Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
QJM ; 106(10): 909-14, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23696676

ABSTRACT

BACKGROUND: It has not been fully elucidated whether antihypertensive medication adherence affects blood pressure (BP) control in hypertension cases. AIM: To investigate the association of adherence to antihypertensive drug regimens and BP control using data from the Combination Pill of Losartan Potassium and Hydrochlorothiazide for Improvement of Medication Compliance Trial (COMFORT) study. DESIGN: An observational analysis from a randomized controlled trial. METHODS: A total of 203 hypertensive subjects were randomly assigned to a daily regimen of a combination pill (losartan 50 mg/hydrochlorothiazide 12.5 mg) or two pills, an angiotensin II receptor blocker and a thiazide diuretic. Medication adherence calculated based on pill counts and BPs was evaluated at 1, 3 and 6 months after randomization. RESULTS: The subjects were divided into three groups according to their adherence, i.e. relatively low-adherence (<90%; n = 19), moderate-adherence (90-99%; n = 71) and high-adherence (100%; n = 113) groups. Clinical characteristics of the subjects including BP, sex, randomized treatments and past medical history did not differ significantly among the three groups. Achieved follow-up BPs over the 6-month treatment period, which were adjusted for age, sex, baseline BP and randomized treatment, were significantly higher in the low-adherence group (135/78 mmHg) compared with the high-adherence (130/74 mmHg; P = 0.02/0.02) and the moderate-adherence (128/74 mmHg; P = 0.003/0.02) groups. CONCLUSION: Low adherence to an antihypertensive-drug regimen was associated with poor BP control.


Subject(s)
Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Hypertension/drug therapy , Medication Adherence , Aged , Antihypertensive Agents/pharmacology , Drug Combinations , Female , Humans , Hydrochlorothiazide/economics , Hydrochlorothiazide/therapeutic use , Hypertension/physiopathology , Japan/epidemiology , Losartan/economics , Losartan/therapeutic use , Male , Middle Aged , Patient Education as Topic , Prospective Studies , Treatment Outcome
2.
Proc Natl Acad Sci U S A ; 99(2): 913-8, 2002 Jan 22.
Article in English | MEDLINE | ID: mdl-11773635

ABSTRACT

A deletion mutation Delta K210 in cardiac troponin T (cTnT) was recently found to cause familial dilated cardiomyopathy (DCM). To explore the effect of this mutation on cardiac muscle contraction under physiological conditions, we determined the Ca(2+)-activated force generation in permeabilized rabbit cardiac muscle fibers into which the mutant and wild-type cTnTs were incorporated by using our TnT exchange technique. The free Ca(2+) concentrations required for the force generation were higher in the mutant cTnT-exchanged fibers than in the wild-type cTnT-exchanged ones, with no statistically significant differences in maximal force-generating capability and cooperativity. Exchanging the mutant cTnT into isolated cardiac myofibrils also increased the free Ca(2+) concentrations required for the activation of ATPase. In contrast, a deletion mutation Delta E160 in cTnT that causes familial hypertrophic cardiomyopathy (HCM) decreased the free Ca(2+) concentrations required for force generation, just as in the case of the other HCM-causing mutations in cTnT. The results indicate that cTnT mutations found in the two distinct forms of cardiomyopathy (i.e., HCM and DCM) change the Ca(2+) sensitivity of cardiac muscle contraction in opposite directions. The present study strongly suggests that Ca(2+) desensitization of force generation in sarcomere is a primary mechanism for the pathogenesis of DCM associated with the deletion mutation Delta K210 in cTnT.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/physiopathology , Sequence Deletion , Troponin T/genetics , Troponin T/metabolism , Adenosine Triphosphatases/metabolism , Animals , Humans , In Vitro Techniques , Muscle Fibers, Skeletal/physiology , Myocardial Contraction/physiology , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
3.
J Mol Cell Cardiol ; 33(12): 2095-107, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11735257

