Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Janet Woodcock-Mitchell is active.

Publication


Featured researches published by Janet Woodcock-Mitchell.


Circulation | 1998

Increased Plasminogen Activator Inhibitor Type 1 in Coronary Artery Atherectomy Specimens From Type 2 Diabetic Compared With Nondiabetic Patients A Potential Factor Predisposing to Thrombosis and Its Persistence

Burton E. Sobel; Janet Woodcock-Mitchell; David J. Schneider; Robert E. Holt; Kousuke Marutsuka; Herman K. Gold

BACKGROUND Inhibition of fibrinolysis attributable to elevated concentrations of plasminogen activator inhibitor type 1 (PAI-1) in blood is associated with insulin resistance, hyperinsulinemia, and type 2 diabetes mellitus. Because we have shown that insulin can stimulate PAI-1 synthesis in vivo and because accelerated vascular disease is common in such patients as well, we hypothesized that increased PAI-1, potentially predisposing to thrombosis, acute occlusion, and accelerating atherosclerosis because of thrombus-associated mitogens, would be present in excess in atheroma from type 2 diabetic subjects. METHODS AND RESULTS Samples acquired by directional coronary atherectomy from 25 patients with type 2 diabetes and 18 patients without diabetes were characterized qualitatively histologically for cellularity and by immunohistochemistry visually and qualitatively and by quantitative image analysis for assessment of urokinase-type plasminogen activator (u-PA) and PAI-1. Patients with and without diabetes were similar with respect to demographic features and the distribution and severity of coronary artery disease. Substantially more PAI-1 and substantially less u-PA were present in the atherectomy samples from subjects with diabetes. CONCLUSIONS The disproportionate elevation of PAI-1 compared with u-PA observed in atheromatous material extracted from vessels of diabetic subjects is consistent with increased gene expression of PAI-1 in vessels as well as the known increase of PAI-1 in blood, presumably reflecting increased synthesis. The increased PAI-1 detected in the atheroma may contribute in vivo to accelerated or persistent thrombosis underlying acute occlusion and to vasculopathy exacerbated by clot-associated mitogens in the vessel wall. Because the changes were observed to be associated with insulin resistance and type 2 diabetes mellitus, they may be modifiable by reduction of insulin resistance with insulin sensitizers and stringent control of hyperglycemia.


Circulation | 1998

Induction of plasminogen activator inhibitor type 1 and type 1 collagen expression in rat cardiac microvascular endothelial cells by interleukin-1 and its dependence on oxygen-centered free radicals

Hitoshi Okada; Janet Woodcock-Mitchell; John J. Mitchell; Tomohiro Sakamoto; Kousuke Marutsuka; Burton E. Sobel; Satoshi Fujii

BACKGROUND Ischemia with or without reperfusion induces the release of diverse products from monocytes, including cytokines such as interleukin-1 (IL-1). To determine whether these phenomena modulate fibrinolysis and potentially exacerbate impairment of the macrocirculation, microcirculation, or both, we characterized the effects of IL-1 on the expression of fibrinolytic system and matrix proteins in rat cardiac microvascular endothelial cells (CMECs). METHODS AND RESULTS Confluent CMECs were exposed to IL-1 in serum-free medium for 24 hours, and cell-conditioned medium was assayed for plasminogen activator inhibitor type 1 (PAI-1), the primary physiological inhibitor of plasminogen activators, and for type 1 collagen with Western blotting. IL-1 (2 ng/mL) specifically increased the accumulation of PAI-1 (4.4 +/- 0.6-fold; mean +/- SD; n = 9) without affecting tissue plasminogen activator (t-PA) or urokinase plasminogen activator (u-PA) levels, which remained unchanged. IL-1 increased the accumulation of collagen in conditioned media by 3.5 +/- 0.7-fold (n = 6). Conversely, the accumulation of both PAI-1 and collagen induced by IL-1 was inhibited with an IL-1 receptor antagonist (200 ng/mL; n = 6) and with cycloheximide (10 micrograms/mL; n = 6), implying that protein synthesis was a requirement for the effect. To determine whether the IL-1 effect was mediated by induction of oxygen-centered free radical production, known to be induced by IL-1, we exposed the cells to the hydroxyl radical scavenger tetramethylthiourea (10 mmol/L) and observed abolition of the IL-1-induced increase in the expression of PAI-1 and collagen (n = 6). Conversely, superoxides (generated with 10 mU/mL xanthine oxidase plus 0.6 mmol/L hypoxanthine, and 100 mumol/L hydrogen peroxide) induced the accumulation of PAI-1 and collagen (n = 6). IL-1 (1 microgram/kg body wt) and lipopolysaccharide (50 micrograms/kg body wt) administered in vivo increased PAI-1 protein in rat hearts as detected with Western blotting and PAI-1 immunostaining of rat heart microvessels, indicating the effects delineated in vitro were paralleled by effects in vivo. CONCLUSIONS These results indicate that IL-1-induced oxygen-centered free radicals stimulate elaboration of PAI-1 and collagen by CMECs. Accordingly, microvascularly mediated inhibition of fibrinolysis may predispose to the persistence of microvascular thrombi, thereby contributing to impaired microcirculatory function, the no-reflow phenomenon, and cardiac dysfunction after ischemia and reperfusion.


