Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Oncogene ; 40(1): 127-139, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33093654

RESUMO

The oncofetal long noncoding RNA (lncRNA) H19 is postnatally repressed in most tissues, and re-expressed in many cancers, including hepatocellular carcinoma (HCC). The role of H19 in carcinogenesis is a subject of controversy. We aimed to examine the role of H19 in chronic inflammation-mediated hepatocarcinogenesis using the Mdr2/Abcb4 knockout (Mdr2-KO) mouse, a well-established HCC model. For this goal, we have generated Mdr2-KO/H19-KO double knockout (dKO) mice and followed spontaneous tumor development in the dKO and control Mdr2-KO mice. Cellular localization of H19 and effects of H19 loss in the liver were determined in young and old Mdr2-KO mice. Tumor incidence and tumor load were both significantly decreased in the liver of dKO versus Mdr2-KO females. The expression levels of H19 and Igf2 were variable in nontumor liver tissues of Mdr2-KO females and were significantly downregulated in most matched tumors. In nontumor liver tissue of aged Mdr2-KO females, H19 was expressed mainly in hepatocytes, and hepatocyte proliferation was increased compared to dKO females. At an early age, dKO females displayed lower levels of liver injury and B-cell infiltration, with higher percentage of binuclear hepatocytes. In human samples, H19 expression was higher in females, positively correlated with cirrhosis (in nontumor liver samples) and negatively correlated with CTNNB1 (beta-catenin) mutations and patients' survival (in tumors). Our data demonstrate that the lncRNA H19 is pro-oncogenic during the development of chronic inflammation-mediated HCC in the Mdr2-KO mouse model, mainly by increasing liver injury and decreasing hepatocyte polyploidy in young mice.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Carcinoma Hepatocelular/patologia , Fibrose/genética , Neoplasias Hepáticas/patologia , RNA Longo não Codificante/genética , beta Catenina/genética , Animais , Carcinoma Hepatocelular/genética , Feminino , Fibrose/complicações , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Knockout , Caracteres Sexuais , Carga Tumoral , Regulação para Cima , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
2.
Oncotarget ; 6(33): 34691-703, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26415227

RESUMO

The function of imprinted H19 long non-coding RNA is still controversial. It is highly expressed in early embryogenesis and decreases after birth and re-expressed in cancer. To study the role of H19 in oncogenesis and pluripotency, we down-regulated H19 expression in vitro and in vivo in pluripotent human embryonic carcinoma (hEC) and embryonic stem (hES) cells. H19 knockdown resulted in a decrease in the expression of the pluripotency markers Oct4, Nanog, TRA-1-60 and TRA-1-81, and in the up-regulation of SSEA1; it further attenuated cell proliferation, decreased cell-matrix attachment, and up-regulated E-Cadherin expression. SCID-Beige mice transplanted with H19 down-regulated hEC cells exhibited slower kinetics of tumor formation, resulting in an increased animal survival. Tumors derived from H19 down-regulated cells showed a decrease in the expression of pluripotency markers and up-regulation of SSEA-1 and E-cadherin. Our results suggest that H19 oncogenicity in hEC cells is mediated through the regulation of the pluripotency state.


Assuntos
Transformação Celular Neoplásica/genética , Células-Tronco Embrionárias , Células-Tronco Pluripotentes , RNA Longo não Codificante/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Transfecção
3.
Radiology ; 276(2): 426-32, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26203709