ABSTRACT

Functional consequences of the six mutations (R145G, R145Q, R162W, DeltaK183, G203S, K206Q) in cardiac troponin I (cTnI) that cause familial hypertrophic cardiomyopathy (HCM) were studied using purified recombinant human cTnI. The missense mutations R145G and R145Q in the inhibitory region of cTnI reduced the intrinsic inhibitory activity of cTnI without changing the apparent affinity for actin. On the other hand, the missense mutation R162W in the second troponin C binding region and the deletion mutation DeltaK183 near the second actin-tropomyosin region reduced the apparent affinity of cTnI for actin without changing the intrinsic inhibitory activity. Ca(2+) titration of a fluorescent probe-labeled human cardiac troponin C (cTnC) showed that only R162W mutation impaired the cTnC-cTnI interaction determining the Ca(2+) affinity of the N-terminal regulatory domain of cTnC. Exchanging the human cardiac troponin into isolated cardiac myofibrils or skinned cardiac muscle fibers showed that the mutations R145G, R145Q, R162W, DeltaK183 and K206Q induced a definite increase in the Ca(2+)-sensitivity of myofibrillar ATPase activity and force generation in skinned muscle fibers. Although the mutation G203S also showed a tendency to increase the Ca(2+) sensitivity in both myofibrils and skinned muscle fibers, no statistically significant difference compared with wild-type cTnI could be detected. These results demonstrated that most of the HCM-linked cTnI mutations did affect the regulatory processes involving the cTnI molecule, and that at least five mutations (R145G, R145Q, R162W, DeltaK183, K206Q) increased the Ca(2+) sensitivity of cardiac muscle contraction.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/genetics , Mutation , Myofibrils/metabolism , Troponin I/genetics , Troponin I/metabolism , Actins/metabolism , Adenosine Triphosphatases/metabolism , Animals , Calcium/metabolism , Cardiomyopathy, Hypertrophic, Familial/metabolism , Fluorescent Dyes/metabolism , Humans , Muscle Fibers, Skeletal/metabolism , Myocardium/metabolism , Naphthalenesulfonates/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Swine , Titrimetry , Troponin C/metabolism
4.
Hypertens Res ; 24(5): 579-88, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11675954

ABSTRACT

Retinoids have been shown to promote vascular smooth muscle cell differentiation, although the underlying mechanism is unclear. In fact, treatment of rat aortic smooth muscle cells with all-trans retinoic acid (ATRA) has been shown to markedly elevate the mRNA and protein levels of smooth muscle alpha-actin. Considering that an exit from the cell cycle is a prerequisite for cell differentiation, we examined the effect of ATRA on cellular events during the progression from Go to S phase. Pretreatment with ATRA dose-dependently inhibited DNA synthesis induced by basic fibroblast growth factor. However, ATRA did not inhibit transient activation of mitogen-activated protein kinase (MAPK) in response to mitogenic stimulation. And ATRA consistently failed to influence the phosphorylation of MAPK kinase (MEK) and the expression of MAPK-specific dual phosphatase (MKP-1). ATRA did not interfere with other early mitogenic signals either, such as the phosphorylation of FGF-1 receptor or the induction of immediate early genes c-fos, c-jun, and c-myc. In contrast, ATRA strongly suppressed the pRb kinase activities of the cyclin-dependent kinases (Cdks) Cdk4, Cdk6, and Cdk2. ATRA did not influence the expressions of Cip/Kip family Cdk inhibitors or those of cyclins D1 and D2, whereas it strongly inhibited the expressions of cyclins D3 and E, Cdk4, Cdk6, and Cdk2. These results suggest that ATRA targets multiple genes essential for entry into the cell cycle and for the subsequent progression to G1 phase, but without interrupting early mitogenic signals upstream of MAPK.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Muscle, Smooth, Vascular/cytology , Tretinoin/pharmacology , Actins/genetics , Animals , Aorta/cytology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , DNA/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
5.
Int J Urol ; 8(4): 184-7, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11260351

ABSTRACT

A case is reported of renal pelvic carcinoma of the horseshoe kidney in a 69-year-old man, which showed an interesting metastatic pattern by implantation in the prostate. A few months after transurethral resection of the prostate for benign prostate hyperplasia and extracorporal shock wave lithotripsy for renal stones, the patient complained of severe back pain due to multiple metastatic bone tumors. Autopsy revealed transitional cell carcinoma in the pelvis as well as in the prostate with remarkable vessel invasion. The clinical course and autopsy findings suggested that the systemic expansion of cancer cells from the renal pelvis was caused not only by direct metastasis but also by implantation in the prostate.


