protective Agents/therapeutic use , Rats , Reactive Oxygen Species/metabolism
9.
FASEB J ; 25(5): 1746-57, 2011 May.
Article in English | MEDLINE | ID: mdl-21321187

ABSTRACT

Methylglyoxal (MG), a reactive dicarbonyl molecule, can modify protein to form advanced glycation endproducts. Increased MG level has been implicated in proliferative vascular diseases, but the underlying mechanisms are not clear yet. The serine/threonine kinase, Akt, regulates multiple signaling pathways that control cell proliferation. Using mass spectrometric analysis, we have detected the modification of Akt1 by MG at Cys(77). This structural modification increased Akt1 phosphorylation at Ser(473) and Thr(308). Akt1 phosphorylation and activity were also increased by MG treatment (<50 µM) in cultured vascular smooth muscle cells (VSMCs). MG treatment of VSMCs led to increased DNA synthesis (EC(50)=5.8 µM), cell proliferation, phosphorylation of p21 and glycogen synthase kinase-3α/ß (GSK-3α/ß), and increased cyclin-dependent kinase 2 (CDK2) activity. These effects of MG were significantly inhibited by silencing Akt1 or by an Akt inhibitor. Overexpression of Akt1 Cys(77)Ser mutant in HEK-293 cells increased cell proliferation and DNA synthesis, concurrent with an increase in Akt1 activity, which could not be further augmented by MG treatment. It is concluded that MG-induced VSMC proliferation is mediated by the activation of Akt1 via the modification of Akt1 at Cys(77).


Subject(s)
Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvaldehyde/pharmacology , Animals , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , HEK293 Cells , Humans , Male , Mass Spectrometry , Myocytes, Smooth Muscle/cytology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Rats
10.
Can J Physiol Pharmacol ; 88(3): 273-84, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20393592

ABSTRACT

Aging is a multifactorial process that involves changes at the cellular, tissue, organ and the whole body levels resulting in decreased functioning, development of diseases, and ultimately death. Oxidative stress is believed to be a very important factor in causing aging and age-related diseases. Oxidative stress is caused by an imbalance between oxidants such as reactive oxygen species (ROS) and antioxidants. ROS are produced from the mitochondrial electron transport chain and many oxidative reactions. Methylglyoxal (MG) is a highly reactive dicarbonyl metabolite formed during glucose, protein and fatty acid metabolism. MG levels are elevated in hyperglycemia and other conditions. An excess of MG formation can increase ROS production and cause oxidative stress. MG reacts with proteins, DNA and other biomolecules, and is a major precursor of advanced glycation end products (AGEs). AGEs are also associated with the aging process and age-related diseases such as cardiovascular complications of diabetes, neurodegenerative diseases and connective tissue disorders. AGEs also increase oxidative stress. In this review we discuss the potential role of MG in the aging process through increasing oxidative stress besides causing AGEs formation. Specific and effective scavengers and crosslink breakers of MG and AGEs are being developed and can become potential treatments to slow the aging process and prevent many diseases.


Subject(s)
Aging/metabolism , Oxidative Stress/physiology , Pyruvaldehyde/metabolism , Aging/drug effects , Animals , Glycation End Products, Advanced/adverse effects , Glycation End Products, Advanced/biosynthesis , Humans , Oxidative Stress/drug effects , Pyruvaldehyde/adverse effects , Reactive Oxygen Species/adverse effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
11.
Antioxid Redox Signal ; 12(9): 1093-100, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19803740

ABSTRACT

Hydrogen sulfide (H(2)S) is a gasotransmitter with multifaceted physiological functions, including the regulation of glucose metabolism. Methylglyoxal (MG) is an intermediate of glucose metabolism and plays an important role in the pathogenesis of insulin resistance syndromes. In the present study, we investigated the effect of MG on H(2)S synthesis and the interaction between these two endogenous substances. In cultured vascular smooth muscle cells (VSMCs), MG (10, 30, and 50 microM) significantly decreased cellular H(2)S levels in a concentration-dependent manner, while H(2)S donor, NaHS (30, 60, and 90 microM), significantly decreased cellular MG levels. The expression level and activity of H(2)S-producing enzyme, cystathionine gamma-lyase (CSE), were significantly decreased by MG treatment. NaHS (30-90 microM) significantly inhibited MG (10 or 30 microM)-induced ROS production. Cellular levels of GSH, cysteine, and homocysteine were also increased by MG or NaHS treatment. Furthermore, direct reaction of H(2)S with MG in both concentration- and time-dependent manners were observed in in vitro incubations. In conclusion, MG regulates H(2)S level in VSMCs by downregulating CSE protein expression and directly reacting with H(2)S molecule. Interaction of MG with H(2)S may be one of future directions for the studies on glucose metabolism and the development of insulin resistance syndromes.


