Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 129(10): 1308-1319, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28082445

RESUMO

p53-related protein kinase (TP53RK, also known as PRPK) is an upstream kinase that phosphorylates (serine residue Ser15) and mediates p53 activity. Here we show that TP53RK confers poor prognosis in multiple myeloma (MM) patients, and, conversely, that TP53RK knockdown inhibits p53 phosphorylation and triggers MM cell apoptosis, associated with downregulation of c-Myc and E2F-1-mediated upregulation of pro-apoptotic Bim. We further demonstrate that TP53RK downregulation also triggers growth inhibition in p53-deficient and p53-mutant MM cell lines and identify novel downstream targets of TP53RK including ribonucleotide reductase-1, telomerase reverse transcriptase, and cyclin-dependent kinase inhibitor 2C. Our previous studies showed that immunomodulatory drugs (IMiDs) downregulate p21 and trigger apoptosis in wild-type-p53 MM.1S cells, Importantly, we demonstrate by pull-down, nuclear magnetic resonance spectroscopy, differential scanning fluorimetry, and isothermal titration calorimetry that IMiDs bind and inhibit TP53RK, with biologic sequelae similar to TP53RK knockdown. Our studies therefore demonstrate that either genetic or pharmacological inhibition of TP53RK triggers MM cell apoptosis via both p53-Myc axis-dependent and axis-independent pathways, validating TP53RK as a novel therapeutic target in patients with poor-prognosis MM.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mieloma Múltiplo/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/imunologia , Apoptose/fisiologia , Western Blotting , Técnicas de Silenciamento de Genes , Humanos , Fatores Imunológicos/farmacologia , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/patologia , Prognóstico , Transdução de Sinais/efeitos dos fármacos
2.
Urology ; 76(4): 1017.e7-12, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20719367

RESUMO

OBJECTIVES: To clarify the basic mechanism involved in the pathophysiology of cystitis by characterizing the urodynamic parameters, pharmacologically relevant (muscarinic and purinergic) receptors, and the in vivo release of adenosine triphosphate (ATP) in the bladder of hydrochloric acid (HCl)-treated rats. METHODS: The muscarinic and purinergic receptors in rat tissue were measured by radioreceptor assays using (N-methyl-³H) scopolamine methyl chloride ([³H]NMS) and αß-methylene-ATP (2,8-³H) tetrasodium salt ([³H]αß-MeATP), respectively. The urodynamic parameters and ATP levels were measured using a cystometric method and the luciferin-luciferase assay, respectively. RESULTS: In the HCl-treated rats, the micturition interval and micturition volume were significantly (48% and 55%, respectively, P <.05) decreased and the number of micturitions was significantly (3.2-fold, P <.05) increased compared with those of the control rats. The maximal number of binding sites for [³H]NMS and [³H]αß-MeATP was significantly (55% and 72%, respectively, P <.001) decreased in the bladder of HCl-treated rats, suggesting downregulation of both muscarinic and purinergic receptors. In the HCl-treated rats, the inhibition constant, K(i), values for oxybutynin, solifenacin, and darifenacin were significantly (1.3-1.4-fold, P <.05) increased, but those for tolterodine and AF-DX116 were unchanged. Similarly, the inhibition constant for A-317491, pyridoxal-phosphate-6-azophenyl-2',4'-disulfonic acid tetrasodium, and MRS2273 was significantly (5.5, 11, and 7.6-fold, respectively, P <.001) increased. Furthermore, the in vivo release of ATP was significantly (P <.05) enhanced in the HCl-treated rat bladder. CONCLUSIONS: Both muscarinic and purinergic mechanisms might be, at least in part, associated with the urinary dysfunction due to cystitis.


Assuntos
Cistite/metabolismo , Ácido Clorídrico/toxicidade , Receptores Muscarínicos/análise , Receptores Purinérgicos/análise , Bexiga Urinária/química , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Animais , Compostos Benzidrílicos/metabolismo , Benzofuranos/metabolismo , Cresóis/metabolismo , Cistite/induzido quimicamente , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Ácidos Mandélicos/metabolismo , N-Metilescopolamina , Organofosfonatos/metabolismo , Fenóis/metabolismo , Fenilpropanolamina/metabolismo , Pirenzepina/análogos & derivados , Pirenzepina/metabolismo , Compostos Policíclicos/metabolismo , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/metabolismo , Pirrolidinas/metabolismo , Quinuclidinas/metabolismo , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Succinato de Solifenacina , Tetra-Hidroisoquinolinas/metabolismo , Tartarato de Tolterodina , Bexiga Urinária/efeitos dos fármacos , Micção , Urodinâmica
3.
Arterioscler Thromb Vasc Biol ; 29(10): 1529-36, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628784

RESUMO

BACKGROUND: The angiotensin II (Ang II) type 1 (AT(1)) receptor is expressed in bone marrow (BM) cells, whereas it remains poorly defined how Ang II regulates differentiation/proliferation of monocyte-lineage cells to exert proatherogenic actions. METHODS AND RESULTS: We generated BM chimeric apoE(-/-) mice repopulated with AT(1)-deficient (Agtr1(-/-)) or wild-type (Agtr1(+/+)) BM cells. The atherosclerotic development was significantly reduced in apoE(-/-)/BM-Agtr1(-/-) mice compared with apoE(-/-)/BM-Agtr1(+/+) mice, accompanied by decreased numbers of BM granulocyte/macrophage progenitors (GMP:c-Kit(+)Sca-1(-)Lin(-)CD34(+)CD16/32(+)) and peripheral blood monocytes. Macrophage-colony-stimulating factor (M-CSF)-induced differentiation from hematopoietic stem cells (HSCs:c-Kit(+)Sca-1(+)Lin(-)) to promonocytes (CD11b(high)Ly-6G(low)) was markedly reduced in HSCs from Agtr1(-/-) mice. The expression of M-CSF receptor c-Fms was decreased in HSCs/promonocytes from Agtr1(-/-) mice, accompanied by a marked inhibition in M-CSF-induced phosphorylation of PKC-delta and JAK2. c-Fms expression in HSCs/promonocytes was mainly regulated by TNF-alpha derived from BM CD45(-)CD34(-) stromal cells, and Ang II specifically regulated the TNF-alpha synthesis and release from BM stromal cells. CONCLUSIONS: Ang II regulates the expression of c-Fms in HSCs and monocyte-lineage cells through BM stromal cell-derived TNF-alpha to promote M-CSF-induced differentiation/proliferation of monocyte-lineage cells and contributes to the proatherogenic action.


Assuntos
Células-Tronco Hematopoéticas/citologia , Monócitos/citologia , Receptor Tipo 1 de Angiotensina/fisiologia , Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Aterosclerose/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Janus Quinase 2/metabolismo , Fator Estimulador de Colônias de Macrófagos/sangue , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteína Quinase C-delta/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/análise , Receptores de LDL/fisiologia , Fator de Necrose Tumoral alfa/fisiologia
4.
Neurosci Lett ; 436(1): 81-4, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18372110

RESUMO

We characterized muscarnic and purinergic receptors and urodynamic parameters in the bladder of cyclophosphamide (CYP)-treated rats to clarify the mechanisms involved in the pathophysiology of interstitial cystitis (IC). In the cystometry of CYP-treated rats compared with control rats, the micturition interval and micturition volume were significantly (55% and 77%, respectively) decreased and the frequency of micturition and basal pressure were significantly (3 and 2.3 times, respectively) increased. These changes in urodynamic parameters may characterize the detrusor overactivity occurring in CYP-treated rats. The maximal number of binding sites (B(max)) for specific binding of [N-methyl-(3)H]scopolamine methyl chloride ([(3)H]NMS) and alphabeta-methylene ATP [2,8-(3)H]tetrasodium salt ([(3)H]alphabeta-MeATP) was significantly (43% and 31%, respectively) decreased in the bladder of CYP-treated rats compared with control rats. On the other hand, the apparent dissociation constant (K(d)) for neither radioligand was significantly altered by the CYP treatment. K(i) value for the inhibition of bladder [(3)H]NMS binding by antimuscarinic agents (oxybutynin, tolterodine, darifenacin, and AF-DX 116) did not differ significantly between control and CYP-treated rats. The inhibition constant (K(i)) for the inhibition of bladder [(3)H]alphabeta-MeATP binding by purinergic antagonists (A-317491, PPADS) was significantly higher in CYP-treated rats than control rats. In conclusion, CYP treatment has been shown to cause down-regulation of pharmacologically relevant (muscarinic and purinergic) receptors in the bladder of rats. Thus, the present study offers further pharmacological evidence that both muscarinic and purinergic mechanisms contribute significantly to the urinary dysfunction due to IC.


Assuntos
Cistite Intersticial/metabolismo , Cistite Intersticial/fisiopatologia , Receptores Muscarínicos/metabolismo , Receptores Purinérgicos/metabolismo , Bexiga Urinária/fisiopatologia , Urodinâmica , Animais , Ciclofosfamida/toxicidade , Cistite Intersticial/induzido quimicamente , Feminino , Imunossupressores/toxicidade , Ratos , Ratos Sprague-Dawley , Bexiga Urinária/efeitos dos fármacos
5.
J Mol Cell Cardiol ; 40(6): 799-809, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16678200

RESUMO

Application of ultrasound-mediated destruction of microbubbles (US + Bubble) to skeletal muscle creates capillary ruptures leading to leakage of the cell components. We studied whether US + Bubble combined with bone-marrow-derived mononuclear cells (BM-MNCs) infusion enables the targeted delivery of endothelial-lineage cells into the myocardium and improves cardiac function of the cardiomyopathy model due to the paucity of neocapillary formation. Pulsed US was applied to the anterior chest of BIOTO2 cardiomyopathy hamsters for 90 s after the intravenous injection of microbubble (Optison) followed by infusion of BM-MNCs. Cardiac samples from US + microbubble + BM-MNCs (US + Bubble + BM), US + Bubble, US + BM without Bubble, and saline infusion control groups were analyzed 12 weeks after treatment. Labeled BM-MNCs transplanted by US + Bubble were found to be mainly localized in the microvessels, but not by US stimulation without microbubble (121.2 +/- 24.5 vs. 2.80 +/- 1.30 cells/mm2, P < 0.001). Capillary densities in US + Bubble + BM group were increased 1.7-fold (P < 0.05) over the control, and neither US + Bubble nor US + BM enhanced neocapillary formation. 99mTc-Tetrofosmin scintigraphy revealed that blood perfusion area in the US + Bubble + BM group was 48% greater than the control (P < 0.01). US + Bubble stimulation induces the expression of adhesion molecules (VCAM-1 and ICAM-1) in capillaries, and the US + Bubble-mediated supply of BM-MNCs increased the myocardial content of VEGF and bFGF. The left ventricular wt/body wt, area of cardiac fibrosis, and apoptotic cell numbers in the US + Bubble + BM group significantly (P < 0.05) decreased by 82%, 73%, and 64% relative to the control, respectively. The cardiac function in myopathic hamsters (assessed by fractional shortening) was markedly improved 36% (P < 0.05) by US + Bubble + BM treatment. Targeted delivery of BM-MNCs by US + Bubble to the myocardium of the cardiomyopathic hamster increased the capillary densities and regional blood flow and inhibited cardiac remodeling, resulting in the prevention of heart failure. This non-invasive cell delivery system may be useful as a novel efficient approach for angiogenic cell therapy to the myocardium.


Assuntos
Ecocardiografia , Células Endoteliais/citologia , Coração/fisiologia , Microbolhas , Miocárdio/citologia , Neovascularização Fisiológica , Células-Tronco/citologia , Indutores da Angiogênese/metabolismo , Animais , Peso Corporal , Células da Medula Óssea/citologia , Moléculas de Adesão Celular/metabolismo , Linhagem da Célula , Transplante de Células , Vasos Coronários , Cricetinae , Citocinas/genética , Tamanho do Órgão , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ultrassom , Remodelação Ventricular
6.
Arterioscler Thromb Vasc Biol ; 26(4): 751-7, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16439710

RESUMO

BACKGROUND: Granulocyte colony-stimulating factor (G-CSF) treatment was shown to inhibit neointimal formation of balloon-injured vessels, whereas neither the identification of progenitor cells involved in G-CSF-mediated endothelial regeneration with a bone marrow (BM) transplant experiment nor the functional properties of regenerated endothelium have been studied. METHODS AND RESULTS: Recombinant human G-CSF (100 microg/kg per day) was injected daily for 14 days starting 3 days before balloon injury in the rat carotid artery. Neointimal formation of denuded vessels on day 14 was markedly attenuated by G-CSF (39% versus the control; P<0.05). Endothelial cell-specific immunostaining revealed an enhancement of re-endothelialization (1.8-fold increase versus the control; P<0.05) and inhibition of extravasation of Evans Blue dye (47%; P=0.02). The regenerated endothelium exhibited acetylcholine-mediated vasodilatation in NO-dependent manner. G-CSF increased the circulating c-Kit+/Flk-1+ cells (9.1-fold; P<0.02), which showed endothelial properties in vitro (acetylated low-density lipoprotein uptake and lectin binding) and incorporated into the regenerated endothelium in vivo. A BM replacement experiment with green fluorescent protein (GFP)-overexpressing cells showed that BM-derived GFP+/CD31+ endothelial cells occupied 39% of the total luminal length in the G-CSF-mediated neo-endothelium (2% in the control). CONCLUSIONS: The G-CSF-induced mobilization of BM-derived c-Kit+/Flk-1+ cells contributes to endothelial regeneration, and this cytokine therapy may be a feasible strategy for the promotion of re-endothelialization after angioplasty.


Assuntos
Lesões das Artérias Carótidas/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Regeneração , Túnica Íntima/fisiopatologia , Animais , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/fisiopatologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Artéria Carótida Primitiva/fisiologia , Proliferação de Células , Células Cultivadas , Endotélio Vascular/patologia , Humanos , Hiperplasia/prevenção & controle , Injeções Subcutâneas , Camundongos , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Proto-Oncogênicas c-kit , Ratos , Ratos Endogâmicos Lew , Proteínas Recombinantes , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Vasodilatação
7.
Hypertens Res ; 29(11): 923-34, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17345793

RESUMO

We examined the effects of angiotensin II (Ang II) on inward rectifier K+ currents (IK1) in rat atrial myocytes. [125I]Ang II-binding assays revealed the presence of both Ang II type 1 (AT1) and type 2 (AT2) receptors in atrial membrane preparations. Ang II inhibited IK1 in isolated atrial myocytes with an IC50 of 46 nmol/l. This inhibition was abolished by the AT, antagonist RNH6270 but not at all by the AT2 antagonist PD123319. Treatment of cells with pertussis toxin or a synthetic decapeptide corresponding to the carboxyl-terminus of Gialpha-3 abolished the inhibition by Ang II, indicating the role of a Gi-dependent signaling pathway. Accordingly, Ang II failed to inhibit IK1 in the presence of forskolin, dibutyryl-cAMP or protein kinase A catalytic subunits. In spite of the increased binding capacities for [125I]Ang II, Ang II failed to affect IKI in cells from spontaneously hypertensive rats (SHR). AT, immunoprecipitation from atrial extracts revealed decreased amounts of Gialpha-2 and Gialpha-3 proteins associated with this receptor in SHR as compared with controls. The reduced coupling of AT, with Gialpha. proteins may underlie the unresponsiveness of atrial IK1 to Ang II in SHR cells.


Assuntos
Angiotensina II/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Animais , Expressão Gênica , Átrios do Coração/citologia , Técnicas In Vitro , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Arterioscler Thromb Vasc Biol ; 25(10): 2128-34, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16051876

RESUMO

OBJECTIVE: Ultrasound (US)-mediated destruction of contrast microbubbles causes capillary rupturing that stimulates arteriogenesis, whereas intramuscular implantation (im) of bone marrow mononuclear cells (BM-MNCs) induces angiogenesis. We therefore studied whether US-targeted microbubble destruction combined with transplantation of BM-MNCs can enhance blood flow restoration by stimulating both angiogenesis and arteriogenesis. METHODS AND RESULTS: US-mediated destruction of phospholipid-coated microbubbles was applied onto ischemic hindlimb muscle and subsequently BM-MNCs were transfused. A significant enhancement in blood flow recovery after Bubble+US+BM-MNC infusion (34% increase, P<0.05) was observed compared with Bubble+US (25%). The ratio of capillary/muscle fiber increased by Bubble+US+BM-MNC-i.v (260%, P<0.01) than that in the Bubble+US group (172%), into which BM-MNCs were incorporated (angiogenesis). Smooth muscle alpha-actin-positive arterioles were also increased, and angiography showed augmented collateral vessel formation (arteriogenesis). Platelet-derived proinflammatory factors activated by Bubble+US induces the expression of adhesion molecules (P-selectin and ICAM-1), leading to the attachment of transplanted BM-MNCs on the endothelium. Flow assay confirmed that the platelet-derived factors cause the adhesion of BM-MNCs onto endothelium under laminar flow. CONCLUSIONS: This study demonstrates that the targeted delivery of BM-MNCs by US destruction of microbubbles enhances regional angiogenesis and arteriogenesis response, in which the release of platelet-derived proinflammatory factors activated by Bubble+US play a key role in the attachment of transplanted BM-MNCs onto the endothelial layer.


Assuntos
Transplante de Medula Óssea/métodos , Isquemia/terapia , Microbolhas , Neovascularização Fisiológica/fisiologia , Ultrassonografia de Intervenção/métodos , Angiografia , Animais , Arteríolas/citologia , Arteríolas/diagnóstico por imagem , Células da Medula Óssea/citologia , Capilares/citologia , Capilares/diagnóstico por imagem , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Linhagem da Célula , Células Cultivadas , Endotélio Vascular/diagnóstico por imagem , Endotélio Vascular/ultraestrutura , Artéria Femoral/citologia , Artéria Femoral/diagnóstico por imagem , Fêmur/irrigação sanguínea , Fêmur/citologia , Isquemia/diagnóstico por imagem , Microscopia Eletrônica , Músculo Esquelético/irrigação sanguínea , Ratos , Fluxo Sanguíneo Regional/fisiologia
9.
Circulation ; 110(3): 317-23, 2004 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-15249508

RESUMO

BACKGROUND: Aldosterone has recently attracted considerable attention for its involvement in the pathophysiology of heart failure, in which apoptotic cell loss plays a critical role. This study examined whether aldosterone directly induces myocyte apoptosis via its specific receptors. METHODS AND RESULTS: Neonatal rat cardiac myocytes were exposed to aldosterone (10(-8) to 10(-5) mol/L). Nuclear staining with Hoechst 33258 showed that aldosterone induced myocyte apoptosis in a dose- and time-dependent fashion. Treatment of myocytes with 10(-5) mol/L aldosterone significantly increased the percentage of apoptosis (15.5+/-1.4%) compared with serum-deprived control (7.3+/-0.6%). Radio ligand binding assay revealed the existence of plasma membrane receptor with high affinity (K(d), 0.2 nmol/L) for aldosterone in cardiac myocytes but not in fibroblasts. Aldosterone rapidly (approximately 30 seconds) mobilized [Ca2+]i that was blocked by neomycin. Aldosterone induced dephosphorylation of the proapoptotic protein Bad, enhancement of mitochondrial permeability transition, decrease in mitochondrial membrane potential, and release of cytochrome c from the mitochondria into the cytosol with concomitant activation of caspase-3. These effects of aldosterone were inhibited by concurrent treatment with either an L-type Ca2+ channel antagonist, nifedipine, or inhibitors for the Ca2+-dependent phosphatase calcineurin, cyclosporin A and FK506. CONCLUSIONS: The present study demonstrates for the first time that the specific plasma membrane receptor (coupled with phospholipase C) for aldosterone is present on cardiac myocytes and that aldosterone accelerates the mitochondrial apoptotic pathway through activation of calcineurin and dephosphorylation of Bad, suggesting that the proapoptotic action of aldosterone may directly contribute to the progression of heart failure.


Assuntos
Aldosterona/farmacologia , Apoptose , Calcineurina/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Animais , Inibidores de Calcineurina , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Proteínas de Transporte/metabolismo , Caspase 3 , Caspases/metabolismo , Citocromos c/metabolismo , Inibidores Enzimáticos/farmacologia , Canais Iônicos/metabolismo , Potenciais da Membrana , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Superfície Celular/análise , Receptores de Mineralocorticoides/análise , Proteína de Morte Celular Associada a bcl
10.
J Gastroenterol Hepatol ; 19(5): 506-11, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15086593

RESUMO

BACKGROUND AND AIM: Attachment of Helicobacter pylori to gastric epithelial cells leads to the production of chemokines, such as interleukin-8 (IL-8), which in turn activate and recruit neutrophils to the site of infection. Lafutidine [(+/-)-2-(furfurylsulfinyl)-N-(4-(4-(piperidinomethyl)-2-pyridyl)oxy-(Z)-2-butenyl)acetamide] is a new type of antiulcer drug that possesses an antisecretory action as well as gastroprotective activity, independent of its antisecretory action. In the present study, we examined the effects of lafutidine on H. pylori-induced IL-8 release and H. pylori adhesion to MKN45 cells. METHODS: MKN45 cells were stimulated with H. pylori, tumor necrosis factor (TNF)-alpha, or IL-1beta, then IL-6 and IL-8 levels in the culture supernatants were determined with a specific enzyme-linked immunosorbent assay kit. RESULTS: Lafutidine significantly inhibited both the release of IL-8 induced by H. pylori and the adhesion of H. pylori to cells in a dose-dependent manner. These properties of lafutidine are unrelated to the blockade of histamine H(2)-receptors, because the same effects have not been observed with other H(2)-receptor antagonists, such as cimetidine and famotidine. Lafutidine also significantly inhibited H. pylori-induced IL-6 release. Both TNF-alpha and IL-1beta-induced IL-8 releases, conversely, were little affected by lafutidine up to a concentration of 10(-5) M. CONCLUSIONS: These results suggest that lafutidine inhibits IL-8 release by inhibiting H. pylori adherence to gastric epithelial cells, indicating a novel mechanism by which lafutidine protects against the mucosal inflammation associated with H. pylori infection.


Assuntos
Acetamidas/farmacologia , Antiulcerosos/farmacologia , Mucosa Gástrica/metabolismo , Helicobacter pylori/efeitos dos fármacos , Interleucina-8/biossíntese , Piperidinas/farmacologia , Piridinas/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Mucosa Gástrica/microbiologia , Humanos , Interleucina-1/farmacologia , Interleucina-6/biossíntese , Fator de Necrose Tumoral alfa/farmacologia
11.
J Pharmacol Sci ; 94(3): 233-9, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15037807

RESUMO

Helicobacter pylori (H. pylori) infection of gastric epithelial cells has been shown to induce interleukin (IL)-8 production, but the signal transduction mechanism leading to IL-8 production has not been clearly defined. Here, we investigate the role of protein kinase C (PKC) in the mechanism of induction of IL-8 release by H. pylori in human gastric epithelial cells. In MKN45 cells, H. pylori-induced IL-8 release was enhanced by treatment with PKC inhibitors (GF109203X and calphostin C) and PKC depletion, which completely inhibited PKC activity. Moreover, PKC inhibitors and PKC depletion increased extracellular signal-regulated kinase (ERK) activity and phosphorylation, but not calcium/calmodulin-dependent protein kinase II (CaMK II) activity, in response to H. pylori infection. PKC activated by H. pylori inhibited activation of ERK induced by H. pylori without affecting the CaMK II activity and negatively regulated IL-8 production in human gastric epithelial cells.


Assuntos
Células Epiteliais/microbiologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Mucosa Gástrica/microbiologia , Helicobacter pylori/metabolismo , Interleucina-8/antagonistas & inibidores , Proteína Quinase C/metabolismo , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Moléculas de Adesão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/genética , Humanos , Quinase I-kappa B , Indóis/metabolismo , Indóis/farmacologia , Interleucina-8/biossíntese , Interleucina-8/química , Maleimidas/metabolismo , Maleimidas/farmacologia , Métodos , Naftalenos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
12.
Circ Res ; 93(10): 980-9, 2003 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-14525810

RESUMO

Peripheral blood (PB)-derived CD14+ monocytes were shown to transdifferentiate into endothelial cell (EC) lineage cells and contribute to neovascularization. We investigated whether bone marrow (BM)- or PB-derived CD34-/CD14+ cells are involved in reendothelialization after carotid balloon injury. Although neither hematopoietic nor mesenchymal stem cells were included in human BM-derived CD34-/CD14+ monocyte lineage cells (BM-MLCs), they expressed EC-specific markers (Tie2, CD31, VE-cadherin, and endoglin) to an extent identical to mature ECs. When BM-MLCs were cultured with vascular endothelial growth factors, hematopoietic markers were drastically decreased and new EC-specific markers (Flk and CD34) were induced. BM-MLCs were intra-arterially transplanted into balloon-injured arteries of athymic nude rats. When BM-MLCs were activated by monocyte chemoattractant protein-1 (MCP-1) in vivo or in vitro, they adhered onto injured endothelium, differentiated into EC-like cells by losing hematopoietic markers, and inhibited neointimal hyperplasia. Ability to prevent neointimal hyperplasia was more efficient than that of BM-derived CD34+ cells. MCP-dependent adhesion was not observed in PB-derived CD34-/CD14+ monocytes. Regenerated endothelium exhibited a cobblestone appearance, blocked extravasation of dye, and induced NO-dependent vasorelaxation. Basal adhesive activities on HUVECs under laminar flow and beta1-integrin expression (basal and active forms) were significantly increased in BM-MLCs compared with PB-derived monocytes. MCP-1 markedly enhanced adhesive activity of BM-MLCs (2.8-fold) on HUVECs by activating beta1-integrin conformation. Thus, BM-MLCs can function as EC progenitors that are more potent than CD34+ cells and acquire the ability to adhere on injured endothelium in a MCP-1-dependent manner, leading to reendothelialization associated with inhibition of intimal hyperplasia. This will open a novel window to MCP-1-mediated biological actions and vascular regeneration strategies by cell therapy.


Assuntos
Células da Medula Óssea/citologia , Quimiocina CCL2/metabolismo , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Monócitos/fisiologia , Células-Tronco/fisiologia , Angioplastia com Balão/efeitos adversos , Animais , Antígenos de Diferenciação/biossíntese , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Adesão Celular/fisiologia , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/farmacologia , Endotélio Vascular/imunologia , Endotélio Vascular/lesões , Técnicas de Transferência de Genes , Humanos , Monócitos/citologia , Monócitos/imunologia , Ratos , Ratos Nus , Células-Tronco/citologia
13.
Hypertension ; 41(1): 156-62, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12511546

RESUMO

It remains undetermined whether continuous endothelial nitric oxide (NO) overexpression exerts angiogenic action. We surgically induced hindlimb ischemia in transgenic mice overexpressing endothelial NO synthase in the endothelium (eNOS-Tg) and studied neocapillary formation, ischemia-induced vascular endothelial growth factor (VEGF) expression, cGMP accumulation, and Akt/PKB signaling. Laser Doppler imaging revealed a markedly increased recovery of blood perfusion in ischemic limbs of eNOS-Tg mice (44% increase) compared with that in wild-type mice. Angiography showed a marked increase in basal and ischemia-induced collateral vessel formation in eNOS-Tg mice. Basal capillary densities and tissue cGMP levels were increased in eNOS-Tg mice (1.8-fold and 1.6-fold versus wild-type mice, respectively). Ischemia-induced neocapillary formation and cGMP accumulation were markedly increased in eNOS-Tg mice (3.6-fold and 4.1-fold versus preischemia levels, respectively), whereas those in wild-type mice were much less (1.8-fold and 1.5-fold, respectively). Basal and time-dependent VEGF expression in ischemic muscles did not differ between eNOS-Tg and wild-type mice. Basal and VEGF-mediated Akt phosphorylation in aortas was similar between eNOS-Tg and wild-type mice. Aortic basal eNOS expression was increased 3.3-fold, and VEGF-mediated eNOS phosphorylation was markedly induced in aortas of eNOS-Tg compared with preischemia levels (4.2-fold), whereas much smaller changes were observed in wild-type mice (1.8-fold increase). Our study demonstrates that overexpression of eNOS protein causes a marked increase in neocapillary formation in response to tissue ischemia without affecting ischemia-induced VEGF expression or VEGF-mediated Akt phosphorylation.


Assuntos
Isquemia/sangue , Neovascularização Fisiológica , Óxido Nítrico Sintase/genética , Proteínas Serina-Treonina Quinases , Angiografia , Animais , Circulação Sanguínea , Capilares/crescimento & desenvolvimento , Circulação Colateral , GMP Cíclico/análise , Fatores de Crescimento Endotelial/genética , Fatores de Crescimento Endotelial/farmacologia , Membro Posterior/irrigação sanguínea , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Isquemia/enzimologia , Isquemia/metabolismo , Fluxometria por Laser-Doppler , Linfocinas/genética , Linfocinas/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , RNA Mensageiro/biossíntese , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
14.
Circulation ; 106(15): 2019-25, 2002 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-12370229

RESUMO

BACKGROUND: Peripheral blood mononuclear cells (PBMNCs), platelets, and polymorphonuclear leukocytes (PMNs) contain various angiogenic factors and cytokines. METHODS AND RESULTS: Unilateral hindlimb ischemia was surgically induced in athymic nude rats, and fluorescence-labeled human blood cells (PBMNCs [10(7) cells]+platelets [10(9)] or PBMNCs [10(7)]+platelets [10(9)]+PMNs [10(7)]) were intramuscularly implanted into the ischemic limbs. Laser Doppler imaging revealed markedly increased blood perfusion in PBMNC+platelet-implanted limbs (44% increase, P<0.001) compared with control implantation of human umbilical vein vascular endothelial cells. The addition of PMNs to PBMNCs+platelets attenuated blood perfusion (27% decrease, P<0.01). Neocapillary densities were increased by implantation of PBMNCs+platelets or platelets alone (3.5-fold and 2.4-fold, respectively; P<0.001), whereas PMNs inhibited (32%, P<0.05) PBMNC+ platelet-mediated capillary formation. There was no incorporation of implanted PBMNCs into neocapillaries, whereas PBMNCs and platelets accumulated around arterioles after implantation. Cellular extract from PBMNCs+platelets, in which vascular endothelial growth factor (VEGF), basic fibroblast growth factor, platelet-derived growth factor-AB, and transforming growth factor-beta were detected, markedly stimulated tubule formation of human umbilical vein vascular endothelial cells. Anti-VEGF neutralizing antibody markedly inhibited tubule formation and in vivo vessel formation. Neutrophil elastase inhibitor blocked the antiangiogenic action of PMNs, whereas inhibitors of oxygen metabolites had no effect. CONCLUSIONS: This study demonstrated that implantation of PBMNCs and platelets into ischemic limbs effectively induces collateral vessel formation by supplying angiogenic factors (mainly VEGF) and cytokines, suggesting that this cell therapy is useful as a novel strategy for therapeutic angiogenesis.


Assuntos
Isquemia/cirurgia , Leucócitos Mononucleares/transplante , Neovascularização Fisiológica , Transfusão de Plaquetas , Indutores da Angiogênese/fisiologia , Angiografia , Animais , Circulação Sanguínea , Plaquetas/fisiologia , Vasos Sanguíneos/química , Vasos Sanguíneos/crescimento & desenvolvimento , Transplante de Medula Óssea , Movimento Celular , Fatores de Crescimento Endotelial/fisiologia , Endotélio Vascular/crescimento & desenvolvimento , Extremidades/irrigação sanguínea , Fator VIII/análise , Fator VIII/imunologia , Humanos , Imuno-Histoquímica , Isquemia/diagnóstico por imagem , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Leucócitos Mononucleares/fisiologia , Linfocinas/fisiologia , Neutrófilos/fisiologia , Neutrófilos/transplante , Ratos , Ratos Nus , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
15.
Biochem Pharmacol ; 64(1): 21-30, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12106602

