Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Funct Biomater ; 14(12)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38132820

RESUMO

Therapeutic interventions for vascular diseases aim at achieving long-term patency by controlling vascular remodeling. The extracellular matrix (ECM) of the vessel wall plays a crucial role in regulating this process. This study introduces a novel photochemical treatment known as Natural Vascular Scaffolding, utilizing a 4-amino substituted 1,8-naphthimide (10-8-10 Dimer) and 450 nm light. This treatment induces structural changes in the ECM by forming covalent bonds between amino acids in ECM fibers without harming vascular cell survival, as evidenced by our results. To further investigate the mechanism of this treatment, porcine carotid artery segments were exposed to 10-8-10 Dimer and light activation. Subsequent experiments subjected these segments to enzymatic degradation through elastase or collagenase treatment and were analyzed using digital image analysis software (MIPAR) after histological processing. The results demonstrated significant preservation of collagen and elastin structures in the photochemically treated vascular wall, compared to controls. This suggests that photochemical treatment can effectively modulate vascular remodeling by enhancing the resistance of the ECM scaffold to degradation. This approach shows promise in scenarios where vascular segments experience significant hemodynamic fluctuations as it reinforces vascular wall integrity and preserves lumen patency. This can be valuable in treating veins prior to fistula creation and grafting or managing arterial aneurysm expansion.

2.
Int J Mol Sci ; 24(8)2023 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-37108733

RESUMO

Rates of arteriovenous fistula maturation failure are still high, especially when suboptimal size veins are used. During successful maturation, the vein undergoes lumen dilatation and medial thickening, adapting to the increased hemodynamic forces. The vascular extracellular matrix plays an important role in regulating these adaptive changes and may be a target for promoting fistula maturation. In this study, we tested whether a device-enabled photochemical treatment of the vein prior to fistula creation facilitates maturation. Sheep cephalic veins were treated using a balloon catheter coated by a photoactivatable molecule (10-8-10 Dimer) and carrying an internal light fiber. As a result of the photochemical reaction, new covalent bonds were created during light activation among oxidizable amino acids of the vein wall matrix proteins. The treated vein lumen diameter and media area became significantly larger than the contralateral control fistula vein at 1 week (p = 0.035 and p = 0.034, respectively). There was also a higher percentage of proliferating smooth muscle cells in the treated veins than in the control veins (p = 0.029), without noticeable intimal hyperplasia. To prepare for the clinical testing of this treatment, we performed balloon over-dilatation of isolated human veins and found that veins can tolerate up to 66% overstretch without notable histological damage.


Assuntos
Fístula Arteriovenosa , Derivação Arteriovenosa Cirúrgica , Humanos , Animais , Ovinos , Diálise Renal , Veias/patologia , Dilatação , Fístula Arteriovenosa/patologia , Resultado do Tratamento
3.
Int J Mol Sci ; 23(2)2022 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-35054866

RESUMO

The development of bioscaffolds for cardiovascular medical applications, such as peripheral artery disease (PAD), remains to be a challenge for tissue engineering. PAD is an increasingly common and serious cardiovascular illness characterized by progressive atherosclerotic stenosis, resulting in decreased blood perfusion to the lower extremities. Percutaneous transluminal angioplasty and stent placement are routinely performed on these patients with suboptimal outcomes. Natural Vascular Scaffolding (NVS) is a novel treatment in the development for PAD, which offers an alternative to stenting by building on the natural structural constituents in the extracellular matrix (ECM) of the blood vessel wall. During NVS treatment, blood vessels are exposed to a photoactivatable small molecule (10-8-10 Dimer) delivered locally to the vessel wall via an angioplasty balloon. When activated with 450 nm wavelength light, this therapy induces the formation of covalent protein-protein crosslinks of the ECM proteins by a photochemical mechanism, creating a natural scaffold. This therapy has the potential to reduce the need for stent placement by maintaining a larger diameter post-angioplasty and minimizing elastic recoil. Experiments were conducted to elucidate the mechanism of action of NVS, including the molecular mechanism of light activation and the impact of NVS on the ECM.