American Journal of Physiology-cell Physiology | 1999

TNF-α and insulin, alone and synergistically, induce plasminogen activator inhibitor-1 expression in adipocytes

Tomohiro Sakamoto; Janet Woodcock-Mitchell; Kousuke Marutsuka; John J. Mitchell; Burton E. Sobel; Satoshi Fujii

Obesity is associated with hyperinsulinemia and elevated concentrations of tumor necrosis factor-alpha (TNF-alpha) in adipose tissue. TNF-alpha has been implicated as an inducer of the synthesis of plasminogen activator inhibitor-1 (PAI-1), the primary physiological inhibitor of fibrinolysis, mediated by plasminogen activators in cultured adipocytes. To identify mechanism(s) through which TNF-alpha induces PAI-1, 3T3-L1 preadipocytes were differentiated into adipocytes and exposed to TNF-alpha for 24 h. TNF-alpha selectively increased the synthesis of PAI-1 without increasing activity of plasminogen activators. Both superoxide (generated by xanthine oxidase plus hypoxanthine) and hydrogen peroxide were potent inducers of PAI-1, and hydroxyl radical scavengers completely abolished the TNF-alpha induction of PAI-1. Exposure of adipocytes to TNF-alpha or insulin alone over 5 days increased PAI-1 production. These agonists exert synergistic effects. Results obtained suggest that TNF-alpha stimulates PAI-1 production by adipocytes, an effect potentiated by insulin, and that adipocyte generation of reactive oxygen centered radicals mediates the induction of PAI-1 production by TNF-alpha. Because induction of PAI-1 by TNF-alpha is potentiated synergistically by insulin, both agonists appear likely to contribute to the impairment of fibrinolytic system capacity typical in obese, hyperinsulinemic patients.


American Journal of Physiology-lung Cellular and Molecular Physiology | 1998

Smooth muscle actin and myosin expression in cultured airway smooth muscle cells

Jean Z. Wong; Janet Woodcock-Mitchell; John D. Mitchell; Patricia Rippetoe; Sheryl L. White; Marlene Absher; Linda Baldor; John N. Evans; Kirk M. McHugh; Robert B. Low

In this study, the expression of smooth muscle actin and myosin was examined in cultures of rat tracheal smooth muscle cells. Protein and mRNA analyses demonstrated that these cells express alpha- and gamma-smooth muscle actin and smooth muscle myosin and nonmuscle myosin-B heavy chains. The expression of the smooth muscle specific actin and myosin isoforms was regulated in the same direction when growth conditions were changed. Thus, at confluency in 1 or 10% serum-containing medium as well as for low-density cells (50-60% confluent) deprived of serum, the expression of the smooth muscle forms of actin and myosin was relatively high. Conversely, in rapidly proliferating cultures at low density in 10% serum, smooth muscle contractile protein expression was low. The expression of nonmuscle myosin-B mRNA and protein was more stable and was upregulated only to a small degree in growing cells. Our results provide new insight into the molecular basis of differentiation and contractile function in airway smooth muscle cells.In this study, the expression of smooth muscle actin and myosin was examined in cultures of rat tracheal smooth muscle cells. Protein and mRNA analyses demonstrated that these cells express α- and γ-smooth muscle actin and smooth muscle myosin and nonmuscle myosin-B heavy chains. The expression of the smooth muscle specific actin and myosin isoforms was regulated in the same direction when growth conditions were changed. Thus, at confluency in 1 or 10% serum-containing medium as well as for low-density cells (50-60% confluent) deprived of serum, the expression of the smooth muscle forms of actin and myosin was relatively high. Conversely, in rapidly proliferating cultures at low density in 10% serum, smooth muscle contractile protein expression was low. The expression of nonmuscle myosin-B mRNA and protein was more stable and was upregulated only to a small degree in growing cells. Our results provide new insight into the molecular basis of differentiation and contractile function in airway smooth muscle cells.