Subject(s)
Carcinoma, Transitional Cell/secondary , Kidney Neoplasms/pathology , Kidney Pelvis , Kidney/abnormalities , Prostatic Neoplasms/secondary , Aged , Humans , Male
6.
Mol Pharmacol ; 58(4): 837-44, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10999955

ABSTRACT

In search of substances useful for the treatment of atherosclerotic vascular diseases, we studied the effects of 15-deoxy-Delta(12, 14)-prostaglandin J(2) (15d-PGJ(2)), a natural ligand for peroxisome proliferator-activated receptor gamma, on the proliferation and differentiation of vascular smooth muscle cells (VSMCs). 15d-PGJ(2) but not WY14643, an agonist for peroxisome proliferator-activated receptor alpha, dose-dependently inhibited VSMC proliferation; the effect was maximal at 12 microM. This compound strongly suppressed the activities of cyclin-dependent kinases (Cdk) 4, 6, and 2, thereby preventing the phosphorylation of the retinoblastoma protein. These Cdks seemed to be inhibited through two mechanisms: the down-regulation of cyclin D1 and the up-regulation of Cdk inhibitor p21(Cip1/Waf1/Sdi1). 15d-PGJ(2) was found to inhibit the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, which mediates cyclin D1 expression. Mitogenic stimulation of quiescent cells decreased the level of mRNA for the smooth muscle-specific myosin heavy-chain SM1, whereas this reduction was prevented by 15d-PGJ(2). A long-term treatment of exponentially growing VSMCs with 15d-PGJ(2) markedly elevated the mRNA level of SM1 and, moreover, induced SM2, another isoform expressed exclusively in mature VSMCs. 15d-PGJ(2) also increased the expression levels of calponin-h1 and smooth muscle alpha-actin. These results suggest that 15d-PGJ(2) induces G(1) arrest by two distinct mechanisms and promotes VSMC differentiation.


Subject(s)
Antigens, Differentiation/biosynthesis , G1 Phase/drug effects , Muscle, Smooth, Vascular/drug effects , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Protein Serine-Threonine Kinases , Cell Division/drug effects , Cells, Cultured , Cyclin D1/antagonists & inhibitors , Cyclin D1/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/biosynthesis , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Up-Regulation
7.
J Hypertens ; 18(7): 901-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10930188

ABSTRACT

OBJECTIVE: To verify whether hypertension confers a hypercoagulable state in a hypertensive animal model. DESIGN: The parameters of blood coagulation were compared between stroke-prone spontaneously hypertensive rats (SHR-SP) and Wistar-Kyoto (WKY) rats. Each rat group consisted of a younger subgroup at 8-12 weeks old (n = 12) and an older subgroup at 16-20 weeks old (n = 12). METHODS: Prothrombin time (PT), activated partial thromboplastin time (APTT), fluorogenic PT, fibrinogen, fibrin/fibrinogen degradation products (FDP), thrombin-anti-thrombin III complex (TAT), factor Xa activity, anti-thrombin III (AT-III), tissue factor pathway inhibitor (TFPI), protein C and C1 inhibitor were measured in both rat groups. RESULTS: There was no significant difference in FDP and TAT levels between SHR-SP and WKY rats even at 16-20 weeks when SHR-SP developed severe hypertensive vascular lesions. Contrary to expectations, fluorogenic PT and factor Xa activity were significantly lower in SHR-SP than in WKY rats. While there was no significant difference in AT-III, TFPI and protein C activities between SHR-SP and WKY rats, C1 inhibitor activity was significantly higher in SHR-SP than in WKY rats. The elevated C1 inhibitor activity was inversely correlated with the reduced factor Xa activity. Gel-filtered fractionated plasma with C1 inhibitor activity had an inhibitory effect on the purified rat factor Xa, and immunodepletion of C1 inhibitor from the fractionated plasma attenuated the inhibitory effect CONCLUSION: These results suggest that SHR-SP get into a hypocoagulable state rather than a hypercoagulable state, and that the reduction of factor Xa activity in SHR-SP may be related to the elevation of C1 inhibitor activity.


Subject(s)
Blood Coagulation Disorders/blood , Blood Coagulation , Escherichia coli Proteins , Hypertension/blood , Membrane Transport Proteins , Stroke/blood , Animals , Blood Cell Count , Blood Coagulation Disorders/complications , Blood Coagulation Disorders/pathology , Carrier Proteins/metabolism , Complement C1 Inactivator Proteins/metabolism , Factor Xa/metabolism , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Hypertension/complications , Hypertension/pathology , Lipoproteins/metabolism , Male , Partial Thromboplastin Time , Protein C/metabolism , Prothrombin Time , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Stroke/complications , Stroke/pathology
8.
Hinyokika Kiyo ; 46(4): 273-6, 2000 Apr.
Article in Japanese | MEDLINE | ID: mdl-10845161

ABSTRACT

We report here 3 cases of papillary adenocarcinoma of the prostate. In all 3 cases, the tumors were discernible on cystourethroscopy and transurethral biopsy established the diagnosis, whereas no significant finding was found on digital rectal examination. Although androgen deprivation therapy was administered in all cases, different surgical procedures were employed according to the stage in each case. In case 1, since the papillary tumor was confined within the prostatic urethra, complete resection was accomplished by transurethral resection (TUR). In case 2, since pelvic lymph nodes metastases were found, local radiation therapy was added. In case 3, since the patient had vesical invasion of tumor total cysto-prostatectomy was performed. Papillary adenocarcinoma of the prostate originates from the prostatic duct, resulting in existence at the "central portion" of the prostate gland. Cystourethroscopy and transurethral biopsy is helpful for diagnosis of this disease, whereas rectal digital examination is useless. As a surgical procedure for the primary site, TUR may be efficient for tumors confined within the prostatic urethra, although more extensive surgery may be necessary for those with a more invasive profile.


Subject(s)
Adenocarcinoma, Papillary , Prostatic Neoplasms , Adenocarcinoma, Papillary/pathology , Adenocarcinoma, Papillary/radiotherapy , Adenocarcinoma, Papillary/surgery , Aged , Combined Modality Therapy , Cystectomy , Cystoscopy , Humans , Male , Prostatectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/surgery
9.
Circ Res ; 86(9): 967-73, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10807869

ABSTRACT

Ligands for peroxisome proliferator-activated receptor gamma, such as the thiazolidinedione class of antidiabetic drugs and 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), modulate various processes in atherogenesis. In search of cells that generate prostaglandin D(2) (PGD(2)), the metabolic precursor of 15d-PGJ(2), we identified PGD(2) from culture medium of endothelial cells. To study how PGD(2) production is regulated in endothelial cells, we investigated the role of fluid shear stress in the metabolism of PGD(2). Endothelial cells expressed the mRNA for the lipocalin-type PGD(2) synthase (L-PGDS) both in vitro and in vivo. Loading laminar shear stress using a parallel-plate flow chamber markedly enhanced the gene expression of L-PGDS, with the maximal effect being obtained at 15 to 30 dyne/cm(2). The expression began to increase within 6 hours after loading shear stress and reached the maximal level at 18 to 24 hours. In contrast, shear stress did not alter the expression levels of PGI(2) synthase and thromboxane A(2) synthase. In parallel with the increase in the expression level of L-PGDS, endothelial cells released PGD(2) and 15d-PGJ(2) into culture medium. These results demonstrate that shear stress promotes PGD(2) production by stimulating L-PGDS expression and suggest the possibility that a peroxisome proliferator-activated receptor gamma ligand is produced in vascular wall in response to blood flow.


Subject(s)
Endothelium, Vascular/enzymology , Intramolecular Oxidoreductases/metabolism , Arteries/metabolism , Cells, Cultured , Culture Media/metabolism , Endothelium, Vascular/cytology , Humans , Lipocalins , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/biosynthesis , Stress, Mechanical
10.
Circ Res ; 86(1): 68-75, 2000.
Article in English | MEDLINE | ID: mdl-10625307

ABSTRACT

Differentiation-inducing factor-1 (DIF-1) is a morphogen that induces differentiation of DICTYOSTELIUM: Recently, DIF-1 has been shown to inhibit proliferation and induce differentiation in tumor cells, although the underlying mechanisms remain unknown. In this study, we examined the effects of DIF-1 on the proliferation and differentiation of vascular smooth muscle cells, to explore novel therapeutic strategies for atherosclerosis. DIF-1 nearly completely inhibited DNA synthesis and cell division in mitogen-stimulated cells. DIF-1 inhibited the phosphorylation of the retinoblastoma protein and the activities of cyclin-dependent kinase (Cdk) 4, Cdk6, and Cdk2, which phosphorylate the retinoblastoma protein. DIF-1 strongly suppressed the expression of cyclins D1, D2, and D3, as well as those of cyclins E and A, which normally began after that of the D-type cyclins. The mRNAs for the smooth muscle myosin heavy chains SM1 and SM2 were expressed in quiescent cells in primary culture, and these expression levels decreased after mitogenic stimulation. In the presence of DIF-1, the rate of the reduction was significantly decelerated. Moreover, the addition of DIF-1 to dedifferentiated cells induced the expressions of SM1 and SM2, accompanied by a reduction in the level of SMemb, a nonmuscle-type myosin heavy chain. Therefore, DIF-1 seemed to interrupt a very early stage of G(1) probably by suppressing the expressions of the D-type cyclins. Furthermore, this compound may prevent phenotypic modulation and induce differentiation of vascular smooth muscle cells.