Assuntos
Prótese Vascular , Matriz Extracelular/efeitos da radiação , Alicerces Teciduais/química , Angioplastia com Balão , Animais , Artérias/fisiologia , Fenômenos Biomecânicos , Reagentes de Ligações Cruzadas/química , Dimerização , Hipercolesterolemia/diagnóstico por imagem , Hipercolesterolemia/fisiopatologia , Hipercolesterolemia/terapia , Luz , Peptídeos/química , Suínos
4.
Chembiochem ; 22(17): 2703-2710, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34161648

RESUMO

The oxidation of proteins generates reactive amino acid (AA) residue intermediates, leading to protein modification and cross-linking. Aerobic studies with peptides and photosensitizers allow for the controlled generation of reactive oxygen species (ROS) and reactive AA residue intermediates, providing mechanistic insights as to how natural protein modifications form. Such studies have inspired the development of abiotic methods for protein modification and crosslinking, including applications of biomedical importance. Dityrosine linkages derived from oxidation at tyrosine (Tyr) residues represent one of the more well-understood oxidation-induced modifications. Here we demonstrate an aerobic, visible light-dependent oxidation reaction of Tyr-containing substrates promoted by a water-soluble 4-amino-1,8-naphthalimide-based photosensitizer. The developed procedure converts Tyr-containing substrates into o,o'-Tyr-Tyr linked dimers. The regioselectively formed o,o'-Tyr-Tyr linkage is consistent with dimeric standards prepared using a known enzymatic method. A crossover study with two peptides provides a statistical mixture of three distinct o,o'-Tyr-Tyr linked dimers, supporting a mechanism that involves Tyr residue oxidation followed by intermolecular combination.


Assuntos
1-Naftilamina/análogos & derivados , Naftalimidas/química , Quinolonas/química , Tirosina/química , 1-Naftilamina/química , Biocatálise , Dimerização , Peroxidase do Rábano Silvestre/metabolismo , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Luz , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Oxirredução , Fármacos Fotossensibilizantes/química , Teoria Quântica , Estereoisomerismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Água/química
5.
Front Bioeng Biotechnol ; 9: 622617, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33681159

RESUMO

Following creation, an arteriovenous fistula (AVF) must mature (i.e., enlarge lumen to allow high blood flow) before being used for hemodialysis. AVF maturation failure rates are high, and currently, there are no effective therapy to treat this problem. The maturation process is likely affected by the integrity of the vascular extracellular matrix (ECM). Natural Vascular Scaffolding (NVS) Therapy is a new technology that interlinks collagen and elastin via photoactivation of a locally delivered small molecule (4-amino-1,8-naphtalamide). We hypothesized that NVS Therapy may improve AVF remodeling by preserving ECM integrity. AVFs were created in Wistar male rats by connecting the femoral vein (end) to femoral artery (side) in the same limb. Immediately after blood flow was restored to dilate the femoral vein by arterial pressure, a 10 µl-drop of the NVS compound (2 mg/ml) was placed on the anastomosis perivascularly. Following 5-min incubation, the NVS treated area was exposed to 1-min illumination by 450-nm light. The control group received 10 µl-drop of phosphate buffered saline (PBS) and the same light activation. The skin was closed, and rats were euthanized 4 weeks (n = 6-9 per group) post-AVF creation for histology, morphometry, immunohistochemistry (IHC), and multiphoton microscopy for second-harmonic-generation evaluation of collagen fibers. The vascular thickness was similar in both groups. The AVF vein's open lumen area and % open lumen area in NVS-treated rats were significantly larger than in PBS-treated rats (4.2-fold p = 0.014 and 2-fold p = 0.009, respectively). The inflammatory markers IL-6 and MMP-9 in the AVF walls were significantly decreased in the NVS group than the PBS group. Collagen fibers in the vascular wall trended toward perpendicular alignment to the lumen circumference in the NVS-treated AVFs, with more defined shape but less area than in the PBS-treated AVFs. These results indicate that the NVS Therapy exerted changes in collagen, which may influence AVF maturation. Rats tolerated the NVS treatment well, and the lack of cell death by the treatment was confirmed in cell culture experiments. These results suggest that NVS treatment is safe and may have therapeutic potential by facilitating lumen expansion to enhanced AVF maturation in patients.