Subject(s)
Hydrogen Sulfide/chemistry , Muscle, Smooth, Vascular/cytology , Pyruvaldehyde/chemistry , Animals , Cell Line , Cysteine/chemistry , Dose-Response Relationship, Drug , Glutathione/metabolism , Homocysteine/chemistry , Hydrogen Sulfide/metabolism , Lyases/metabolism , Myocytes, Smooth Muscle/cytology , Pyruvaldehyde/metabolism , Rats , Reactive Oxygen Species , Time Factors
12.
Am J Physiol Heart Circ Physiol ; 295(3): H999-H1007, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18621849

ABSTRACT

Hemin, an oxidized form of heme, is an essential regulator of gene expression and cell cycle progression. Our laboratory previously reported (34) that chronic hemin treatment of spontaneously hypertensive rats reversed the eutrophic inward remodeling of small peripheral arteries. Whether long-term treatment of cultured vascular smooth muscle cells (VSMCs) with hemin alters the proliferation status of these cells has been unknown. In the present study, hemin treatment at 5 muM for 4, 7, 14, and 21 days significantly inhibited the proliferation of cultured rat aortic VSMCs (A-10 cells) by arresting cells at G0/G1 phases so as to decelerate cell cycle progression. Heme oxygenase (HO) activity and inducible HO-1 protein expression were significantly increased by hemin treatment. HO inhibitor tin protoporphyrin IX (SnPP) abolished the effects of hemin on cell proliferation and HO activity. Interestingly, hemin-induced HO-1 expression was further increased in the presence of SnPP. Hemin treatment had no significant effect on the expression of constitutive HO-2. Expression of p21 protein and the level of reactive oxygen species (ROS) were decreased by hemin treatment, which was reversed by application of SnPP. After removal of hemin from culture medium, inhibited cell proliferation and increased HO-1 expression in VSMCs were returned to control level within 1 wk. Transfection with HO-1 small interfering RNA significantly knocked down HO-1 expression and decreased HO activity, but had no effect on HO-2 expression, in cells treated with or without hemin for 7 days. The inhibitory effect of hemin on cell proliferation was abolished in HO-1 silenced cells. It is concluded that induction of HO-1 and, consequently, increased HO activity are responsible for the chronic inhibitory effect of hemin on VSMC proliferation. Changes in the levels of p21 and ROS might also participate in the cellular effects of hemin.


Subject(s)
Hemin/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Flow Cytometry , Heme Oxygenase-1/biosynthesis , Oncogene Protein p21(ras)/biosynthesis , RNA, Small Interfering/pharmacology , Rats , Reactive Oxygen Species/metabolism
13.
J Hypertens ; 26(4): 765-72, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18327087

ABSTRACT

OBJECTIVES: Methylglyoxal is a reactive dicarbonyl intermediate of metabolism produced in the body. It reacts with certain proteins and forms damaging advanced glycation endproducts (AGEs) such as N epsilon-carboxyethyl-lysine (CEL) and N epsilon-carboxymethyl-lysine (CML). Increased methylglyoxal levels are found in diabetes mellitus and associated with hypertension development in the spontaneously hypertensive rats (SHR). The purpose of this study was to investigate whether increased endogenous formation of methylglyoxal and methylglyoxal-induced AGEs caused hypertension development in normotensive Sprague Dawley rats. METHODS: The rats were fed chronically for 16 weeks with fructose, a known precursor of methylglyoxal formation. One group of rats was cotreated with fructose and metformin, an AGEs formation inhibitor. Methylglyoxal and reduced glutathione (GSH) were measured by high performance liquid chromatography, whereas hydrogen peroxide was measured by a dicholorofluorescin assay. Immunohistochemistry was performed for endothelial nitric oxide synthase (eNOS), CEL and CML. RESULTS: Fructose-fed rats had elevated blood pressure, serum methylglyoxal and triglycerides and reduced serum levels of GSH. Methylglyoxal, hydrogen peroxide and CEL were increased in the aorta, whereas eNOS was reduced. CEL and CML were also increased in the mesenteric artery endothelium along with media/lumen ratio, signifying structural remodelling. All the harmful changes in fructose-fed rats were attenuated in metformin and fructose cotreated rats. CONCLUSION: Increased methylglyoxal, AGEs, oxidative stress and reduced eNOS along with structural remodeling of the vessel wall in the aorta and mesenteric artery likely play a role in the pathogenesis of hypertension.


Subject(s)
Fructose/pharmacology , Hypertension/etiology , Hypertension/metabolism , Pyruvaldehyde/blood , Animals , Aorta/metabolism , Dietary Carbohydrates/pharmacology , Glutathione/blood , Glycation End Products, Advanced/metabolism , Hydrogen Peroxide/metabolism , Hypertension/drug therapy , Hypoglycemic Agents/pharmacology , Lysine/analogs & derivatives , Lysine/metabolism , Male , Mesenteric Arteries/metabolism , Metformin/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
14.
Am J Hypertens ; 20(6): 629-36, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17531919

ABSTRACT

BACKGROUND: Methylglyoxal (MG), a metabolite of glucose, and MG-induced advanced glycation endproducts (AGEs) are causatively associated with vascular complications of diabetes mellitus. We have previously reported elevated levels of MG and MG-induced AGEs in spontaneously hypertensive rats (SHR). The purpose of this study was to investigate the causative role of MG and MG-induced AGEs in the pathogenesis of hypertension in SHR. METHODS: Young SHR were treated with an AGE inhibitor, aminoguanidine, for 9 weeks. HPLC was used to determine plasma and aortic MG and reduced glutathione levels. The MG-induced AGEs, N epsilon-carboxyethyl-lysine (CEL) and argpyramidine, in the aorta were determined by immunohistochemistry. Vascular relaxation of small mesenteric arteries was measured using myograph. RESULTS: Chronic treatment with aminoguanidine attenuated age-dependent blood pressure (BP) increase in SHR. Plasma and aortic MG levels, and aortic levels of MG-induced AGEs, were significantly reduced after aminoguanidine treatment, which were comparable to those from age-matched Wistar Kyoto rats. Free radical level was significantly lowered, whereas reduced glutathione level was significantly increased by aminoguanidine treatment in the aortic tissues from SHR. Moreover, aminoguanidine therapy prevented the morphologic damage of vascular tissues in SHR and restored the endothelium-dependent relaxation to acetylcholine. Chronic aminoguanidine treatment also increased aortic endothelial nitric oxide synthase expression and reduced inducible nitric oxide synthase expression. CONCLUSIONS: The MG and MG-induced AGEs contribute to the pathogenesis of hypertension by altering the redox balance, causing vascular eutrophic inward remodeling, and inducing endothelial dysfunction in SHR.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glycation End Products, Advanced/metabolism , Guanidines/therapeutic use , Hypertension/metabolism , Hypertension/prevention & control , Pyruvaldehyde/metabolism , Animals , Aorta/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Endothelium, Vascular/physiopathology , Free Radicals/metabolism , Glutathione/metabolism , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/pathology , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Inbred SHR , Rats, Inbred WKY
15.
Biochem Biophys Res Commun ; 351(2): 485-91, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17069760

ABSTRACT

Hydrogen sulfide (H(2)S) is an important gasotransmitter that generated in mammalian cells from l-cysteine metabolism. Little is known about its protective role in oxidative stress. In the present study, we investigated whether H(2)S could affect homocysteine (HCY)-induced cytotoxicity and oxidative stress in vascular smooth muscle cells. Cultured A-10 cells were exposed to HCY treatment in the presence or absence of NaHS (donor of H(2)S). HCY induced cytotoxicity, increased levels of H(2)O(2), ONOO(-), and O2- in a time- and concentration-dependent manner. Low levels of NaHS (30 or 50microM) protected A-10 cells from cytotoxicity, decreased the production of H(2)O(2), ONOO(-), and O2- in the presence of HCY. Furthermore, NaHS enhanced inhibitory effects of NAC, GSH, DPI, SOD, L-NAME, or vitamin C on oxidized DCF or O2- formation induced by HCY. In conclusion, our findings provide the first evidence that low levels of H(2)S decrease reactive oxygen species and improve cell viability and by doing so limit cellular damage induced by HCY.


Subject(s)
Homocysteine/physiology , Hydrogen Sulfide/metabolism , Muscle, Smooth, Vascular/metabolism , Oxidative Stress , Reactive Nitrogen Species/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Catalase/metabolism , Cell Line , Glutathione/pharmacology , Homocysteine/pharmacology , Hydrogen Sulfide/pharmacology , Muscle, Smooth, Vascular/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Onium Compounds/pharmacology , Rats , Reactive Oxygen Species/metabolism , Sulfides/metabolism , Superoxide Dismutase/metabolism , Superoxides/metabolism
16.
Life Sci ; 79(26): 2448-54, 2006 Nov 25.
Article in English | MEDLINE | ID: mdl-16950408

ABSTRACT

Methylglyoxal (MG), a highly reactive molecule, has been implicated in the development of insulin resistance. We investigated whether fructose, a precursor of MG, induced ONOO(-) generation and whether this process was mediated via endogenously increased MG formation. Fructose significantly increased MG generation in vascular smooth muscle cells (VSMCs) in a concentration and time dependent manner. The intracellular production of MG was increased by 153+/-23% or 259+/-28% after cells were treated 6 h with fructose (15 mM or 30 mM), compared with production from untreated cells (p<0.01, n=4 for each group). A significant increase in the production of ONOO(-), NO, and O(2)(*-), was found in the cells treated with fructose (15 mM) or MG (10 microM). Fructose- or MG-induced ONOO(-) generation was significantly inhibited by MG scavengers, including reduced glutathione or N-acetyl-l-cysteine, and by O(2)(*-) or NO inhibitors, such as diphenylene iodonium, superoxide dismutase or N-nitro-l-arginine methyl ester. Moreover, an enhanced iNOS expression was also observed in the cells treated directly with MG which was significantly inhibited when co-application with N-acetyl-l-cysteine. Our results demonstrated that fructose is capable of inducing a significant increase in ONOO(-) production, which is mediated by an enhanced formation of endogenous MG in VSMCs.


Subject(s)
Fructose/pharmacology , Muscle, Smooth, Vascular/drug effects , Peroxynitrous Acid/biosynthesis , Pyruvaldehyde/metabolism , Acetylcysteine/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glutathione/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Superoxide Dismutase/pharmacology , Superoxides/metabolism , Time Factors
17.
Am J Hypertens ; 19(7): 678-85, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16814121

ABSTRACT

Correlation of blood pressure (BP) with expression levels of large-conductance, voltage- and Ca2+-activated K+ (BK) channel beta1 subunit in vascular tissues from spontaneously hypertensive rats (SHR), Wistar-Kyoto rats (WKY), and Sprague-Dawley rats (SD) at different ages was investigated. Systolic BP and BK beta1 expression in mesenteric arteries at either mRNA or protein levels were not different among 4-week-old SHR, WKY, and SD. With hypertension developed at 7 weeks and reached plateau at 12 weeks, expression levels of BK beta1 mRNA in mesenteric arteries and aortae from SHR during this period of time were significantly higher than in age-matched normotensive WKY. The BK beta1 protein expression was significantly higher in mesenteric arteries from 12-week-old but not 7-week-old SHR when compared with age-matched WKY and SD. The BK beta1 protein levels in aortae were not different among 7-week-old SHR, WKY, and SD but were significantly lower in 12-week-old WKY than in age-matched SHR and SD. Captopril treatment normalized BP of 12-week-old SHR. This treatment downregulated BK beta1 protein in mesenteric arteries but upregulated it in aortae. No significant difference in BK alpha subunit expression was detected in mesenteric arteries from three strains of rats as well as the captopril-treated SHR. It appears that expression patterns of BK beta1 in vascular tissues vary depending on tissue types, animal age, and animal strains. Expression of BK beta1 in mesenteric arteries is closely correlated with BP in SHR. Increased BK beta1 expression in mesenteric arteries may represent a compensatory reaction to limit the development of hypertension.


Subject(s)
Aorta/metabolism , Hypertension/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Mesenteric Arteries/metabolism , Animals , Antihypertensive Agents/pharmacology , Aorta/drug effects , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/analysis , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Mesenteric Arteries/drug effects , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred Strains
18.
Can J Physiol Pharmacol ; 84(12): 1229-38, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17487230

ABSTRACT

Pathogenic mechanisms for essential hypertension are unclear despite striking efforts from numerous research teams over several decades. Increased production of reactive oxygen species (ROS) has been associated with the development of hypertension and the role of ROS in hypertension has been well documented in recent years. In this context, it is important to better understand pathways and triggering factors for increased ROS production in hypertension. This review draws a causative linkage between elevated methylglyoxal level, methylglyoxal-induced production of ROS, and advanced glycation end products in the development of hypertension. It is proposed that elevated methylglyoxal level and resulting protein glycation and ROS production may be the upstream links in the chain reaction leading to the development of hypertension.


Subject(s)
Glycation End Products, Advanced/metabolism , Hypertension/metabolism , Oxidative Stress , Pyruvaldehyde/metabolism , Animals , Glycosylation , Humans , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Signal Transduction
19.
J Hypertens ; 23(8): 1565-73, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16003184

ABSTRACT

OBJECTIVES: The pathogenic process of diabetes mellitus is associated with increased methylglyoxal (MG). MG causes non-enzymic glycation of proteins to form irreversible advanced glycation endproducts (AGEs). However, the correlation between MG and essential hypertension is unknown. The aim of the present study was to investigate whether MG, MG-induced AGEs, and oxidative stress were increased in the aorta of spontaneously hypertensive rats (SHR) and whether an increased formation of MG and related AGEs was correlated with the development of high blood pressure in these rats. METHODS: High-performance liquid chromatography (HPLC) was used to determine MG and reduced glutathione levels in plasma and aorta. MG-induced AGEs, N(epsilon)-carboxyethyl-lysine (CEL) and N(epsilon)-carboxymethyl-lysine (CML), in aorta were determined using immunohistochemistry. Hydrogen peroxide and superoxide levels in aorta and glutathione peroxidase and reductase activities were also determined. RESULTS: Aortic and plasma MG levels were significantly elevated in SHR, but not in Wistar-Kyoto (WKY) rats, at 8, 13 and 20 weeks of age, in parallel with blood pressure increase. Immunohistochemistry revealed more intense staining for CML and CEL in aorta from SHR than those of WKY rats from 8 weeks onwards. Most of the staining was localized to endothelial cells. Superoxide and hydrogen peroxide levels were significantly elevated in aorta of SHR at 13 weeks, whereas reduced glutathione levels, glutathione peroxidase and glutathione reductase activities were significantly decreased compared to WKY rats. CONCLUSIONS: Increased aortic MG, AGE formation and oxidative stress were associated with blood pressure increase in SHR, which may cause endothelial dysfunction and altered vascular reactivity.


Subject(s)
Aorta/metabolism , Glycation End Products, Advanced/metabolism , Hypertension/genetics , Hypertension/metabolism , Pyruvaldehyde/analysis , Age Factors , Animals , Glutathione/analysis , Glutathione Peroxidase/analysis , Glutathione Reductase/analysis , Hydrogen Peroxide/analysis , Immunohistochemistry , Male , Oxidative Stress , Pyruvaldehyde/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Species Specificity , Superoxides/analysis
20.
Free Radic Biol Med ; 38(2): 286-93, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15607912

ABSTRACT

Methylglyoxal (MG) is a metabolite of glucose. Our previous study demonstrated an elevated MG level with an increased oxidative stress in vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats. Whether MG causes the generation of nitric oxide (NO) and superoxide anion (O2*-), leading to peroxynitrite (ONOO-) formation in VSMCs, was investigated in the present study. Cultured rat thoracic aortic SMCs (A-10) were treated with MG or other different agents. Oxidized DCF, reflecting H2O2 and ONOO- production, was significantly increased in a concentration- and time-dependent manner after the treatment of SMCs with MG (3-300 microM) for 45 min-18 h (n = 12). MG-increased oxidized DCF was effectively blocked by reduced glutathione or N-acetyl-l-cysteine, as well as L-NAME (p < 0.05, n = 12). Both O2*- scavenger SOD and NAD(P)H oxidase inhibitor DPI significantly decreased MG-induced oxidized DCF formation. MG significantly and concentration-dependently increased NO and O2*- generation in A-10 cells, which was significantly inhibited by L-NAME and SOD or DPI, respectively. In conclusion, MG induces significant generation of NO and O2*- in rat VSMCs, which in turn causes ONOO- formation. An elevated MG level and the consequential ROS/RNS generation would alter cellular signaling pathways, contributing to the development of different insulin resistance states such as diabetes or hypertension.


Subject(s)
Muscle, Smooth, Vascular/cytology , Nitric Oxide/metabolism , Peroxynitrous Acid/pharmacology , Pyruvaldehyde/pharmacology , Animals , Aorta/cytology , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Free Radicals , Glucose/metabolism , Hydrogen Peroxide/pharmacology , Insulin Resistance , Models, Chemical , Myocytes, Smooth Muscle/cytology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/chemistry , Oxidative Stress , Oxygen/metabolism , Peroxynitrous Acid/chemistry , Peroxynitrous Acid/metabolism , Rats , Reactive Oxygen Species , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...