Coronary Artery Disease | 1998

Pathogenetic implications of hyaluronan-induced modification of vascular smooth muscle cell fibrinolysis in diabetes.

Kousuke Marutsuka; Janet Woodcock-Mitchell; Tomohiro Sakamoto; Burton E. Sobel; Satoshi Fujii

BackgroundProteolysis, modulated in part by intramural fibrinolytic system proteins and their inhibitors, appears to influence vascular smooth muscle cell (SMC) migration and proliferation and remodeling of extracellular matrix (ECM). Alterations of fibrinolysis in circulating blood and of proteolysis within vessel walls in experimental animals and patients with diabetes have been associated with accelerated vascular disease. Hyaluronan, a prominent component of ECM in normal vessels, is increased in the tunica media of macroscopically normal arterial vessels from patients with type 2 diabetes. ObjectiveTo determine whether hyaluronan alters the expression of the fibrinolytic system protein, plasminogen activator inhibitor type-1 (PAI-1), in human vascular SMCs, thereby potentially accelerating vascular disease in patients with type 2 diabetes. MethodsUrokinase-type and tissue-type plasminogen activators (uPA and tPA) and PAI-1 were assayed in vascular SMC conditioned media and in cell lysates, using enzyme-linked immunosorbent assay and western blotting. ResultsHyaluronan increased the 24-h release of PAI-1 into conditioned media in a concentration-dependent and time-dependent manner (1.8-fold compared with control with 1 mg/ml hyaluronan; n = 9, P < 0.01). Although the accumulation of uPA in conditioned media tended to increase also, uPA content was reduced in cell lysates (64% of control with 0.1 mg/ml hyaluronan at 24 h; n = 9, P < 0.01) without any change in PAI-1. Concentrations of tPA in conditioned media and cell lysates were unchanged. Digestion of hyaluronan with hyaluronidase (50 turbidity reducing units (TRU)/ml) or exposure of the smooth muscle cells to antihuman CD44 antibody (1 μg/ml) that binds to the hyaluronan cell surface receptor obviated the effects of hyaluronan. ConclusionOur results indicate that increases in hyaluronan increase vascular SMC expression of PAI-1, a phenomenon that may alter the balance between proteolysis and its inhibition in vessels of patients with type 2 diabetes, thereby contributing to the acceleration of macroangiopathy.


Angiogenesis | 1999

Long-term blockade of nitric oxide synthesis in rats modulates coronary capillary network remodeling.

Daisuke Goto; Satoshi Fujii; A.K.M. Tarikuz Zaman; Ichiro Sakuma; Ming Gao; Tomiyasu Koyama; John J. Mitchell; Janet Woodcock-Mitchell; Burton E. Sobel; Akira Kitabatake

Long-term blockade of nitric oxide synthesis with Nω-nitro-L-arginine methyl ester (L-NAME) induces cardiac perivascular fibrosis in rats. Its relationship to expression of angiogenic growth factors and capillary network remodeling is not understood. This study was designed to determine whether capillary proliferation and angiogenic growth factor regulation occur in response to L-NAME. Three groups of rats were studied: C, control; L1, L-NAME 13 mg/kg/day; L2, 130 mg/kg/day. One and eight weeks later the hearts were removed and subjected to morphometric analysis and analysis of gene expressions of molecules related to angiogenesis. Arterial hypertension was observed within 8 weeks in the L1 and L2 groups compared with control. After 1 week immunohistochemical assays demonstrated basic fibroblast growth factor (bFGF) in the arteriolar media. Northern blot analysis revealed increase in bFGF and transforming growth factor-β (TGF-β) mRNA during this period. At 8 weeks arteriolar medial thickening and perivascular fibrosis were seen microscopically in the L1 and L2 groups, which were accompanied by only a modest remodeling of capillary network due to increase in venular or intermediate capillary portions. Concomitantly immunoreactivity for vascular endothelial growth factor (VEGF) and TGF-β were detected in perivascular area. These results suggest that (1) blockade of NO synthesis induces expression of angiogenic growth factors as well as vessel wall remodeling, and (2) TGF-β may counteract angiogenic growth factors and limit subsequent alterations in capillary network remodeling.