6.
J Phys Chem A ; 125(11): 2294-2307, 2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33724853

RESUMO

The solvent-dependent photophysics of two 4-amino-substituted 1,8-naphthalene imides (AIs) were studied using fluorescence spectroscopy and laser flash photolysis. The compounds were functionalized with water-soluble 2,2'(ethylenedioxy) diethylamine groups, yielding a monomer (AI3) and a dimer (AI4). The radiative and nonradiative singlet-state deactivation processes of AI3 and AI4 were quantified in 10 solvents and at different pH values. The fluorescence quantum yield for the AI4 dimer in water was more than 100× lower than in other solvents, or for the monomeric AI3. The enhanced nonradiative decay of aqueous solutions of dimeric AI4 is accompanied by biexponential decay kinetics, suggesting equilibration with a dark excited state. An oxygen-quenchable triplet state (T1) of AI3 was produced upon 416 nm excitation in both water and n-octanol. In water, the T1 state evolved into a long-lived transient that was unreactive toward oxygen or several electron donors. This species was not observed in n-octanol. The transient observed upon 416 nm excitation of AI4 in water was extremely weak. However, production of T1 in both AI3 and AI4 was evidenced by the photoinduced electron transfer to methyl viologen, albeit in low quantum yield (0.0503 and 0.00778 for AI3 and AI4, respectively). The photophysics and reactivity are consistent with the production of an intramolecular charge transfer (ICT) state that is stabilized in water. Significantly, this stabilization enhances nonradiative decay pathways, particularly in the AI4 dimer. The results indicate that the photochemistry of these compounds can be environmentally mediated, switching from radical- to triplet-initiated processes.

7.
J Cardiovasc Transl Res ; 14(3): 441-448, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32748207

RESUMO

In this study, we investigated natural vascular scaffolding (NVS) treatment on vascular functionality using freshly isolated human popliteal arteries in vitro. Arteries were exposed to intraluminal NVS treatment consisting of a compound (4 amino-1,8-naphthalimide) photoactivated by a 450-nm light-emitting light fiber placed inside the artery. This procedure results in covalent linking between the extracellular matrix proteins to achieve a larger vessel diameter post-angioplasty and minimizing elastic recoil. Immediately following NVS treatment, rings were cut from the treated arteries and mounted in organ baths for contractility testing in response to U46619 and sodium nitroprusside. We also investigated the effect of NVS treatment on IL-6 cytokine release from vascular rings following a 4-h organoculture post-NVS treatment. Based on our results, we conclude that exposure of the vessels to NVS treatment does not adversely affect the contractile responsiveness of the vascular smooth muscle and exerts no pro-inflammatory effect. Graphical abstract.


Assuntos
1-Naftilamina/análogos & derivados , Reagentes de Ligações Cruzadas/farmacologia , Proteínas da Matriz Extracelular/metabolismo , Matriz Extracelular/efeitos dos fármacos , Naftalimidas/farmacologia , Artéria Poplítea/efeitos dos fármacos , Quinolonas/farmacologia , 1-Naftilamina/farmacologia , 1-Naftilamina/efeitos da radiação , Idoso , Idoso de 80 Anos ou mais , Reagentes de Ligações Cruzadas/efeitos da radiação , Elasticidade , Matriz Extracelular/metabolismo , Humanos , Interleucina-6/metabolismo , Pessoa de Meia-Idade , Naftalimidas/efeitos da radiação , Processos Fotoquímicos , Artéria Poplítea/metabolismo , Quinolonas/efeitos da radiação , Técnicas de Cultura de Tecidos , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia
8.
Exp Hematol ; 60: 73-82.e3, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29337222

RESUMO

Vaso-occlusive crisis (VOC) is the most common and debilitating complication of sickle cell disease (SCD); recurrent episodes cause organ damage and contribute to early mortality. Plasma placental growth factor (PlGF) levels are elevated in SCD and can further increase under hypoxic conditions in SCD mice. Treatment with a PlGF-neutralizing antibody (anti-PlGF Ab) in SCD mice reduced levels of monocyte chemoattractant protein-3, eotaxin, macrophage colony-stimulating factor, and plasminogen activator inhibitor-1 significantly, and of macrophage-derived chemokine and macrophage inflammatory protein-3ß moderately; this may contribute to inhibition of leukocyte recruitment, activation, and thrombosis. In subsequent experiments, anti-PlGF Ab treatment significantly reduced plasma lactate dehydrogenase levels, indicating possible reduction in cellular destruction and/or hemolysis. Histopathology studies revealed decreased incidence and severity of congestion in the lungs and spleen with repeated anti-PlGF Ab treatment. Furthermore, anti-PlGF Ab significantly reduced vaso-occlusion events under hypoxic conditions in a modified dorsal skinfold chamber model in SCD mice. Therefore, elevated PlGF levels may contribute to recruitment and activation of leukocytes. This can subsequently lead to increased pathology of affected organs in addition to mediating acute hypoxia/reoxygenation-triggered vaso-occlusion under SCD conditions. Thus, targeting PlGF may offer a therapeutic approach to reduce acute VOC and possibly alleviate long-term vascular complications in patients with SCD.


Assuntos
Anemia Falciforme/tratamento farmacológico , Anticorpos Neutralizantes/farmacologia , Proteínas/antagonistas & inibidores , Doenças Vasculares/tratamento farmacológico , Anemia Falciforme/sangue , Anemia Falciforme/complicações , Animais , Modelos Animais de Doenças , Humanos , Proteínas de Membrana , Camundongos , Proteínas/metabolismo , Doenças Vasculares/sangue , Doenças Vasculares/etiologia
9.
Handb Exp Pharmacol ; 232: 59-70, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26552401

RESUMO

Drug development is a resource-intensive process requiring significant financial and time investment. Preclinical target validation studies and in vivo testing of the therapeutic molecules in clinically relevant disease models can accelerate and significantly de-risk later stage clinical development. In this chapter, we will focus on (1) in vivo animal models and (2) pharmacological tools for target validation.


Assuntos
Descoberta de Drogas , Terapia de Alvo Molecular , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Camundongos
10.
J Pharmacol Exp Ther ; 343(3): 547-55, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22918043

RESUMO

{4-[(5,6-Diphenylpyrazin-2-yl)(isopropyl)amino]butoxy}acetic acid (ACT-333679) is the main metabolite of the selective prostacyclin (PGI(2)) receptor (IP receptor) agonist selexipag. The goal of this study was to determine the influence of IP receptor selectivity on the vasorelaxant efficacy of ACT-333679 and the PGI(2) analog treprostinil in pulmonary artery under conditions associated with pulmonary arterial hypertension (PAH). Selexipag and ACT-333679 evoked full relaxation of pulmonary artery from control and monocrotaline (MCT)-PAH rats, and ACT-333679 relaxed normal pulmonary artery contracted with either endothelin-1 (ET-1) or phenylephrine. In contrast, treprostinil evoked weaker relaxation than ACT-333679 of control pulmonary artery and failed to induce relaxation of pulmonary artery from MCT-PAH rats. Treprostinil did not evoke relaxation of normal pulmonary artery contracted with either ET-1 or phenylephrine. Expression of prostaglandin E(3) (EP(3)) receptor mRNA was increased in pulmonary artery from MCT-PAH rats. In contraction experiments, the selective EP(3) receptor agonist sulprostone evoked significantly greater contraction of pulmonary artery from MCT-PAH rats compared with control rats. The presence of a threshold concentration of ET-1 significantly augmented the contractile response to sulprostone in normal pulmonary artery. ACT-333679 did not evoke direct contraction of rat pulmonary artery, whereas treprostinil evoked concentration-dependent contraction that was inhibited by the EP(3) receptor antagonist (2E)-3-(3',4'-dichlorobiphenyl-2-yl)-N-(2-thienylsulfonyl)acrylamide. Antagonism of EP(3) receptors also revealed a relaxant response to treprostinil in normal pulmonary artery contracted with ET-1. These data demonstrate that the relaxant efficacy of the selective IP receptor agonist selexipag and its metabolite ACT-333679 is not modified under conditions associated with PAH, whereas relaxation to treprostinil may be limited in the presence of mediators of disease.


Assuntos
Acetamidas/farmacologia , Acetatos/farmacologia , Epoprostenol/análogos & derivados , Artéria Pulmonar/efeitos dos fármacos , Pirazinas/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Alprostadil/análogos & derivados , Alprostadil/metabolismo , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epoprostenol/farmacologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Técnicas In Vitro , Masculino , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Ratos , Ratos Wistar , Receptores de Epoprostenol/agonistas , Vasoconstritores/farmacologia
11.
Nephron Extra ; 2(1): 205-18, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22811694

RESUMO

Amlodipine reduces blood pressure; however, its effect in the diabetic kidney irrespective of its blood pressure-lowering effects is unclear. This study examined the effects of amlodipine (0, 5, 10 and 20 mg/kg; D(A0), D(A5), D(A10) and D(A20), respectively) for 12 weeks on renal functional and structural changes in the streptozotocin-induced diabetic rat, a nonhypertensive model of diabetes-associated hyperfiltration. Compared with nondiabetic rats, diabetes (D) was associated with increased urine albumin excretion (UAE, 12.6 ± 3.40 vs. 3.73 ± 1.14 mg/day), glomerular filtration rate (2.17 ± 0.09 vs. 1.64 ± 0.12 ml/min/g kidney weight), glomerulosclerosis (0.21 ± 0.03 vs. 0.05 ± 0.01 AU) and infiltration of inflammatory cells (18.5 ± 2.78 vs. 6.92 ± 0.70 cells/cm(2)), but did not affect mean arterial pressure (MAP, 110 ± 4.70 vs. 109 ± 5.33 mm Hg). While D(A20) abolished glomerular hyperfiltration (1.49 ± 0.05 ml/min/g kidney weight) and inflammatory cell abundance (6.0 ± 0.79 cells/cm(2)), it exacerbated UAE (43.5 ± 8.49 mg/day) and increased MAP (132 ± 3.76 mm Hg), but had no effect on renal pathology. These data suggest that amlodipine reduces renal inflammation and abolished glomerular hyperfiltration, but increases blood pressure and exacerbates albuminuria in the rat model of normotensive diabetic kidney disease. We conclude that amlodipine may have limited renoprotective effects in the face of hyperfiltration and absence of elevated blood pressure.

12.
Hypertension ; 57(4): 795-801, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21357272

RESUMO

The renin-angiotensin system is a well-known regulator of blood pressure and plays an important role in the pathogenesis of cardiovascular disease and renal damage. Genetic factors, including single nucleotide polymorphisms and sex, are increasingly recognized as potential risk factors for the development of cardiovascular disease. Double transgenic rats (dTGRs), harboring human renin and angiotensinogen genes, were used in this study to investigate potential sex differences influencing renal function and renal gene expression. dTGR males and females had comparable increases in blood pressure, whereas body weight, albuminuria/proteinuria, and urine flow rate were higher in males. At 8 weeks of age, renal plasma flow and glomerular filtration rate were proportionally lower in males, and renal vascular resistance tended to be higher. Males developed more severe tubulointerstitial and vascular lesions. By the end of week 8, 40%of the males but none of the females had died. Genome expression studies were performed with RNA from kidneys of 7-week-old male and female dTGRs and control rats to further investigate the sex-related differences on a molecular level. Forty-five genes showed sex-dependent expression patterns in dTGRs that were significantly different compared to controls. Cathepsin L, one of the genes differentially expressed between the sexes, was also shown to be strongly associated with the degree of renal injury. In dTGRs, urinary cathepsin L at week 7 was higher in males (nanograms per 24 hours: male, 512±163; female, 132±70). These results reveal a potential new biomarker for the personalized diagnosis and management of chronic kidney disease.


Assuntos
Angiotensinogênio/genética , Catepsina L/genética , Rim/metabolismo , Renina/genética , Caracteres Sexuais , Análise de Variância , Angiotensinogênio/metabolismo , Animais , Biomarcadores/metabolismo , Pressão Sanguínea/fisiologia , Catepsina L/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Taxa de Filtração Glomerular/fisiologia , Humanos , Imuno-Histoquímica , Rim/patologia , Rim/fisiopatologia , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Circulação Renal/fisiologia , Renina/metabolismo , Sistema Renina-Angiotensina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Resistência Vascular/fisiologia
13.
Lab Invest ; 90(11): 1573-81, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20585312

RESUMO

The role of angiotensin II (Ang II) in promoting cardiac hypertrophy is well known; however, its role in a spontaneous model of hypertrophy in mice lacking the protein caveolin-1 (Cav-1 KO) has not been explored. In this study, WT and Cav-1 KO mice were treated with angiotensin receptor blocker (ARB), telmisartan (Telm), and cardiac function was assessed by echocardiography. Treatment of Cav-1 KO mice with Telm significantly improved cardiac function compared with age-matched vehicle-treated Cav-1 KO mice, whereas Telm did not affect cardiac function in WT mice. Both left ventricular (LV) weight to body weight ratios and LV to tibial length ratios were also reverted by Telm in Cav-1 KO but not in WT mice. LV hypertrophy was associated with increased expression of natriuretic peptides A and B, ß-myosin heavy chain and TGF-ß, and Telm treatment normalized the expression of these genes. Telm reduced the expression of collagen genes (Col1A and Col3A) and associated perivascular fibrosis in intramyocardial vessels in Cav-1 KO mice. In conclusion, Telm treatment reduces indexes of cardiac hypertrophy in this unique genetic model of spontaneous LV hypertrophy.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Benzimidazóis/uso terapêutico , Benzoatos/uso terapêutico , Caveolina 1/deficiência , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Angiotensina II/sangue , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Tipo 2 de Angiotensina/fisiologia , Telmisartan
14.
Nitric Oxide ; 16(4): 403-12, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17452114

RESUMO

Endothelial nitric oxide synthase (ecNOS) derived nitric oxide (NO) is a key contributor to the angiogenic process. By augmenting angiogenesis NO could potentially promote tumor progression. The object of this study was to determine how knockdown of ecNOS affects endothelial NO production and the angiogenic response in endothelial cells. EOMA cells derived from a spontaneously arising murine hemangioendothelioma were genetically manipulated to stably express siRNA targeting ecNOS. Knockdown of ecNOS in different stably transfected EOMA cell lines was demonstrated by quantitative RT-PCR, Western blot and ecNOS specific ELISA. An EOMA cell line with near complete knockdown of ecNOS exhibited dramatically altered morphology and changes in the expression of mRNAs encoding proteins involved in angiogenesis. This cell line exhibited a 4-fold increase in proliferation in vitro, altered tube formation in matrigel and formed tumors in mice more rapidly than the parental cells. In contrast, a cell line in which ecNOS protein levels were reduced only 5-fold did not show changes in proliferation rate, tube formation or tumor growth. These results suggest that ecNOS derived nitric oxide reduces the growth of hemangioendothelioma derived tumors, and underscore the importance of careful consideration of the tumor type when selecting modulation of nitric oxide signaling as a treatment strategy.


Assuntos
Proliferação de Células , Endotélio Vascular/enzimologia , Hemangioendotelioma/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , RNA Interferente Pequeno/biossíntese , Animais , Linhagem Celular Tumoral , Células Cultivadas , Regulação para Baixo , Endotélio Vascular/química , Regulação da Expressão Gênica , Vetores Genéticos/genética , Hemangioendotelioma/química , Hemangioendotelioma/patologia , Lentivirus/genética , Camundongos , Transplante de Neoplasias , Óxido Nítrico Sintase Tipo III/genética , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/genética
15.
J Biol Chem ; 281(40): 29568-74, 2006 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-16880208

RESUMO

Cellular senescence prevents the proliferation of cells at risk for neoplastic transformation. Nonetheless, the senescence response is thought to be antagonistically pleiotropic and thus contribute to aging phenotypes, including, ironically, late life cancers. The cancer-promoting activity of senescent cells is likely due to secreted molecules, the identity of which remains largely unknown. Here, we have shown that senescent fibroblasts, much more than presenescent fibroblasts, stimulate tumor vascularization in mice. Weakly malignant epithelial cells co-injected with senescent fibroblasts had larger and greater numbers of blood vessels compared with controls. Accordingly, increased vascular endothelial growth factor (VEGF) expression was a frequent characteristic of senescent human and mouse fibroblasts in culture. Importantly, conditioned medium from senescent fibroblasts, more than medium from presenescent cells, stimulates cultured human umbilical vein endothelial cells to invade a basement membrane, a hallmark of angiogenesis. Increased VEGF expression was specific to the senescent phenotype and increased whether senescence was induced by replicative exhaustion, overexpression of p16(Ink4a), or overexpression of oncogenic RAS. The senescence-dependent increase in VEGF production was accompanied by very little increase in hypoxic-inducible (transcription) factor 1 alpha protein levels, and hypoxia further induced VEGF in senescent cells. This result suggests the rise in VEGF expression at senescence is not a hypoxic response. Our findings may in part explain why senescent cells stimulate tumorigenesis in vivo and support the idea that senescent cells may facilitate age-associated cancer development by secreting factors that promote malignant progression.


Assuntos
Senescência Celular/fisiologia , Fibroblastos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Cultivadas , Meios de Cultivo Condicionados , Humanos , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/etiologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia , Neovascularização Patológica/etiologia , Neovascularização Patológica/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese
16.
J Cardiovasc Pharmacol ; 47(4): 587-93, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16680073

RESUMO

Morbidity and mortality of peripheral arterial occlusive disease significantly increases with age, often exhibiting more severe disease pathology and decreased treatment effectiveness. Therapeutic angiogenesis with angiogenic growth factors may represent a valuable treatment option for the severely ill, older adult patient population. Aging is considered an independent cardiovascular risk factor, but pathomechanistically it is not well understood. Diminished endothelial nitric oxide (EDNO) production has been considered as a major contributor to the aging process. To investigate the effect of age on postischemic revascularization independent of changes in EDNO, we used endothelial nitric oxide synthase-deficient (ecNOS-KO) mice. We found an age-dependent acceleration in ischemic injury following unilateral femoral artery ligation in these animals compared to C57BL/J6 mice. Postischemic revascularization, quantified by measuring von Willebrand factor expression, was significantly impaired, suggesting that factors other than progressive EDNO deterioration are also involved in the age-dependent severe disease phenotype. Ischemia led to an increase in the expression of vascular endothelial growth factor receptor-2, KDR, in younger ecNOS-KO; however, this increase in KDR expression was absent in the older animals. Lack of increased KDR expression may provide a mechanistic explanation for the severe ischemic injury and perhaps can be used as a clinical marker to identify severe, vascular endothelial growth factor refractory patient population.


Assuntos
Isquemia Miocárdica/fisiopatologia , Óxido Nítrico Sintase Tipo III/deficiência , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Envelhecimento , Animais , Endotélio Vascular/metabolismo , Artéria Femoral/fisiologia , Expressão Gênica/fisiologia , Proteínas Hedgehog , Isquemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/fisiologia , Necrose , Óxido Nítrico/metabolismo , Óxido Nítrico/fisiologia , Fluxo Sanguíneo Regional/fisiologia , Transativadores/genética , Regulação para Cima
17.
Life Sci ; 78(26): 3066-73, 2006 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-16413583

RESUMO

Myocardial infarction (MI) activates the renin-angiotensin system in the heart and increases local production of aldosterone. This hormone may increase reactive fibrosis in the myocardium favoring heart failure development. To elucidate the potential contribution of aldosterone to cardiac remodeling following MI, we evaluated the expression of mineralocorticoid receptors (MCR) in the left ventricle (LV) and kidney of rats after MI and captopril treatment. MI was induced by ligation of the coronary artery in Wistar rats, which were separated into (1) sham-operated group, (2) MI group, (3) MI-captopril treated group (cap, 50 mg kg(-1) day(-1)). One month later angiotensin converting enzyme (ACE) activity was assayed in the plasma, LV and kidney. Cardiac and renal angiotensin II (Ang II) levels were determined by ELISA and MCR mRNA expression and protein were measured by Taqman RT-PCR and Western blot, respectively. Cardiac MCR mRNA and protein levels increased nearly by 80% after MI and Cap treatment normalized cardiac MCR protein and mRNA expression. Kidney MCR expression was not affected. ACE activity increased 34% in the plasma and 83% in the LV after MI. This increase was prevented by Cap. Ang II concentration increased 225% in the LV and 193% in kidney, which was partially attenuated by Cap. Our data demonstrate upregulation of MCR in the heart following MI what may facilitate the effects of aldosterone in the ventricular remodeling process. ACE inhibitors may reduce reactive fibrosis not only by decreasing Ang II production but also by attenuating the aldosterone-signaling pathway by decreasing the expression of MCR receptors.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Captopril/farmacologia , Rim/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptores de Mineralocorticoides/biossíntese , Angiotensina II/metabolismo , Animais , Western Blotting , Hemodinâmica/fisiologia , Masculino , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar
18.
Proc Natl Acad Sci U S A ; 102(31): 10999-1004, 2005 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-16043715

RESUMO

The genetic loss of endothelial-derived nitric oxide synthase (eNOS) in mice impairs vascular endothelial growth factor (VEGF) and ischemia-initiated blood flow recovery resulting in critical limb ischemia. This result may occur through impaired arteriogenesis, angiogenesis, or mobilization of stem and progenitor cells. Here, we show that after ischemic challenge, eNOS knockout mice [eNOS (-/-)] have defects in arteriogenesis and functional blood flow reserve after muscle stimulation and pericyte recruitment, but no impairment in endothelial progenitor cell recruitment. More importantly, the defects in blood flow recovery, clinical manifestations of ischemia, ischemic reserve capacity, and pericyte recruitment into the growing neovasculature can be rescued by local intramuscular delivery of an adenovirus encoding a constitutively active allele of eNOS, eNOS S1179D, but not a control virus. Collectively, our data suggest that endogenous eNOS-derived NO exerts direct effects in preserving blood flow, thereby promoting arteriogenesis, angiogenesis, and mural cell recruitment to immature angiogenic sprouts.