RESUMO

Infecting gastric epithelial cells with Helicobacter pylori (H. pylori) has been shown to induce interleukin-8 (IL-8) production, but the signal transduction mechanism leading to IL-8 production is not defined clearly. In the present study, we investigated the molecular mechanism responsible for H. pylori-induced IL-8 release in human gastric epithelial cells. IL-8 levels in culture supernatants were determined by an enzyme linked-immunosorbent assay. Extracellular signal-regulated kinase (ERK) activity was tested using an in vitro kinase assay, which measured the incorporation of [gamma-33P]ATP into a synthetic peptide that is a specific ERK substrate. ERK phosphorylation and IkappaBalpha degradation by H. pylori infection were assessed by western blotting. In MKN45 cells, H. pylori-induced IL-8 release in a time-dependent manner. This IL-8 release was abolished by treatment with intracellular Ca2+ chelators (BAPTA-AM and TMB-8) but not by EGTA or nifedipine. The Ca2+ ionophore A23187 also induced IL-8 release to an extent similar to that of H. pylori infection. Calmodulin inhibitors (W7 and calmidazolium) and tyrosine kinase inhibitors (genistein and ST638) completely blocked IL-8 release by H. pylori and A23187. PD98059, an ERK pathway inhibitor, completely abolished H. pylori-induced IL-8 release. Moreover, BAPTA-AM, calmidazolium, and genistein, but not nifedipine, suppressed the ERK activation induced by H. pylori infection. PD98059 as well as MG132, an NF-kappaB pathway inhibitor, blocked both IL-8 production and degradation of IkappaBalpha induced by H. pylori infection, whereas only PD98059 inhibited ERK activity in response to H. pylori. There was no significant difference between IL-8 production induced by the cagA positive wild-type strain and the cagA negative isogenic mutant strain of H. pylori; therefore, CagA is not involved in the IL-8 production pathway. H. pylori-induced IL-8 production is dominantly regulated by Ca2+/calmodulin signaling, and ERK plays an important role in signal transmission for the efficient activation of H. pylori-induced NF-kappaB activity, resulting in IL-8 production.


Assuntos
Ácido Egtázico/análogos & derivados , Helicobacter pylori/metabolismo , Interleucina-8/biossíntese , Mucosa Intestinal/microbiologia , Transdução de Sinais/fisiologia , Aderência Bacteriana/efeitos dos fármacos , Cálcio/metabolismo , Calmodulina/metabolismo , Células Cultivadas , Ácido Egtázico/farmacologia , Humanos , Interleucina-1/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
16.
Br J Pharmacol ; 135(6): 1487-94, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11906962

RESUMO

1. Capsaicin sensitive afferent nerves play an important role in gastric mucosal defensive mechanisms. Capsaicin stimulates afferent nerves and enhances the release of calcitonin gene-related peptide (CGRP), which seems to be the predominant neurotransmitter of spinal afferents in the rat stomach, exerting many pharmacological effects by a direct mechanism or indirectly through second messengers such as nitric oxide (NO). 2. Lafutidine is a new type of anti-ulcer drug, possessing both an antisecretory effect, exerted via histamine H(2) receptor blockade, and gastroprotective activities. Studies with certain antagonists or chemical deafferentation techniques suggest the gastroprotective actions of lafutidine to be mediated by capsaicin sensitive afferent nerves, but this is an assumption based on indirect techniques. In order to explain the direct relation of lafutidine to afferent nerves, we conducted the following studies. 3. We determined CGRP and NO release from rat stomach and specific [(3)H]-resiniferatoxin (RTX) binding to gastric vanilloid receptor subtype 1 (VR1), which binds capsaicin, using EIA, a microdialysis system and a radioreceptor assay, respectively. 4. Lafutidine enhanced both CGRP and NO release from the rat stomach induced by a submaximal dose of capsaicin, but had no effect on specific [(3)H]-RTX and capsaicin binding to VR1. 5. In conclusion, our findings demonstrate that lafutidine modulates the activity of capsaicin sensitive afferent nerves in the rat stomach, which may be a key mechanism involved in its gastroprotective action.


Assuntos
Acetamidas/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Piperidinas/farmacologia , Piridinas/farmacologia , Estômago/efeitos dos fármacos , Animais , Antiulcerosos/farmacologia , Capsaicina/farmacologia , Relação Dose-Resposta a Droga , Mucosa Gástrica/metabolismo , Masculino , Neurônios Aferentes/metabolismo , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...