Thrombosis Research | 1997

A NEW BUTADIENE DERIVATIVE, T-686, INHIBITS PLASMINOGEN ACTIVATOR INHIBITOR TYPE-1 PRODUCTION IN VITRO BY CULTURED HUMAN VASCULAR ENDOTHELIAL CELLS AND DEVELOPMENT OF ATHEROSCLEROTIC LESIONS IN VIVO IN RABBITS

Boris Vinogradsky; Stephen P. Bell; Janet Woodcock-Mitchell; Akio Ohtani; Satoshi Fujii

Plasminogen activator inhibitor-1 (PAI-1), the major physiologic inhibitor of tissue-type plasminogen activator and urokinase, is abundantly expressed in atherosclerotic vascular wall. To determine the role of PAI-1 in vascular wall, we have used a novel inhibitor of PAI-1, (3E, 4E)-3-benzylidene-4-(3,4,5-trimethoxy-benzylidene) -pyrrolidine-2,5-dione (T-686). T-686 was given to human vascular endothelial cells in vitro and to rabbits subjected to high cholesterol diet and mechanical injury in vivo. T-686 attenuated the augmentation of PAI-1 antigen accumulation induced by transforming growth factor beta in conditioned medium from the human umbilical vein endothelial cells. In rabbits with aortic atherosclerosis induced by hypercholesterolemia and implantation of indwelling plastic tubing, oral administration of T-686 (30mg/kg body weight/day) for 8 weeks attenuated the increase in plasma PAI-1 activity induced by vascular injury without decreasing blood triglyceride and cholesterol. This was accompanied by the reduction in aortic PAI-1 mRNA expression and the inhibition of development of atherosclerosis lesions. Thus, T-686 not only decreased PAI-1 synthesis in vascular cells in vitro but also protected against the development of vascular lesions in vivo. This compound may be useful in defining the role of PAI-1 in atherothrombotic states.


Coronary Artery Disease | 1996

Leukaemia inhibitory factor and oncostatin M modulate expression of urokinase plasminogen activator and fibrinogen

Hitoshi Okada; Janet Woodcock-Mitchell; Satoshi Fujii

BackgroundPathogenetic effects of active immune cell products on the coagulation and fibrinolytic system proteins in liver and endothelial cells - primary sites of synthesis of these proteins - have not been elucidated. MethodsWe incubated highly differentiated human hepatoma cells (Hep G2) and human umbilical vein endothelial cells (HUVECs) for 24 h with recombinant human leukaemia inhibitory factor (LIF) and oncostatin M (OSM) - cytokines that are implicated in acute phase reactions during inflammation and which bind to the same cell surface receptor unit (glycoprotein gp130). UF was. also given to mice in vivo. Contents of coagulation and fibrinolytic system proteins in cell culture supematants and in mouse liver were determined. ResultsLIF increased the accumulation of urokinase plasminogen activator (u-PA) in the Hep G2 cell culture supematants determined by enzyme-linked immunosorbent assay (ELISA) (0.21 ± 0.03 (SE) μg/ml at baseline; 0.40 ± 0.05 μg/ml at 100 μ/ml, P< 0.05; 0.57 ± 0.06 ng/ml at 500 U/ml, P< 0.01; n = 9) without altering total protein content. OSM elicited a similar effect (0.25 ± 0.04 ng/ml at baseline, 0.62 ± 0.19 ng/ml at 1 ng/ml; P<0.05, n = 6). A monoclonal antibody against gp130 abrogated the response to both agents (n = 9). Rasminogen activator inhibitor type-1 (PAI-1) (assayed by ELISA, n = 9), the PAI-1 binding protein, vitronectin (immunoprecipitation, n = 3) and tissue factor (ELISA, n = 3) were not affected by LIF, but fibrinogen production increased up to twofold with LIF (500 U/ml; Western blot, n = 3). In HUVECs, synthesis of tissue type plasminogen activator or PAI-1 was not altered by UF or OSM (ELISA, n = 9). In vivo, intraperitoneal recombinant murine LIF (2 μg) increased liver concentrations of u-PA by 30% and fibrinogen by 220% in mice. ConclusionsLIF and OSM produced by immune cells may modify fibrinolysis and coagulation by altering expression of u-PA and fibrinogen, thereby contributing to coagulopathy.


Differentiation | 1988

α-Smooth muscle actin is transiently expressed in embryonic rat cardiac and skeletal muscles

Janet Woodcock-Mitchell; John J. Mitchell; Robert B. Low; Madeleine Kieny; Philippe Sengel; Laura Rubbia; Omar Skalli; Brian W. Jackson; Giulio Gabbiani


American Journal of Respiratory Cell and Molecular Biology | 1990

Smooth Muscle Cell Markers in Developing Rat Lung

John J. Mitchell; Susan E. Reynolds; Kevin O. Leslie; Robert B. Low; Janet Woodcock-Mitchell

Collaboration


Dive into the Janet Woodcock-Mitchell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenneth B. Adler

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John D. Mitchell

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge