Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Stem Cells ; 42(7): 593-606, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38655770

ABSTRACT

Cycling myeloid cells (CMCs) are often detected from various tissues using single-cell RNA sequencing (scRNA-seq) datasets, however, their research value was not noticed before. For the first time, our study preliminarily revealed the origin, differentiation, and roles of CMCs in physiological processes. Particularly, subgroup a of cycling myeloid cells (aCMCs) were conclusively identified as belonging to a specific cell type. In an active state, aCMCs rapidly proliferate during the early stages of an embryonic development. With an individual maturing, most aCMCs differentiate into specialized cells, while a small portion of them enter an inactive or dormant state. Under pathological conditions, aCMCs restore their proliferative and differentiation capacities via activation or revival. The present study has set the stage for future research on CMCs by linking them with progenitors of immune cells, and provided a crucial starting point to understand the origin, differentiation, and roles of CMCs in various physiological and pathological processes, particularly those related to traumatic injury, cancer, and pathogen infection, leading to develop targeted therapies or interventions.


Subject(s)
Cell Differentiation , Myeloid Cells , Single-Cell Analysis , Myeloid Cells/metabolism , Single-Cell Analysis/methods , Animals , Cell Differentiation/genetics , RNA-Seq/methods , Humans , Mice , Sequence Analysis, RNA/methods , Cell Cycle/genetics , Cell Proliferation/genetics , Single-Cell Gene Expression Analysis
2.
Front Surg ; 9: 881006, 2022.
Article in English | MEDLINE | ID: mdl-35548186

ABSTRACT

Purpose: This study aimed to observe the application value of dezocine and ketorolac tromethamine in patient-controlled intravenous analgesia (PCIA) of patients undergoing laparoscopic cholecystectomy (LC). Methods: A total of 154 patients who underwent LC surgery in our hospital and received PCIA after surgery from September 2020 to September 2021 were selected, they were divided into group A (n = 77) and group B (n = 77). Group A was given dezocine and group B was given ketorolac tromethamine. The analgesia, sedation, comfort, and adverse reactions of the two groups were closely observed at 4, 8, 12, and 24 h after surgery. Results: At 4, 8, 12, and 24 h after surgery, the visual analog scale scores in group B were lower than those in group A (P < 0.05). At 4, 8, 12, and 24 h after surgery, the Ramsay scores in group B were higher than those in group A (P < 0.05). At 4, 8, 12, and 24 h after surgery, there was no significant difference in Bruggrmann comfort scale scores between the two groups (P > 0.05). There was no significant difference in the incidence of adverse reactions between the two groups (P > 0.05). Conclusion: Both dezocine and ketorolac tromethamine have high clinical application value in patients who underwent LC surgery and received PCIA, with higher patient comfort and fewer adverse reactions. But compared with dezocine, ketorolac tromethamine can achieve better sedative and analgesic effects, which is worthy of clinical promotion.

3.
Mol Med Rep ; 16(5): 7641-7648, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28944865

ABSTRACT

Prenatal examinations, including serological screening and ultrasound screening, are the methods determining a risk of fetal chromosomal disease. The current study is aimed to assess whether ultrasound screening can effectively assist the screening for fetal chromosomal disease among pregnant women with a single abnormal serum marker. Following serologic screening, pregnant women at 18­32 weeks underwent systematic fetal ultrasound analysis. In this study, 99 pregnant women with an abnormal serum marker and fetal ultrasound abnormalities underwent prenatal diagnosis of amniotic fluid or umbilical cord blood, with confirmation by pathological examination performed following birth or induced labor. A total of 95 cases with an abnormal serum marker but no fetal ultrasound abnormalities were used as the control group, and underwent prenatal karyotype analysis. The rate of fetal chromosomal abnormalities of women with ultrasound abnormalities was significantly higher than in the control group. The fetal chromosomal abnormalities rate in pregnant women with a history of abnormal gestation/birth was higher than in pregnant women with a normal history. The present results suggest that ultrasound examination can facilitate screening for fetal chromosome abnormalities in pregnant women with a single abnormal serum marker.