Subject(s)
G1 Phase/drug effects , Hexanones/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Enzyme Inhibitors/metabolism , Humans , Isoenzymes/metabolism , Mitogens/physiology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myosin Heavy Chains/metabolism , Signal Transduction/drug effects
11.
Circ Res ; 86(2): 185-90, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10666414

ABSTRACT

Alterations in the functions of vascular endothelial cells (ECs) induced by fluid shear stress may play a pivotal role in both the development and prevention of vascular diseases. We found that DNA synthesis of bovine aortic and human umbilical vein ECs, determined by [(3)H]thymidine incorporation, was inhibited by steady laminar shear stress (5 and 30 dyne/cm(2)). This growth inhibition due to shear stress was associated with suppression of cell transition from the G(1) to S phase of the cell cycle. Therefore, we studied G(1)-phase events to find the molecules responsible for this cell cycle arrest. Shear stress inhibited the phosphorylation of a retinoblastoma protein (pRb) and the activity of cyclin-dependent kinase (cdk) 2 and cdk4, which phosphorylate pRb. The level of cdk inhibitor p21(Sdi1/Cip1/Waf1) protein, but not that of p27(Kip1), increased as a result of shear stress, and the amount of p21 protein associated with cdk2 also increased, although the protein level of cdk2 was unchanged. Shear stress markedly elevated the mRNA level of p21, and this elevation in mRNA faded after the release of cells from shear stress, concomitant with a recovery of DNA synthesis. These results suggest that steady laminar shear stress induces cell cycle arrest by upregulating p21. Derangement of the steady laminar flow may release cells from this inhibition and induce cell proliferation, which, in turn, may cause atherosclerosis through the induction of EC stability disruption.


Subject(s)
CDC2-CDC28 Kinases , Cyclins/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Proto-Oncogene Proteins , Animals , Aorta/cytology , Cattle , Cell Division/physiology , Cells, Cultured , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA/biosynthesis , Enzyme Activation/physiology , Flow Cytometry , Gene Expression Regulation, Enzymologic , Humans , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/analysis , Retinoblastoma Protein/metabolism , Stress, Mechanical , Substrate Specificity , Umbilical Veins/cytology
12.
Biochem Biophys Res Commun ; 268(1): 25-30, 2000 Feb 05.
Article in English | MEDLINE | ID: mdl-10652206

ABSTRACT

The Jak/STAT pathway is activated following stimulation of the type I angiotensin II receptor. To examine whether this pathway is shared among other G-protein-coupled receptors, we studied the linkage between the alpha(1) adrenergic receptor and this pathway. The alpha(1) agonist phenylephrine induced tyrosine phosphorylation of Jak2, Tyk2, and STAT1 in vascular smooth muscle cells. The phosphorylation of Jak2 was prevented by the alpha(1) receptor antagonists prazosin and chloroethylclonidine, but not by WB4101, and that of STAT1 was inhibited by prazosin and the Jak2 inhibitor AG490. After stimulation with phenylephrine, Jak2 and STAT1 were found to associate with alpha(1B) receptor. Phenylephrine stimulated the DNA binding activity of STAT1. Protein synthesis promoted by phenylephrine was inhibited by prazosin, AG490, and the introduction of a decoy oligonucleotide for STAT1. These results suggested that alpha(1) receptor is linked to the Jak/STAT pathway and that this pathway mediates alpha(1) agonist-induced smooth muscle hypertrophy.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-Agonists/pharmacology , Amino Acid Sequence , Base Sequence , Cell Division/physiology , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Janus Kinase 2 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Oligodeoxyribonucleotides/genetics , Phenylephrine/pharmacology , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Proteins/chemistry , Proteins/metabolism , STAT1 Transcription Factor , Signal Transduction , TYK2 Kinase , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection , Tyrosine/metabolism
13.
Mol Pharmacol ; 56(5): 938-46, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10531398