Assuntos
Isquemia/enzimologia , Óxido Nítrico Sintase/fisiologia , Animais , Extremidades/irrigação sanguínea , Expressão Gênica , Técnicas de Transferência de Genes , Isquemia/patologia , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/irrigação sanguínea , Neovascularização Patológica , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Pericitos/patologia , Fluxo Sanguíneo Regional
19.
Protein Expr Purif ; 42(2): 295-304, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15925519

RESUMO

Prolyl-4-hydroxylase domain-containing enzymes (PHDs) mediate the oxygen-dependent regulation of the heterodimeric transcription factor hypoxia-inducible factor-1 (HIF-1). Under normoxic conditions, one of the subunits of HIF-1, HIF-1alpha, is hydroxylated on specific proline residues to target HIF-1alpha for degradation by the ubiquitin-proteasome pathway. Under hypoxic conditions, the hydroxylation by the PHDs is attenuated by lack of the oxygen substrate, allowing HIF-1 to accumulate, translocate to the nucleus, and mediate HIF-mediated gene transcription. In several mammalian species including humans, three PHDs have been identified. We report here the cloning of a full-length rat cDNA that is highly homologous to the human and murine PHD-1 enzymes and encodes a protein that is 416 amino acids long. Both cDNA and protein are widely expressed in rat tissues and cell types. We demonstrate that purified and crude baculovirus-expressed rat PHD-1 exhibits HIF-1alpha specific prolyl hydroxylase activity with similar substrate affinities and is comparable to human PHD-1 protein.


Assuntos
Clonagem Molecular , Pró-Colágeno-Prolina Dioxigenase/química , Pró-Colágeno-Prolina Dioxigenase/genética , Sequência de Aminoácidos , Animais , Humanos , Cinética , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos/genética , Pró-Colágeno-Prolina Dioxigenase/biossíntese , RNA Mensageiro/metabolismo , Ratos , Spodoptera
20.
Circulation ; 111(17): 2219-26, 2005 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-15851596

RESUMO

BACKGROUND: Angiotensin II (Ang II) accelerates atherosclerosis and induces abdominal aortic aneurysm (AAA) in an experimental mouse model. Agonism of a G protein-coupled receptor by Ang II activates Rho-kinase and other signaling pathways and results in activation of proteolysis and apoptosis. Enhanced proteolysis and smooth muscle cell apoptosis are important mechanisms associated with AAA. In this study, we tested the hypothesis that fasudil, a Rho-kinase inhibitor, could attenuate Ang II-induced AAA formation by inhibiting vascular wall apoptosis and extracellular matrix proteolysis. METHODS AND RESULTS: Six-month-old apolipoprotein E-deficient mice were infused with Ang II (1.44 mg x kg(-1) x d(-1)) for 1 month. Animals were randomly assigned to treatment with fasudil (136 or 213 mg x kg(-1) x d(-1) in drinking water) or tap water. Ang II infusion induced AAA formation in 75% of the mice, which was accompanied by an increase in proteolysis detected by zymographic analysis and quantified by active matrix metalloproteinase-2 activity, as well as apoptosis detected by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and quantified by both caspase-3 activity and histone-associated DNA fragmentation. The level of DNA fragmentation in the suprarenal aorta correlated with AAA diameter. Ang II also increased atherosclerotic lesion area and blood pressure. Fasudil treatment resulted in a dose-dependent reduction in both the incidence and severity of AAA. At the higher dose, fasudil decreased AAA by 45% while significantly inhibiting both apoptosis and proteolysis, without affecting atherosclerosis or blood pressure. CONCLUSIONS: These data demonstrate that inhibition of Rho-kinase by fasudil attenuated Ang II-induced AAA through inhibition of both apoptosis and proteolysis pathways.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Angiotensina II/farmacologia , Aneurisma da Aorta Abdominal/tratamento farmacológico , Apolipoproteínas E/deficiência , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Apoptose , Endotélio Vascular/citologia , Matriz Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Inibidores de Proteases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Quinases Associadas a rho
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...