Subject(s)
Aneuploidy , Down Syndrome/diagnostic imaging , Prenatal Diagnosis/methods , Trisomy , Ultrasonography, Prenatal/methods , Adult , Amniotic Fluid/chemistry , Biomarkers/blood , Case-Control Studies , Chorionic Gonadotropin/blood , Down Syndrome/genetics , Female , Fetal Blood/chemistry , Fetus , Gestational Age , Humans , Karyotyping , Pregnancy , Pregnancy Trimester, Second , alpha-Fetoproteins/metabolism
4.
Acta Diabetol ; 49(4): 243-54, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21947383

ABSTRACT

Cardiovascular disease is the predominant cause of morbidity in people with type 2 diabetes. Hypertension frequently coexists with diabetes and substantially increases the risk of developing end-organ damage. Controlling hypertension in patients with diabetes is therefore critical to reducing microvascular and macrovascular complications. Agents that block the renin-angiotensin system are increasingly used in patients with diabetes based on their cardiovascular and renoprotective effects, in addition to their direct effects on reducing blood pressure. Telmisartan, an angiotensin II receptor blocker (ARB), has a number of distinguishing pharmacological properties such as having the longest half-life and highest lipophilicity in its class. The ONgoing Telmisartan Alone and in combination with Ramipril Global Endpoint Trial (ONTARGET(®)) trial showed that telmisartan reduces cardiovascular morbidity (including myocardial infarction and stroke) in subjects with a broad spectrum of cardiovascular risk factors, including type 2 diabetes. Telmisartan is the only ARB indicated for the reduction of cardiovascular morbidity in patients with diabetes and end-organ damage, as well as in patients without diabetes but with a history of coronary artery disease, peripheral artery disease, or previous stroke. Trials of telmisartan in patients with diabetes and varying degrees of nephropathy also suggest that this drug can slow the progression of renal disease, an effect that appears to be at least partly independent of reduction in blood pressure. Telmisartan is therefore an important therapeutic option for optimizing cardiovascular and renal protection in the type 2 diabetic population.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Benzimidazoles/therapeutic use , Benzoates/therapeutic use , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/prevention & control , Renin-Angiotensin System/physiology , Cardiovascular Diseases/etiology , Humans , Hypertension/drug therapy , Renin-Angiotensin System/drug effects , Risk Factors , Telmisartan
5.
Kidney Int ; 79(2): 199-209, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20962744

ABSTRACT

Cell division autoantigen 1 (CDA1) modulates cell proliferation and transforming growth factor-ß (TGF-ß) signaling in a number of cellular systems; here we found that its levels were elevated in the kidneys of two animal models of diabetic renal disease. The localization of CDA1 to tubular cells and podocytes in human kidney sections was similar to that seen in the rodent models. CDA1 small interfering RNA knockdown markedly attenuated, whereas its overexpression increased TGF-ß signaling, modulating the expression of TGF-ß, TGF-ß receptors, connective tissue growth factor, collagen types I, III, IV, and fibronectin genes in HK-2 cells. CDA1 and TGF-ß together were synergistic in stimulating TGF-ß signaling and target gene expression. CDA1 knockdown effectively blocked TGF-ß-stimulated expression of collagen genes. This was due to its ability to modulate the TGF-ß type I, but not the type II, receptor, leading to increased phosphorylation of Smad3 and extracellular signal-regulated kinase mitogen-activated protein kinase. Furthermore, the Smad3 inhibitor, SIS3, markedly attenuated the activities of CDA1 in stimulating TGF-ß signaling as well as gene expression of collagens I, III, and IV. Thus, our in vitro and in vivo findings show that CDA1 has a critical role in TGF-ß signaling in the kidney.


Subject(s)
Autoantigens/physiology , Diabetic Nephropathies/etiology , Transforming Growth Factor beta/physiology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Autoantigens/genetics , Base Sequence , Cell Line , DNA Primers/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Fibrosis , Gene Knockdown Techniques , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Inbred SHR , Signal Transduction
6.
J Diabetes Investig ; 2(4): 243-7, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-24843491

ABSTRACT

As the increasing prevalence of diabetes reaches epidemic proportions worldwide, diabetic nephropathy and associated end-stage renal failure will be an unavoidable major health burden to not only individuals with diabetes and their families, but also to the health systems both in developed and developing countries. Over the past decade, a large body of research has focused on diabetic nephropathy ranging from studies in molecular signaling, hemodynamic regulation and pharmaceutical intervention to clinical outcomes. It is likely that the pathophysiology of diabetic nephropathy involves a multifactorial interaction between metabolic and hemodynamic factors. Metabolic factors involve glucose-dependent pathways, such as advanced glycation end-products and their receptors. Hemodynamic factors include various vasoactive hormones, such as components of the renin-angiotensin system. It is likely that these metabolic and hemodynamic factors interact through shared molecular and signaling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells and protein kinase C with associated reactive oxygen species generation. It is likely that these contributing factors cause pathological damage not only to the glomerulus, in particular podocytes, but also to the tubulointerstitium. Specific inhibitors of the various pathways are now available and these emerging pharmaceutical interventions might have potential implications for the prevention and treatment of diabetic nephropathy. The mainstay of therapy remains the achievement of optimal glycemic and blood pressure control in order to slow the progression of diabetic nephropathy. Agents that interrupt the renin-angiotensin system have been shown to be particularly useful as renoprotective agents in both hypertensive and normotensive type 1 and type 2 diabetic subjects. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2011.00131.x, 2011).

7.
Genes (Basel) ; 1(3): 335-48, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-24710090

ABSTRACT

Cell Division Autoantigen 1 (CDA1) was discovered following screening a human expression library with serum from a patient with Discoid Lupus Erythematosus. CDA1, encoded by TSPYL2 on the X chromosome, shares anti-proliferative and pro­fibrotic properties with TGF-b. It inhibits cell growth through p53, pERK1/2 and p21­mediated pathways and is implicated in tumorigenesis and the DNA damage response. Its pro-fibrotic property is mediated through cross-talk with TGF-b that results in upregulation of extracellular matrix proteins. The latter properties have identified a key role for CDA1 in diabetes associated atherosclerosis. These dual properties place CDA1 as an attractive molecular target for treating tumors and vascular fibrosis including atherosclerosis and other vascular disorders associated with enhanced TGF-ß action and tissue scarring.

8.
J Biol Chem ; 282(16): 11722-31, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17317670

ABSTRACT

We previously reported that overexpression of cell division autoantigen 1 (CDA1) in HeLa cells arrests cell growth and inhibits DNA synthesis at S-phase. Here we show that CDA1-induced arrest of cell growth is accompanied by increases in protein and mRNA levels of the cyclin-dependent kinase (Cdk) inhibitor protein, p21(Waf1/Cip1) (p21). Both p21 induction and cell growth arrest are reversed when CDA1 expression is inhibited. CDA1 also increases p53 protein, but not its mRNA, in a time- and dose-dependent manner. MDM2, a ubiquitin ligase regulating p53 degradation, is inactivated by CDA1, suggesting that p53 protein accumulation is due to decreased protein degradation. Knockdown of p53, using siRNA targeting two sites of p53 mRNA, abrogates transcriptional induction of p21 by CDA1. Deletion of the p53 responsive element in the distal region of p21 promoter attenuates promoter activity in response to CDA1. DNA damage caused by camptothecin treatment increases mRNA and protein levels of CDA1, accompanied by induction of p53. The DNA damage-induced p53 induction is markedly attenuated by CDA1 knockdown. CDA1 induces phosphorylation of ERK1/2(p44/42), an activity blocked by PD98059 and U0126, inhibitors of the upstream kinase MEK1/2. The MEK inhibitors also block induction of p21 mRNA and abrogate p21 promoter activity stimulated by CDA1. Cell cycle kinases, Cdk1, -2, -4, and -6 are inhibited by CDA1 overexpression. We conclude that CDA1 induces p53- and MEK/ERK1/2 MAPK-dependent expression of p21 by acting through the p53 responsive element in the p21 promoter and that this contributes to its antiproliferative activity.


Subject(s)
Autoantigens/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Autoantigens/chemistry , Cell Proliferation , HeLa Cells , Humans , Luciferases/metabolism , MAP Kinase Signaling System , Signal Transduction , Transcriptional Activation
9.
J Am Soc Nephrol ; 16(10): 2976-84, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16107577

ABSTRACT

Although hemodynamic and metabolic factors are individually implicated in the development of diabetic nephropathy, their interaction has not been defined clearly. In this study, the effects of angiotensin II (Ang II) and advanced glycation end products (AGE) both individually on each other are explored and compared. In the first study arm, Sprague-Dawley rats received a continuous infusion of AGE-modified rat serum albumin (RSA) or unmodified RSA for 4 wk with or without the angiotensin receptor type 1 antagonist valsartan. In the second arm, animals received a continuous infusion of Ang II (58.3 ng/kg per min) with or without the AGE inhibitor pyridoxamine. Components of the intrarenal renin-angiotensin system were measured using real time reverse transcription-PCR, immunohistochemistry, and standard angiotensin-converting enzyme (ACE) activity assays. Renal and serum AGE were quantified by immunohistochemistry, ELISA, and AGE-fluorescence. After an infusion of AGE-RSA, renal expression of angiotensinogen, ACE, renin, and angiotensin receptor type 1 were increased significantly (all P < 0.01), and ACE activity was elevated. This was associated with tubular and glomerular hypertrophy and AGE accumulation, which could be antagonized by valsartan. However, valsartan had no effect on increased filtration fraction associated with an AGE-RSA infusion. At the same time, an infusion of Ang II increased the serum and renal accumulation of AGE and advanced oxidation protein products and induced renal hypertrophy and salt retention that could be antagonized by pyridoxamine. However, pyridoxamine had no effect on renal vasoconstriction manifested by reduced renal blood flow. AGE and Ang II have overlapping activities in the kidney. The beneficial effects of blockade of either pathway underline the importance of this interaction in diabetic renal disease and the aging kidney.


Subject(s)
Glycation End Products, Advanced/biosynthesis , Kidney/metabolism , Renin-Angiotensin System/physiology , Animals , Hypertrophy , Kidney/pathology , Male , Rats , Rats, Sprague-Dawley
10.
J Histochem Cytochem ; 53(10): 1245-56, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15956033

ABSTRACT

The temporal and spatial expression of transforming growth factor (TGF)-beta(1) and connective tissue growth factor (CTGF) was assessed in the left ventricle of a myocardial infarction (MI) model of injury with and without angiotensin-converting enzyme (ACE) inhibition. Coronary artery ligated rats were killed 1, 3, 7, 28, and 180 days after MI. TGF-beta(1), CTGF, and procollagen alpha1(I) mRNA were localized by in situ hybridization, and TGF-beta(1) and CTGF protein levels by immunohistochemistry. Collagen protein was measured using picrosirius red staining. In a separate group, rats were treated for 6 months with an ACE inhibitor. There were temporal and regional differences in the expression of TGF-beta(1), CTGF, and collagen after MI. Procollagen alpha1(I) mRNA expression increased in the border zone and scar peaking 1 week after MI, whereas collagen protein increased in all areas of the heart over the 180 days. Expression of TGF-beta(1) mRNA and protein showed major increases in the border zone and scar peaking 1 week after MI. The major increases in CTGF mRNA and protein occurred in the viable myocardium at 180 days after MI. Long-term ACE inhibition reduced left ventricular mass and decreased fibrosis in the viable myocardium, but had no effect on cardiac TGF-beta(1) or CTGF. TGF-beta(1) is involved in the initial, acute phase of inflammation and repair after MI, whereas CTGF is involved in the ongoing fibrosis of the heart. The antifibrotic benefits of captopril are not mediated through a reduction in CTGF.


Subject(s)
Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Captopril/pharmacology , Collagen Type I/biosynthesis , Connective Tissue Growth Factor , Fibrosis , Immunohistochemistry , In Situ Hybridization , Myocytes, Cardiac/metabolism , RNA, Messenger/biosynthesis , Rats , Time Factors , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta1 , Ventricular Remodeling
11.
J Hypertens ; 23(1): 153-64, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15643138

ABSTRACT

OBJECTIVE: It has been postulated that vascular endothelial growth factor (VEGF) plays a role in the progression of renal injury. However, the role of other angiogenic factors and their receptors, such as the angiopoietins and Tie2, and in particular their relation to renoprotective therapies, such as agents that interrupt the renin-angiotensin system, have not been studied in the context of diabetes-related renal injury. DESIGN AND METHODS: Renal expression of VEGF, angiopoietin-1 (Ang-1), angiopoietin-2 (Ang-2) and their receptors, VEGF-R2 and Tie-2, were assessed using reverse transcription-polymerase chain reaction, immunohistochemistry and Western blotting, in control and streptozotocin diabetic rats, untreated or receiving the AT1 receptor antagonist, valsartan, or the AT2 receptor antagonist, PD123319. RESULTS: Diabetes was associated with increased gene and protein expression of VEGF, VEGF-R2, Ang-1, Ang-2 and Tie-2. AT1 receptor antagonism attenuated gene expression of these cytokines and receptors, yet PD123319, which had no effect on blood pressure, reduced VEGF-R2 and Ang-1 gene expression and decreased VEGF, Ang-1 and Ang-2 protein levels. CONCLUSIONS: In experimental diabetes, there is significant upregulation within the kidney of various angiogenic cytokines and their receptors. Furthermore, the effects of angiotensin II receptor blockade on these parameters is consistent with the VEGF-VEGF-R2 and angiopoietin-Tie-2 axes being modulated in the kidney by haemodynamic factors in the diabetic context.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Kidney/physiology , Renin-Angiotensin System/physiology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Angiopoietin-1/genetics , Angiopoietin-2/genetics , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Gene Expression/physiology , Imidazoles/pharmacology , Male , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Receptor, TIE-2/genetics , Vasoconstrictor Agents/pharmacology
12.
Kidney Int ; 66(3): 1090-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15327403