ABSTRACT

Cyclin-dependent kinase inhibitor p21(Waf1/Cip1/Sdi1) has been suggested to be involved in the antiproliferative effect of nitric oxide (NO) in vascular smooth muscle cells (VSMCs). To elucidate the mechanism underlying NO-induced p21 expression, we investigated the roles of tumor suppressor p53 and the guanylate cyclase-cGMP pathway. The induction of p21 by the NO donor S-nitroso-N-acetylpenicillamine (SNAP) seemed to be due to transactivation because SNAP elevated the activity of p21 promoter but did not stabilize p21 mRNA and protein. Because SNAP did not stimulate the deletion mutant of p21 promoter that lacked p53 binding sites, we tested the involvement of p53. The expression level of p53 was down-regulated after mitogenic stimulation, whereas it was sustained in the presence of SNAP. SNAP markedly stimulated DNA binding activity of p53. Furthermore, SNAP failed to induce p21 in VSMCs obtained from p53-knock out mice and in A431 cells that contained mutated p53. The antiproliferative effect of SNAP also was attenuated in these cells. NO stimulates guanylate cyclase and its product cGMP has been shown to inhibit VSMC proliferation. However, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, a guanylate cyclase inhibitor, did not prevent SNAP-induced p21 expression. 8-Bromo-cGMP, 3-isobutyl-1-methylxanthine, and their combination did not induce p21. Although 8-bromo-cGMP had a small antiproliferative effect, the elevation of cGMP concentration induced by SNAP was little throughout the G(1) phase. The antiproliferative effect of SNAP was not attenuated by Rp-8-bromoguanosine-3',5'-monophosphorothioate, an inhibitor of cGMP-dependent protein kinase. These results suggested that NO induces p21 through a p53-dependent but cGMP-independent pathway.


Subject(s)
Cyclic GMP/physiology , Cyclins/biosynthesis , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/physiology , Tumor Suppressor Protein p53/physiology , Cell Division/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Genes, Tumor Suppressor/physiology , Humans , Muscle, Smooth, Vascular/cytology , RNA, Messenger/biosynthesis
14.
Pathol Int ; 49(6): 571-6, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10469403

ABSTRACT

A myoepithelioma occurred in the subcutaneous tissue of the right shoulder of a 28-year-old man. The well-demarcated nodular tumor (3. 0 x 2.8 cm) was located in the subcutaneous tissue with no adhesion to the deltoid muscle. The tumor was composed of a fascicular proliferation of spindle cells with variable amounts of stroma and showed areas of sheets of epithelioid cells. In most areas, the tumor cells had uniform nuclei, but pleomorphic epithelioid cells were focally present. Mitotic activity was three per 10 high-power fields. No ductular structure was found throughout the tumor. Immunohistochemical and ultrastructural studies confirmed the myoepithelial origin of the tumor cells. The occurrence of myoepithelioma in the subcutaneous tissue has been rarely reported. Even though the tumor showed no aggressive behavior on the 2-year follow-up, it is still too early to comment definitely on the behav- ior of myoepithelioma of the subcutaneous tissue. This case provides further information about soft tissue myoepithelioma.


Subject(s)
Myoepithelioma/pathology , Soft Tissue Neoplasms/pathology , Adult , Antibodies, Monoclonal , Biomarkers, Tumor/analysis , Diagnosis, Differential , Epithelioid Cells/ultrastructure , Humans , Immunoenzyme Techniques , Leiomyosarcoma/diagnosis , Male , Myoepithelioma/chemistry , Myoepithelioma/surgery , Shoulder/pathology , Soft Tissue Neoplasms/chemistry , Soft Tissue Neoplasms/surgery
15.
Hum Genet ; 104(3): 205-10, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10323243

ABSTRACT

p57KIP2 is a potent tight-binding inhibitor of several G1 cyclin/cyclin-dependent kinase (Cdk) complexes, and is a negative regulator of cell proliferation. The gene encoding p57KIP2 is located at 11p15.5, a region implicated in both sporadic cancers and Beckwith-Wiedemann syndrome (BWS). Previously we demonstrated that p57KIP2 is imprinted and only the maternal allele is expressed in both mice and humans. We also showed mutations found in p57KIP2 in patients with BWS that were transmitted from the patients' carrier mothers, indicating that the expressed maternal allele was mutant and that the repressed paternal allele was normal. In the study reported here, we performed functional analysis of the two mutated p57KIP2 genes. We showed that the nonsense mutation found in the Cdk inhibitory domain in a BWS patient rendered the protein inactive with consequent complete loss of its role as a cell cycle inhibitor and of its nuclear localization. We also showed that the mutation in the QT domain, although completely retaining its cell cycle regulatory activity, lacked nuclear localization and was thus prevented from performing its role as an active cell cycle inhibitor. Consequently, no active p57KIP2 would have existed, which might have caused the disorders in BWS patients.


Subject(s)
Beckwith-Wiedemann Syndrome/genetics , CDC2-CDC28 Kinases , Nuclear Proteins/genetics , Animals , COS Cells , Cell Nucleus/enzymology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p57 , Cyclin-Dependent Kinases/antagonists & inhibitors , Escherichia coli/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Bacterial , Humans , Mutation , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , Plasmids/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
16.
Atherosclerosis ; 138(2): 247-53, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9690907

ABSTRACT

Effect of interleukin 4 (IL-4) on the production of matrix metalloproteinase 1 (MMP-1) by normal and immortalized human intimal smooth muscle cells (SMC) was investigated. The production of the precursors of MMP-1 by intimal SMC was enhanced in a dose-dependent manner by addition of IL-4 to the culture medium, whereas the cytokine also showed an inhibitory effect on DNA synthesis in the cells. In addition, mRNA of IL-4 was found in the atherosclerotic and nonatherosclerotic areas of the intima. Although the production of MMP-1 and the proliferation of SMC are thought to play an important role in reconstruction of the intima during atherogenesis, our results suggest a possible role of IL-4 induced MMP-1 in inhibiting tissue remodeling caused by a variety of arterial disorders including atherosclerosis.


Subject(s)
Collagenases/biosynthesis , Interleukin-4/pharmacology , Muscle, Smooth, Vascular/metabolism , Aorta/enzymology , Aorta/metabolism , Arteriosclerosis/metabolism , Cells, Cultured , Collagenases/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Humans , Interleukin-4/metabolism , Matrix Metalloproteinase 1 , Muscle, Smooth, Vascular/enzymology , Promoter Regions, Genetic , RNA, Messenger/biosynthesis
17.
Biochem Mol Biol Int ; 45(4): 841-7, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9713709

ABSTRACT

Poikilocytosis of red blood cells (RBCs) was observed to be associated with anemia in rats given subcutaneous injections of cadmium (Cd). Phase-contrast light and scanning electron microscopic examinations revealed that acanthocytes appeared in the early stages of administration, and that the number of RBC fragments increased later. Ultrastructural analysis of RBC ghosts by negative staining demonstrated that the normal lattice structure of the membrane skeleton was abolished. The osmotic fragility curve of the Cd-exposed RBCs disclosed that most of the cells were less fragile than control RBCs. These data indicate that the RBC membrane skeleton is initially altered by Cd-exposure, followed by deformation of the cell, thus promoting intrasplenic hemolysis, and resulting in anemia.


Subject(s)
Anemia, Hypochromic/chemically induced , Cadmium/toxicity , Erythrocyte Membrane/drug effects , Acanthocytes/pathology , Anemia, Hypochromic/blood , Animals , Erythrocyte Count , Erythrocyte Membrane/ultrastructure , Erythrocytes, Abnormal/pathology , Female , Hemolysis , Male , Microscopy, Electron , Osmotic Fragility , Rats
18.
J UOEH ; 20(1): 11-9, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9551525

ABSTRACT

Splenomegaly was observed both in male and female Sprague-Dawley rats after 1 week of exposure to CdCl2 (0.6 mg Cd/kg/day). Spleen weight reached about double that in controls by 8 weeks of Cd exposure. Histopathological examination of the enlarged spleen revealed that iron- and lipid-laden histiocytes were clustered in the periarterial lymphatic sheath, and the red pulp appeared to be expanded. It is noteworthy that electron microscopy revealed marked poikilocytosis and Heinz body formation in red blood cells (RBCs) in both the sinus and cord. Histiocytes were swollen by a granular substance in the cytoplasm and also many secondary lysosomes. These morphological findings indicate that degradation of damaged RBCs induced by exposure to Cd might be promoted in the spleen and possibly cause splenomegaly. This RBC damage-hemolysis-splenomegaly sequence is also considered to be associated with the etiology of Cd-induced anemia. In addition to the abnormal RBC degradation, nuclei of lymphocytes in the Cd-exposed spleen exhibited high electron density, consistent with a preapoptotic state suggesting the immunosuppressive effect of Cd.


Subject(s)
Anemia/chemically induced , Cadmium Poisoning/complications , Cadmium/toxicity , Splenomegaly/pathology , Anemia/blood , Anemia/complications , Animals , Erythrocytes, Abnormal/drug effects , Female , Male , Microscopy, Electron , Rats , Rats, Sprague-Dawley , Spleen/ultrastructure , Splenomegaly/chemically induced
19.
J Biol Chem ; 272(15): 10050-7, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9092548

ABSTRACT

Nitric oxide-generating vasodilators inhibit vascular smooth muscle cell proliferation. To elucidate the mechanism underlying this process, we investigated the effect of S-nitroso-N-acetylpenicillamine (SNAP), a nitric oxide-releasing agent, on the smooth muscle cell cycle. When G0 cells were stimulated with fetal bovine serum and basic fibroblast growth factor, DNA synthesis assessed by [3H]thymidine incorporation started about 15 h later. SNAP dose-dependently inhibited this incorporation, and this effect was maximal at 100 microM. This inhibition was attenuated when SNAP was added after 9-12 h. SNAP inhibited the activity of cyclin-dependent kinase 2 (Cdk2) and phosphorylation of the retinoblastoma protein, both of which usually increased from about 9 h, whereas it did not inhibit the activities of cyclin D-associated kinase(s), Cdk4, and Cdk6, which normally increased from 0-3 h. Although SNAP reduced the mRNA levels of cyclins E and A, it neither reduced their protein levels nor impaired their association with Cdk2. SNAP did not reduce the mRNA levels of cyclins G, C, and D1, Cdk2, Cdk4, and Cdk5, which were normally elevated from 0-3 h. The mRNA and protein levels of the Cdk inhibitor p21 were high in the early G1 phase, peaking at 3 h and then rapidly decreasing after 6 h. In the presence of SNAP, however, p21 expression was enhanced, and moreover, the later decrease disappeared. SNAP also increased the amount of Cdk2-associated p21. These results suggested that nitric oxide inhibits the G1/S transition by inhibiting Cdk2-mediated phosphorylation of the retinoblastoma protein and that p21 induction is involved in the Cdk2 inhibition.


Subject(s)
Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/biosynthesis , Enzyme Inhibitors/metabolism , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Tumor Suppressor Proteins , Vasodilator Agents/pharmacology , Animals , Cattle , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , DNA Replication/drug effects , Humans , Microtubule-Associated Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Phosphorylation , Proteins , RNA, Messenger/metabolism , S-Nitroso-N-Acetylpenicillamine , Tumor Suppressor Protein p53/pharmacology , Up-Regulation/drug effects
20.
Atherosclerosis ; 129(2): 207-13, 1997 Mar 21.
Article in English | MEDLINE | ID: mdl-9105563

ABSTRACT

It has been confirmed that the receptor protein encoded by the c-kit proto-oncogene is expressed by cells of the hematopoietic, gonadal, pigment, and mast cell lineages and that its ligand, stem cell factor (SCF), is mainly expressed in their microenvironment. In a previous study we investigated the expression of the c-kit gene in human aortic endothelial cells (EC). In the present study we investigated the expression of SCF in human aortic EC and smooth muscle cells (SMC). Reverse transcription (RT)-PCR and Northern blot analyses showed that both human arterial EC and SMC expressed mRNA specific for the SCF gene. In addition, tissue-specific expression of the SCF gene was confirmed by in situ hybridization in the EC and the SMC. Western blot analysis and immunocytochemistry showed evidence of production of SCF protein in both the EC and the SMC. These results indicate the existence of mast cell-SMC interaction and of an autocrine loop of c-kit and its ligand on the surface of EC, suggesting that the interaction between c-kit protein and SCF may play an important role in metabolism of arterial wall and in the pathogenesis of atherosclerosis in the arterial intima.


Subject(s)
Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Stem Cell Factor/biosynthesis , Animals , Aorta/cytology , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Cell Line, Transformed , Cell Lineage , Cells, Cultured , Endothelium, Vascular/cytology , Gene Expression Regulation , Humans , In Situ Hybridization , Mast Cells/metabolism , Mice , Molecular Probe Techniques , Muscle, Smooth, Vascular/cytology , Polymerase Chain Reaction , Proto-Oncogene Mas , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...