ABSTRACT

BACKGROUND: Diabetic nephropathy is associated with increased albuminuria and accumulation of extracellular matrix proteins within the kidney. Clinical studies have shown some beneficial effects of calcium channel blockers (CCB) on diabetic nephropathy, even though they are generally considered to be less renoprotective than agents that interrupt the renin angiotensin system. However, effects of CCBs on renal injury, and in particular, expression of extracellular matrix proteins in a model of normotensive diabetic nephropathy, are poorly characterized. METHODS: Experimental diabetes was induced by injection of streptozocin in Sprague-Dawley rats. Amlodipine, a CCB which blocks the L channel, and mibefradil, a CCB blocking the T as well as the L channels, were given to diabetic rats for six months. Albumin excretion rate (AER), pathologic injury, and expression of the extracellular matrix proteins, collagen I, and fibronectin were assessed. RESULTS: Increased AER in diabetic rats (13.2 x//1.3 mg/d, geometric mean x// tolerance factor) was attenuated by either amlodipine (3.2 x// 1.4 mg/d) or mibefradil (2.6 x// 1.4 mg/d). Increased glomerulosclerosis and tubulointerstitial injury in diabetic animals were attenuated by amlodipine and mibefradil. There was increased collagen accumulation in the kidney of diabetic rats as assessed by picro-sirius red staining. Gene expression of both collagen I and fibronectin were also increased in the kidneys from diabetic animals, as assessed by reverse transcription-polymerase chain reaction (RT-PCR). These markers of fibrosis were attenuated by treatment with either amlodipine or mibefradil. Blood pressure in diabetic rats (136 +/- 2 mm Hg) was modestly reduced by amlodipine (126 +/- 3 mm Hg) but not by mibefradil treatment (134 +/- 3 mm Hg). CONCLUSION: Calcium channel blockers attenuated albuminuria, pathologic injury, and accumulation of extracellular matrix proteins in this normotensive model of diabetic nephropathy. These findings suggest that CCBs may be useful in preventing pathologic injury in the diabetic kidney.


Subject(s)
Amlodipine/pharmacology , Calcium Channel Blockers/pharmacology , Diabetic Nephropathies/drug therapy , Mibefradil/pharmacology , Albuminuria/drug therapy , Albuminuria/pathology , Animals , Azo Compounds , Blood Pressure/drug effects , Collagen Type I/genetics , Coloring Agents , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Fibronectins/genetics , Gene Expression/drug effects , Kidney/metabolism , Kidney/pathology , Male , Organ Size , Rats , Rats, Sprague-Dawley
13.
J Am Soc Nephrol ; 15(8): 2139-51, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15284299

ABSTRACT

NF-kappaB-dependent pathways play an important role in macrophage infiltration and kidney injury. NF-kappaB is regulated by angiotensin II (AII). However, the role of this pathway in diabetic nephropathy has not been clearly delineated. First, the activation of NF-kappaB, monocyte chemoattractant protein-1 (MCP-1), and macrophage infiltration in the diabetic kidney were explored, in a temporal manner. The active subunit of NF-kappaB, p65, was elevated in the diabetic animals in association with increased MCP-1 gene expression and macrophage infiltration. Second, the effects of treatment for 4 wk with the AII type 1 receptor antagonist valsartan, the AII type 2 receptor antagonist PD123319, or pyrrolidine dithiocarbamate, an inhibitor of NF-kappaB and on these parameters were assessed. These treatments were associated with a reduction in p65 activation, MCP-1 gene expression, and macrophage infiltration. These findings demonstrate a role for activation of NF-kappaB, in particular the p65 subunit, in the pathogenesis of early renal macrophage infiltration in experimental diabetes. In the context of the known proinflammatory effects of AII, it is postulated that the renoprotection conferred by angiotensin II receptor antagonism is at least partly related to the inhibition of NF-kappaB-dependent pathways.


Subject(s)
Angiotensin II/metabolism , Diabetic Nephropathies/pathology , Macrophages/pathology , NF-kappa B/metabolism , Angiotensin II Type 1 Receptor Blockers , Angiotensin II Type 2 Receptor Blockers , Animals , Antioxidants/pharmacology , Biomarkers , Chemokine CCL2/genetics , Diabetic Nephropathies/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression , I-kappa B Proteins/metabolism , Male , NF-KappaB Inhibitor alpha , NF-kappa B p50 Subunit , Pyrrolidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Thiocarbamates/pharmacology , Transcription Factor RelA
14.
Circulation ; 109(12): 1536-42, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15023892

ABSTRACT

BACKGROUND: It remains controversial whether specific blockade of the renin-angiotensin system confers superior antiatherosclerotic effects over other antihypertensive agents in diabetes. Therefore, the aim of this study was to compare equihypotensive doses of the angiotensin II subtype 1 (AT1) receptor blocker irbesartan with the calcium antagonist amlodipine on diabetes-induced plaque formation in the apolipoprotein E (apoE)-null mouse and to explore molecular and cellular mechanisms linked to vascular protection. METHODS AND RESULTS: Diabetes was induced by injection of streptozotocin in 6-week-old apoE-null mice. Diabetic animals were randomized to no treatment, irbesartan, or amlodipine for 20 weeks. Diabetes was associated with an increase in plaque area and complexity in the aorta in association with a significant increase in aortic AT1 receptor expression, cellular proliferation, collagen content, macrophage- and alpha-smooth muscle actin-positive cell infiltration, as well as an increased expression of platelet-derived growth factor-B (PDGF-B), monocyte chemoattractant protein-1 (MCP-1), and vascular cell adhesion molecule-1 (VCAM-1). Irbesartan but not amlodipine treatment attenuated the development of atherosclerosis, collagen content, cellular proliferation, and macrophage infiltration as well as diabetes-induced AT1 receptor, PDGF-B, MCP-1, and VCAM-1 overexpression in the aorta despite similar blood pressure reductions by both treatments. CONCLUSIONS: Diabetes-associated atherosclerosis is ameliorated by AT1 receptor blockade but not by calcium channel antagonism, providing further evidence for the vascular renin-angiotensin system playing a pivotal role in the development and acceleration of atherosclerosis in diabetes.


Subject(s)
Amlodipine/therapeutic use , Angiotensin II Type 1 Receptor Blockers , Antihypertensive Agents/therapeutic use , Arteriosclerosis/prevention & control , Biphenyl Compounds/therapeutic use , Calcium Channel Blockers/therapeutic use , Diabetes Mellitus, Experimental/complications , Tetrazoles/therapeutic use , Amlodipine/pharmacology , Animals , Antihypertensive Agents/pharmacology , Aortic Diseases/etiology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Biphenyl Compounds/pharmacology , Calcium Channel Blockers/pharmacology , Cell Division , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Collagen/metabolism , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Irbesartan , Macrophages/drug effects , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-sis/biosynthesis , Proto-Oncogene Proteins c-sis/genetics , Receptor, Angiotensin, Type 1/biosynthesis , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/physiology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Streptozocin , Tetrazoles/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
15.
Arterioscler Thromb Vasc Biol ; 24(5): 935-42, 2004 May.
Article in English | MEDLINE | ID: mdl-14988091

ABSTRACT

OBJECTIVE: Diabetes is associated with accelerated atherosclerosis, the major factor contributing to increased mortality and morbidity in the diabetic population. The molecular mechanisms by which diabetes promotes atherosclerosis are not fully understood. Platelet-derived growth factor has been shown to play a major role in the pathology of vascular diseases, but whether it plays a role in atherosclerosis associated with diabetes remains unknown. The aims of this study were to assess whether platelet-derived growth factor-dependent pathways are involved in the development of diabetes-induced atherosclerosis and to determine the effects of platelet-derived growth factor receptor antagonism on this disorder. METHODS AND RESULTS: Diabetes was induced by injection of streptozotocin in 6-week-old apolipoprotein E knockout mice. Diabetic animals received treatment with a tyrosine kinase inhibitor that inhibits platelet-derived growth factor action, imatinib (STI-571, 10 mg/kg per day), or no treatment for 20 weeks. Nondiabetic apolipoprotein E knockout mice served as controls. Induction of diabetes was associated with a 5-fold increase in plaque area in association with an increase in aortic platelet-derived growth factor-B expression and platelet-derived growth factor-beta receptor phosphorylation as well as other prosclerotic and proinflammatory cytokines. Imatinib treatment prevented the development of atherosclerotic lesions and diabetes-induced inflammatory cytokine overexpression in the aorta. CONCLUSIONS: Tyrosine kinase inhibition with imatinib appears to be a novel therapeutic option to retard the development of atherosclerosis, specifically in the context of diabetes.


Subject(s)
Arteriosclerosis/drug therapy , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/drug therapy , Piperazines/therapeutic use , Proto-Oncogene Proteins c-sis/physiology , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Animals , Aorta/pathology , Aortic Diseases/drug therapy , Aortic Diseases/etiology , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/etiology , Arteriosclerosis/pathology , Benzamides , Cytokines/biosynthesis , Cytokines/genetics , Diabetes Mellitus, Experimental/chemically induced , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Genes, abl , Genes, sis , Glycated Hemoglobin/analysis , Imatinib Mesylate , Intercellular Signaling Peptides and Proteins , Lipids/blood , Male , Mice , Mice, Knockout , Organometallic Compounds/metabolism , Peptides/genetics , Peptides/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-abl/biosynthesis , Proto-Oncogene Proteins c-sis/biosynthesis , Pyrimidines/pharmacology , Random Allocation , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , Streptozocin , Systole/drug effects , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
16.
Hypertension ; 43(2): 276-81, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14718351

ABSTRACT

Angiotensin II is a known stimulus for the expression of vascular endothelial growth factor (VEGF). This action of angiotensin II is mediated by the angiotensin type 1 (AT1) receptor. However, the role of the angiotensin type 2 (AT2) receptor subtype in inducing VEGF expression has been controversial. The aim of the present study was to assess the effects of AT2 receptor blockade on VEGF expression in the retina, initially in experimental diabetic rats induced by injection of streptozotocin. The AT1 receptor antagonist, valsartan, or the AT2 receptor antagonists, PD123319, were administered to diabetic rats for 4 weeks. Increased gene and protein expressions of VEGF, as assessed by real-time reverse transcription-polymerase chain reaction and immunostaining, respectively, were observed in the retina in diabetic rats. Treatment with either valsartan or PD123319 attenuated retinal VEGF expression. To further explore the link between angiotensin receptor subtypes and VEGF expression, valsartan, or PD123319 were administered to rats that were infused with angiotensin II for 2 weeks. VEGF expression was also increased in the retina from angiotensin II infused rats, and this was attenuated by valsartan and PD123319. These findings suggest that VEGF expression is modulated by AT1 and AT2 receptors, thereby implicating angiotensin II receptor subtypes in retinal diseases such as diabetic retinopathy.


Subject(s)
Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/physiology , Retina/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiotensin II/pharmacology , Angiotensin II Type 2 Receptor Blockers , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Gene Expression/drug effects , Imidazoles/pharmacology , Immunohistochemistry , Male , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/genetics
17.
J Am Soc Nephrol ; 14(12): 3061-71, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14638905

ABSTRACT

A link between angiotensin II and cell proliferation has previously been reported. However, there remains controversy as to the role of the individual angiotensin II receptor subtypes in mediating these effects and their link to angiogenic cytokines and their receptors. Male Sprague-Dawley rats were infused with either angiotensin II or vehicle for 14 d at a dose of 58.3 ng/min. Angiotensin II-infused rats received no treatment, an AT(1) receptor antagonist valsartan (30 mg/kg per d), or an AT(2) receptor antagonist PD123319 (830 ng/min). Gene expression of vascular endothelial growth factor (VEGF) and receptor VEGF-R2, as well as Tie-2 and its ligands angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2) were assessed by reverse transcription-PCR. Protein expression was assessed by Western blotting and immunohistochemistry. Gene and protein expression of VEGF, Ang-1, and Ang-2 were increased by angiotensin II infusion. Valsartan and PD123319 attenuated angiotensin II-associated increases in VEGF gene and protein expression. Ang-1 and Ang-2 gene but not protein expression were reduced by both treatments. These changes occurred in the context of attenuation of angiotensin II-induced glomerular cell proliferation by both valsartan and PD123319. In situ hybridization and immunohistochemical studies localized VEGF, Ang-1, and Ang-2 expression to the epithelial cells of the glomerulus, and VEGF-R2 and Tie-2 receptors to the endothelial cells of the kidney. These findings extend the increasing evidence that the AT(2) receptor, in addition to the AT(1) receptor subtype, plays an important role in mediating the proliferative actions of angiotensin II in the kidney.


Subject(s)
Angiopoietins/biosynthesis , Angiotensin II/physiology , Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/physiology , Vascular Endothelial Growth Factors/biosynthesis , Angiotensin II/administration & dosage , Animals , Gene Expression , Kidney/metabolism , Male , Protein Biosynthesis , Rats , Rats, Sprague-Dawley
18.
Am J Pathol ; 163(3): 879-87, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12937129

ABSTRACT

There is evidence that angiotensin II, vascular endothelial growth factor (VEGF), angiopoietins, and their cognate receptors participate in retinal angiogenesis. We investigated whether angiotensin type 2-receptor blockade (AT2-RB) reduces retinal angiogenesis and alters the expression of VEGF/VEGF-R2 and angiopoietin-Tie2. Retinopathy of prematurity (ROP) was induced in Sprague Dawley (SD) rats by exposure to 80% oxygen from postnatal (P) days 0 to 11, followed by 7 days in room air. ROP shams were in room air from P0-18. A group of ROP rats received the AT2-RB, PD123319, by mini-osmotic pump (5 mg/kg/day) from P11-18 (angiogenesis period). Evaluation of the retinal status of the AT2 receptor indicated that this receptor, as assessed by real-time PCR, immunohistochemistry, and in vitro autoradiography, was present in the retina, was more abundant than the AT1 receptor in the neonatal retina, and was increased in the ROP model. AT2-RB reduced retinal angiogenesis. VEGF and VEGF-R2 mRNA were increased in ROP and localized to blood vessels, ganglion cells, and the inner nuclear layer, and were decreased by PD123319. Angiopoietin2 and Tie2, but not angiopoietin1 mRNA were increased with ROP, and angiopoietin2 was reduced with PD123319. This study has identified a potential retinoprotective role for AT2-RB possibly mediated via interactions with VEGF- and angiopoietin-dependent pathways.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Endothelial Growth Factors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Neovascularization, Pathologic/physiopathology , Receptors, Angiotensin/metabolism , Retinal Diseases/physiopathology , Retinal Vessels/physiopathology , Aging/metabolism , Angiopoietin-2 , Animals , Animals, Newborn , Autoradiography , Computer Systems , Endothelial Growth Factors/genetics , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Neovascularization, Pathologic/pathology , Oxygen , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Retina/metabolism , Retinal Diseases/chemically induced , Retinal Diseases/pathology , Retinal Vessels/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factors
19.
Kidney Int ; 64(2): 565-71, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12846751

ABSTRACT

BACKGROUND: Angiotensin II type 1 (AT1) receptor antagonists and 3-hydroxy-3-methylglutaryl conenzyme A (HMG-CoA) reductase inhibitors have been shown to confer renoprotection. However, the renal effects of the combination of an AT1 receptor antagonist and an HMG-CoA reductase inhibitor in experimental diabetes are unknown. METHODS: Diabetes was induced by injection of streptozotocin in Wistar rats. Diabetic rats were randomly treated with losartan, an AT1 receptor antagonist, or simvastatin, an HMG-CoA reductase inhibitor, as well as the combination of both for eight weeks. Albumin excretion rate (AER) and plasma concentrations of blood urea nitrogen (BUN), creatinine, cholesterol, and triglycerides were measured. Renal injury was evaluated. Immunohistochemical staining of transforming growth factor beta1 (TGF beta 1) and vascular endothelial growth factor (VEGF) were performed. RESULTS: Increased AER in diabetic rats was attenuated by treatment with either losartan or simvastatin and further reduced by the combination of the two. Elevated plasma concentrations of BUN and creatinine were only reduced by the combination. There was no significant difference in plasma concentrations of cholesterol and triglycerides between control and diabetic rats and neither was influenced by losartan or simvastatin. Kidney pathologic injury was attenuated by losartan, but not simvastatin, compared to diabetic animals. Overexpression of TGF beta 1 and VEGF was observed in the glomeruli of diabetic rats and was attenuated by losartan, simvastatin, or the combination of both to a similar level. CONCLUSION: The combination of an angiotensin antagonist with an HMG-CoA reductase inhibitor confers superiority over monotherapies on renal function, as assessed by prevention of albuminuria and rise in plasma BUN and creatinine. However, no advantage of combination therapy was seen with respect to attenuating renal structural injury and renal expression of TGF beta and VEGF in experimental diabetes.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Antihypertensive Agents/pharmacology , Diabetic Nephropathies/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Losartan/pharmacology , Simvastatin/pharmacology , Albuminuria/drug therapy , Albuminuria/metabolism , Animals , Blood Urea Nitrogen , Creatinine/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Drug Therapy, Combination , Female , Kidney/metabolism , Kidney/pathology , Male , Rats , Rats, Wistar , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Vascular Endothelial Growth Factor A/metabolism
20.
Kidney Int ; 63(6): 2152-61, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12753302

ABSTRACT

BACKGROUND: The renal clearance of organic cations is important for the homeostasis of a number of exogenous and endogenous compounds. The organic cation transporters (OCTs) situated on the basolateral surface of proximal tubular cells mediate active cation excretion. Alterations of cation transport may occur in diabetes, although the role of the OCTs has not been previously assessed. METHODS: Experimental diabetes was induced in rats with streptozotocin (55 mg/kg) and animals were randomly assigned to receive ramipril (3 mg/mL) in drinking water for 24 weeks. In a second protocol, rats were infused with angiotensin II (Ang II) at a dose of 58.3 ng/kg/min for 2 weeks via an implanted osmotic pump. Expression of the OCTs and renal clearance of the endogenous cation N-methyl-nicotinamide (NMN) was assessed. RESULTS: Diabetes was associated with a reduction in gene and protein expression of both OCT-1 and OCT-2 and a reduction in NMN clearance. These effects were prevented by ramipril, associated with the prevention of albuminuria and tubular injury as demonstrated by the expression of osteopontin and glutathione peroxidase 3 (GPX-3). An infusion of Ang II also reduced NMN clearance but without altering the renal expression of OCTs. CONCLUSION: We hypothesize that reduced expression of OCTs in diabetes may be a marker of tubular injury. However, Ang II may also directly augment renal cation clearance independent of changes in transporter expression. Together these effects may provide additional mechanism to explain treatment-related improvements in creatinine clearance and renoprotection in diabetes following blockade of the renin-angiotensin system (RAS).


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Kidney Tubules/metabolism , Niacinamide/analogs & derivatives , Ramipril/pharmacology , Animals , Biological Transport/drug effects , Cations/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Gene Expression , Male , Niacinamide/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...