RESUMO

PURPOSE: To compare hepatocellular carcinoma (HCC) development after radiofrequency (RF) ablation, partial surgical hepatectomy, and a sham operation and to inhibit HCC recurrence after RF ablation in a mouse model of spontaneously forming HCC in the setting of chronic inflammation (ie, the MDR2 knockout model). MATERIALS AND METHODS: Animal experiments were performed according to an approved animal care committee protocol. The authors compared the survival of MDR2 knockout mice (an inflammation-induced HCC model) that underwent RF ablation, 35% partial hepatectomy (ie, left lobectomy), or a sham operation (controls) by using Kaplan-Meier survival curve analysis. Tumor load and tumor frequency in mice that underwent sham operation were further compared with those of mice treated with RF ablation at 1 month after therapy by using a two-tailed Student t test. Liver slices from mice treated with RF ablation were stained for α-smooth muscle actin and Ki-67 to establish the role of liver regeneration in the tumorigenic effect of RF ablation. Finally, tumor load and tumor incidence were evaluated in mice treated with a c-met inhibitor after RF ablation by using the Mann-Whitney U test. RESULTS: Ablation of 3.5% ± 0.02 of the MDR2 knockout mice liver induced increased tumor load (P = .007) and reduced survival (P = .03) in comparison to that of controls, with no significant difference to the 10-fold volume removal of partial hepatectomy. Seven days after RF treatment, the border zone of the coagulation zone was surrounded by α-smooth muscle actin-positive activated myofibroblasts. A significant elevation of hepatocyte proliferation was also seen 7 days after RF ablation in the distant liver (ablated lobe: P = .003; untreated lobe: P = .02). A c-met inhibitor significantly attenuated HCC development in MDR2 knockout mice treated with RF ablation (P = .001). CONCLUSION: Liver regeneration induced by RF ablation facilitates c-met/hepatocyte growth factor axis-dependent HCC tumor formation after treatment in the MDR2 knockout model. Blockage of the c-met/hepatocyte growth factor axis attenuates HCC recurrence, raising the potential for therapeutic intervention to reverse this potentially deleterious tumorigenic effect.


Assuntos
Técnicas de Ablação/efeitos adversos , Carcinogênese , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/cirurgia , Eletrocirurgia/efeitos adversos , Hepatectomia/métodos , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/cirurgia , Animais , Camundongos , Recidiva Local de Neoplasia/prevenção & controle
4.
Radiology ; 276(2): 416-25, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25822472

RESUMO

PURPOSE: To determine the kinetics of innate immune and hepatic response to the coagulation necrosis area that remains in situ after radiofrequency (RF) ablation, the cytokine profile of this response, and its local and global effect on the whole organ in a small-animal model. MATERIALS AND METHODS: A standardized RF ablation dose (70°C for 5 minutes) was used to ablate more than 7% of the liver in 91 C57BL6 mice (wild type) according to a protocol approved by the animal care committee. The dynamic cellular response in the border zone surrounding ablation-induced coagulation and in the ablated lobe and an untreated lobe were characterized with immunohistochemistry 24 hours, 72 hours, 7 days, and 14 days after ablation (the time points at which cells migrate to necrotic tissues). After characterization of the cellular populations that reacted to the RF treatment, cytokines secreted by these cells were blocked, either by using interleukin-6 knockout mice (n = 24) or c-met inhibitor PHA 665752 (n = 15), to elucidate the key factors facilitating the wound healing response to RF ablation. Statistical significance was assessed with nonparametric analysis of variance. RESULTS: RF ablation induces a strong time-dependent immunologic response at the perimeter of the necrotic zone. This includes massive accumulation of neutrophils, activated myofibroblasts, and macrophages peaking at 24 hours, 7 days, and 14 days after ablation, respectively. In correlation with myofibroblast accumulation, RF ablation induced hepatocyte proliferation in both the ablated lobe and an untreated lobe (mean, 165.15 and 230.4 cyclin-dependent kinase 47-positive cells per ×20 field, respectively, at day 7; P < .02). Blockade of either IL-6 or c-met significantly reduced global hepatocyte proliferation (P < .05 for both), with the former reducing the accumulation of both macrophages and myofibroblasts surrounding the coagulation necrosis area (42.9 and 113.6 vs 7.3 and 46.6 macrophages and activated myofibroblasts per ×20 field, respectively; P < .036 for both). CONCLUSION: Hepatic RF ablation induces not only a local periablational inflammatory zone but also more global proliferative effects on the liver. These IL-6- and/or c-met-mediated changes could potentially account for some of the local and distant tumor recurrence observed after treatment.


Assuntos
Técnicas de Ablação , Eletrocirurgia , Regeneração Hepática , Fígado/patologia , Fígado/cirurgia , Técnicas de Ablação/efeitos adversos , Animais , Eletrocirurgia/efeitos adversos , Inflamação/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Necrose/etiologia
5.
Hepatology ; 58(3): 1021-30, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23526353

RESUMO

UNLABELLED: Human hepatocellular carcinoma (HCC) is an inflammation-induced cancer, which is the third-leading cause of cancer mortality worldwide. We investigated the role of the chemokine receptors, CCR5 and CCR1, in regulating inflammation and tumorigenesis in an inflammation-induced HCC model in mice. Multidrug resistance 2 gene (Mdr2)-knockout (Mdr2-KO) mice spontaneously develop chronic cholestatic hepatitis and fibrosis that is eventually followed by HCC. We generated two new strains from the Mdr2-KO mouse, the Mdr2:CCR5 and the Mdr2:CCR1 double knockouts (DKOs), and set out to compare inflammation and tumorigenesis among these strains. We found that in Mdr2-KO mice lacking the chemokine receptor, CCR5 (Mdr2:CCR5 DKO mice), but not CCR1 (Mdr2:CCR1 DKO), macrophage recruitment and trafficking to the liver was significantly reduced. Furthermore, in the absence of CCR5, reduced inflammation was also associated with reduced periductal accumulation of CD24(+) oval cells and abrogation of fibrosis. DKO mice for Mdr2 and CCR5 exhibited a significant decrease in tumor incidence and size. CONCLUSIONS: Our results indicate that CCR5 has a critical role in both the development and progression of liver cancer. Therefore, we propose that a CCR5 antagonist can serve for HCC cancer prevention and treatment.


Assuntos
Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/fisiopatologia , Hepatite Crônica/complicações , Cirrose Hepática/complicações , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/fisiopatologia , Receptores CCR5/fisiologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/deficiência , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/fisiologia , Animais , Carcinoma Hepatocelular/epidemiologia , Quimiocina CCL5/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Hepatite Crônica/genética , Incidência , Fígado/patologia , Fígado/fisiopatologia , Cirrose Hepática/genética , Neoplasias Hepáticas/epidemiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CCR1/deficiência , Receptores CCR1/genética , Receptores CCR1/fisiologia , Receptores CCR5/deficiência , Receptores CCR5/genética , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
6.
J Phys Chem B ; 116(23): 6945-51, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22482826

RESUMO

This paper uses advances in the ultrafast manipulation of light to address a general need in medicine for a clinical approach that can provide a solution to a variety of disorders requiring subsurface tissue manipulation with ultralow collateral damage. Examples are age-related macular degeneration (AMD), fungal infections, tumors surrounded by overlying tissue, cataracts, etc. Although lasers have revolutionized the use of light in clinical settings, most lasers employed in medicine cannot address such problems of depth-selective tissue manipulation. This arises from the fact that they are mostly based on one photon based laser tissue interactions that provide a cone of excitation where the energy density is sufficiently high to excite heat or fluorescence in the entire cone. Thus, it is difficult to excite a specific depth of a tissue without affecting the overlying surface. However, the advent of femtosecond (fs) lasers has caused a revolution in multiphoton microscopy (Zipfel et al. Nat. Biotechnol. 2003, 21, 1369-1377; Denk et al. Science 1990, 248, 73-76) and fabrication (Kawata et al. Nature 2001, 412, 697-698). With such lasers, the photon energy density is only high enough for multiphoton processes in the focal volume, and this opens a new direction to address subsurface tissue manipulation. Here we show in an AMD animal model, Ccr2 KO knockout mutant mice, noninvasive, selective fs two-photon photobleaching of pigments associated with AMD that accumulate under and in ultraclose proximity to the overlying retina. Pathological evidence is presented that indicates the lack of collateral damage to the overlying retina or other surrounding tissue.


Assuntos
Lentes de Contato , Degeneração Macular/patologia , Fotodegradação , Lesões Experimentais por Radiação/prevenção & controle , Retina/patologia , Pigmentos da Retina/metabolismo , Animais , Lasers , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência por Excitação Multifotônica
7.
Exp Hematol ; 39(3): 282-92, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21138752

RESUMO

OBJECTIVE: The chemokine receptor CXCR4 and its ligand CXCL12 are involved in the progression and dissemination of a diverse number of solid and hematological malignancies. Binding CXCL12 to CXCR4 activates a variety of intracellular signal transduction pathways that regulate cell chemotaxis, adhesion, survival, proliferation, and apoptosis. MATERIALS AND METHODS: Here, we demonstrate that the CXCR4 antagonist, 4F-benzoyl-TN14003 (BKT140), but not AMD3100, exhibits a CXCR4-dependent preferential cytotoxicity toward malignant cells of hematopoietic origin. BKT140 significantly and preferentially stimulated multiple myeloma apoptotic cell death. BKT140 treatment induced morphological changes, phosphatidylserine externalization, decreased mitochondrial membrane potential, caspase-3 activation, sub-G1 arrest, and DNA double-stranded breaks. RESULTS: In vivo, subcutaneous injections of BKT140 significantly reduced, in a dose-dependent manner, the growth of human acute myeloid leukemia and multiple myeloma xenografts. Tumors from animals treated with BKT140 were smaller in size and weights, had larger necrotic areas and high apoptotic scores. CONCLUSIONS: Taken together, these results suggest a potential therapeutic use for BKT140 in multiple myeloma and leukemia patients.


Assuntos
Apoptose/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Mieloma Múltiplo/tratamento farmacológico , Peptídeos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Animais , Fármacos Anti-HIV/farmacologia , Antineoplásicos , Benzilaminas , Quimiocina CXCL12/metabolismo , Ciclamos , Células HL-60 , Compostos Heterocíclicos/farmacologia , Humanos , Células Jurkat , Leucemia Mieloide Aguda/metabolismo , Camundongos , Camundongos SCID , Mieloma Múltiplo/metabolismo , Transplante de Neoplasias , Receptores CXCR4/metabolismo
8.
PLoS One ; 4(4): e5125, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19340288

RESUMO

The chemokine receptor CXCR4 and its ligand CXCL12 is overexpressed in the majority of tumors and is critically involved in the development and metastasis of these tumors. CXCR4 is expressed in malignant tumor cells whereas its ligand SDF-1 (CXCL12) is expressed mainly by cancer associated fibroblasts (CAF). Similarly to CXCR4, the chemokine CCL20 is overexpressed in variety of tumors; however its role and regulation in tumors is not fully clear. Here, we show that the chemokine receptor CXCR4 stimulates the production of the chemokine CCL20 and that CCL20 stimulates the proliferation and adhesion to collagen of various tumor cells. Furthermore, overexpression of CCL20 in tumor cells promotes growth and adhesion in vitro and increased tumor growth and invasiveness in vivo. Moreover, neutralizing antibodies to CCL20 inhibit the in vivo growth of tumors that either overexpress CXCR4 or CCL20 or naturally express CCL20. These results reveal a role for CCL20 in CXCR4-dependent and -independent tumor growth and suggest a therapeutic potential for CCL20 and CCR6 antagonists in the treatment of CXCR4- and CCL20-dependent malignancies.


Assuntos
Divisão Celular/fisiologia , Quimiocina CCL20/fisiologia , Neoplasias/patologia , Receptores CXCR4/fisiologia , Adesão Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Invasividade Neoplásica , Neoplasias/irrigação sanguínea , Neovascularização Patológica , Reação em Cadeia da Polimerase
9.
FASEB J ; 21(13): 3522-33, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17575264

RESUMO

A femtosecond laser beam gene transduction (SG-LBGT) system is described as a novel and efficient method of intradermal (i.d.) nonviral gene delivery in mice by permeabilizing cells utilizing femtosecond laser pulses. Using this approach, significant gene expression and efficient dermal transduction lasting for >7 months were obtained. The ability of this new DNA gene transfer method to enhance genetic vaccination was tested in BALB/C mice. A single i.d. injection of a plasmid (10 microg) containing the hepatitis B virus (HBV) surface antigen (HBsAg), followed by pulses of laser, induced high titers of HBsAg-specific antibodies lasting for >210 days and increased levels of IgG1, IgG2a, IFNgamma, and IL-4, indicating the activation of both Th1 and Th2 cells. Moreover, mice vaccinated using the SG-LBGT followed by challenge with pHBV showed increased protection against viral challenge, as detected by decreased levels of HBV DNA, suggesting an efficient Th1 effect against HBV-infected replicating cells. Tumor growth retardation was induced in vaccinated mice challenged with an HBsAg-expressing syngeneic tumor. In most of the parameters tested, administration of plasmid followed by laser application was significantly more effective and prolonged than that of plasmid alone. Tissue damage was not detected and integration of the plasmid into the host genomic DNA probably did not occur. We suggest that the LBGT method is an efficient and safe technology for in vivo gene expression and vaccination and emphasizes its potential therapeutic applications for i.d. nonviral gene delivery.


Assuntos
DNA/administração & dosagem , Expressão Gênica , Vacinas de DNA/administração & dosagem , Animais , Células Cultivadas , Antígenos de Superfície da Hepatite B/genética , Antígenos de Superfície da Hepatite B/imunologia , Vírus da Hepatite B/imunologia , Imunoglobulina G/metabolismo , Interferon gama/metabolismo , Interleucina-4/metabolismo , Lasers , Camundongos , Camundongos Endogâmicos BALB C , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
10.
Mol Ther ; 15(4): 772-81, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17299399

RESUMO

Replication-deficient adenoviruses are useful vectors for the transfer of therapeutic transgenes to malignant and non-malignant tissues. Yet their clinical application is limited by the potential toxicity of viral infection and the transient nature of transgene expression. Although transgene expression from adenovirus vectors is initially higher than expression of transgenes transduced by other viral or non-viral vectors, it is often insufficient to generate a significant therapeutic effect. We addressed this issue by searching for DNA-targeted viral-induced host responses potentially restricting transgene expression. Nuclear protein extracts from livers of rats systemically infected with replication-deficient adenovirus exhibited enhanced topoisomerase I activity compared with extracts from uninfected animals. Consequently, the inhibition of topoisomerase I by the anti-cancer drug topotecan greatly enhanced transgene expression in adenovirus-infected hepatic cells, colon cancer and prostate cancer cell cultures, mouse liver, human ex vivo tumor specimens, and mouse tumor in vivo. The enhancement could not be ascribed to non-specific genotoxic stress, cell death, or cell-cycle perturbation. These findings are significant for gene therapy as they reveal novel aspects of the host anti-adenovirus response and set the stage for the development of a rational molecular-pharmacological approach to increase the effectiveness, and safety, of adenovirus-mediated cancer therapeutics.


Assuntos
Adenoviridae/genética , DNA Topoisomerases Tipo I/biossíntese , Vírus Defeituosos/genética , Vetores Genéticos , Adenoviridae/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Vírus Defeituosos/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Terapia Genética/métodos , Humanos , Técnicas In Vitro , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/terapia , Ratos , Ratos Endogâmicos Lew , Inibidores da Topoisomerase I , Topotecan/farmacologia , Transdução Genética
11.
Shock ; 27(3): 266-73, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17304107

RESUMO

Adenosine receptors (AR) and extracellular signal-regulated kinases (ERK) have been implicated in tissue protection and apoptosis regulation during ischemia/reperfusion (I/R) injury. This study tests the hypothesis that reduction of reperfusion lung injury after A2A AR activation is associated with attenuation of apoptosis, modulation of ERK activation, and alterations in antiapoptotic and proapoptotic protein expression (Bcl-2 and Bax, respectively). Experiments were performed in intact-chest, spontaneously breathing cats in which the arterial branch of the left lower lung lobe was occluded for 2 h and reperfused for 3 h (I/R group). Animals were treated with the selective A2A AR agonist ATL313 given 5 min before reperfusion alone or in combination with the selective A2A AR antagonist ZM241385. Western blot analysis showed significant reduction in expression of Bcl-2 and increase in expression of Bax after reperfusion, compared with control lungs. Phosphorylated ERK1/2 levels were also increased after reperfusion. Compared with the I/R group, ATL313 markedly (P < 0.01) attenuated indices of injury and apoptosis including the percentage of injured alveoli, wet-dry weight ratio, myeloperoxidase activity, in situ terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling-positive cells, and caspase 3 activity and expression. Furthermore, compared with reperfused lungs, in ATL313-pretreated lungs, Western blot analysis demonstrated substantial ERK1/2 activation, increased expression of Bcl-2, and attenuated expression of Bax. The protective effects of ATL313 were blocked by pretreatment with ZM241385. In summary, the present study shows that in vivo activation of A2A AR confers protection against reperfusion lung injury. This protection is associated with decreased apoptosis and involves ERK1/2 activation and alterations in antiapoptotic Bcl-2 and proapoptotic Bax proteins.


Assuntos
Apoptose , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Lesão Pulmonar , Pulmão/patologia , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor A2A de Adenosina/metabolismo , Traumatismo por Reperfusão/metabolismo , Proteína X Associada a bcl-2/metabolismo , Agonistas do Receptor A2 de Adenosina , Animais , Gatos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Piperidinas/química , Alvéolos Pulmonares/metabolismo , Triazinas/farmacologia , Triazóis/farmacologia , Ferimentos e Lesões
12.
Crit Care ; 10(2): R65, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16623960

RESUMO

INTRODUCTION: Although activation of A3 adenosine receptors attenuates reperfusion lung injury and associated apoptosis, the signaling pathway that mediates this protection remains unclear. Adenosine agonists activate mitogen-activated protein kinases, and these kinases have been implicated in ischemia/reperfusion injury; the purpose of this study was therefore to determine whether A3 adenosine receptor stimulation with reperfusion modulates expression of the different mitogen-activated protein kinases. In addition, we compared the effect of the A3 adenosine agonist IB-MECA with the newly synthesized, highly selective A3 adenosine receptor agonist MRS3558 on injury in reperfused lung. METHOD: Studies were performed in an in vivo spontaneously breathing cat model, in which the left lower lobe of the lung was isolated and subjected to 2 hours of ischemia and 3 hours of reperfusion. The selective A3 adenosine receptor agonists IB-MECA (0.05 mg/kg, 0.1 mg/kg, or 0.3 mg/kg) and MRS3558 (0.05 mg/kg or 0.1 mg/kg) were administered before reperfusion. RESULTS: Both A3 adenosine receptor agonists administered before reperfusion markedly (P < 0.01) attenuated indices of injury and apoptosis, including the percentage of injured alveoli, wet/dry weight ratio, myeloperoxidase activity, TUNEL (in situ TdT-mediated dUTP nick end labeling)-positive cells, and caspase 3 activity and expression. The more pronounced effects at low doses were observed with MRS3558. Increases in phosphorylated c-Jun amino-terminal protein kinase (JNK), p38, and extracellular signal-regulated kinase (ERK)1/2 levels were observed by the end of reperfusion compared with controls. Pretreatment with the A3 agonists upregulated phosphorylated ERK1/2 levels but did not modify phosphorylated JNK and p38 levels. CONCLUSION: The protective effects of A3 adenosine receptor activation are mediated in part through upregulation of phosphorylated ERK. Also, MRS3558 was found to be more potent than IB-MECA in attenuating reperfusion lung injury. The results suggest not only that enhancement of the ERK pathway may shift the balance between cell death and survival toward cell survival, but also that A3 agonists have potential as an effective therapy for ischemia/reperfusion-induced lung injury.


Assuntos
Pneumopatias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptor A3 de Adenosina/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose/fisiologia , Gatos , Pneumopatias/genética , Pneumopatias/patologia , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Proteínas Quinases Ativadas por Mitógeno/genética , Receptor A3 de Adenosina/biossíntese , Receptor A3 de Adenosina/genética , Traumatismo por Reperfusão/genética
13.
Blood ; 106(7): 2280-6, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15798000

RESUMO

Gene therapy holds a major promise. However, until now, this promise was fulfilled only in few cases, in rare genetic diseases. One very common clinical condition is anemia. Patients with anemia of chronic renal failure are treated with erythropoietin. The objective of this study was to develop a therapeutic platform for serum-secreted proteins like erythropoietin. We developed a tissue protein factory based on dermal cores (Biopump) harvested and implanted autologously. In this study, an adenovector was designed to express the human erythropoietin under the control of the cytomegalovirus (CMV) promoter. This vector transduced the harvested dermal cores ex vivo. The transduced cores were implanted, and erythropoietin and reticulocyte counts were measured. Dermal cores were harvested from 13 patients with chronic renal failure, and implantation was performed in 10. There were no significant drug-related side effects to this procedure. Erythropoietin serum levels increased significantly to therapeutic levels from day 1 after implantation reaching a peak during the first week of follow-up. The expression period was transient for up to 14 days. The rise of erythropoietin was followed by a transient significant increase in reticulocyte counts. The decrease of erythropoietin expression coincided with a significant dermal infiltrate of CD8 cytotoxic T cells. Antierythropoietin antibodies were not detected until day 90 following implantation. Implantation of dermal cores ex vivo transduced with human genes could eventually be used in the clinical setting to express therapeutic serum proteins. However, nonimmunogenic delivery system should be tested as gene vehicles.


Assuntos
Eritropoetina/genética , Terapia Genética/métodos , Falência Renal Crônica/genética , Falência Renal Crônica/terapia , Adenoviridae/genética , Adulto , Idoso , Anemia/terapia , Linfócitos T CD8-Positivos/citologia , Citomegalovirus/genética , Eritropoetina/sangue , Eritropoetina/metabolismo , Vetores Genéticos , Humanos , Imuno-Histoquímica , Inflamação , Pessoa de Meia-Idade , Proteínas Recombinantes , Reticulócitos/citologia , Reticulócitos/metabolismo , Pele/metabolismo , Pele/patologia , Fatores de Tempo
14.
Am J Transplant ; 4(12): 1941-8, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15575895

RESUMO

Apoptosis has been described in various models of ischemia-reperfusion (IR) injury, including lung transplantation. A3 adenosine receptor (AR) has been linked to a variety of apoptotic processes. The effect of A3AR activation on lung injury and apoptosis, following IR, has not been reported to date. In a spontaneously breathing cat model, in which the left lower lobe of the lung was isolated and subjected to 2 h of ischemia and 3 h of reperfusion, we tested the effect of IB-MECA, a selective A3AR agonist, on lung apoptosis and injury. Significant increase in the extent of apoptosis was observed following lung reperfusion. IB-MECA, administered before IR, and before or with reperfusion, markedly (p < 0.01) attenuated indices of injury and apoptosis including the percentage of injured alveoli, wet/dry weight ratio, myeloperoxidase activity, in situ terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) positive cells, and caspase 3 activity and expression. The protective effects of IB-MECA were completely blocked by pretreatment with the selective A3AR antagonist MRS-1191. In summary, even when given after the onset of ischemia, the A3AR agonist IB-MECA conferred a powerful protection against reperfusion lung injury, which was associated with decreased apoptosis. This suggests a potentially important role for A3AR in lung IR injury.


Assuntos
Adenosina/análogos & derivados , Adenosina/farmacologia , Apoptose/fisiologia , Pulmão/irrigação sanguínea , Receptor A3 de Adenosina/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Gatos , Modelos Animais de Doenças , Marcação In Situ das Extremidades Cortadas , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lesão Pulmonar , Traumatismo por Reperfusão/patologia
15.
Anesthesiology ; 101(5): 1153-9, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15505451

RESUMO

BACKGROUND: A3 adenosine receptor (AR) activation worsens or protects against renal and cardiac ischemia-reperfusion (IR) injury, respectively. The aims of the current study were to examine in an in vivo model the effect of A3AR activation on IR lung injury and investigate the mechanism by which it exerts its effect. METHODS: The arterial branch of the left lower lung lobe in intact-chest, spontaneously breathing cats was occluded for 2 h and reperfused for 3 h (IR group). Animals were treated with the selective A3 receptor agonist IB-MECA (300 microg/kg intravenously) given 15 min before ischemia or with IB-MECA as described, with pretreatment 15 min earlier with the selective A3AR antagonist MRS-1191, the nonsulfonylurea adenosine triphosphate-sensitive potassium channel-blocking agent U-37883A, or the nitric oxide synthase inhibitor N-nitro-l-arginine benzyl ester. RESULTS: IB-MECA markedly (P < 0.01) reduced the percentage of injured alveoli (IR, 48 +/- 4%; IB-MECA, 18 +/- 2%), wet:dry weight ratio (IR, 8.2 +/- 0.4; IB-MECA, 4 +/- 2), and myeloperoxidase activity (IR, 0.52 +/- 0.06 U/g; IB-MECA, 0.17 +/- 0.04 U/g). This protective effect was completely blocked by pretreatment with the selective A3AR antagonist MRS-1191 and the adenosine triphosphate-sensitive potassium channel blocking agent U-37883A but not the nitric oxide synthase inhibitor N-nitro-l-arginine benzyl ester. CONCLUSIONS: In the feline lung, the A3AR agonist IB-MECA confers a powerful protection against IR lung injury. This effect is mediated by a nitric oxide synthase-independent pathway and involves opening of adenosine triphosphate-sensitive potassium channels. Therefore, selective activation of A3AR may be an effective means of protecting the reperfused lung.


Assuntos
Adamantano/análogos & derivados , Agonistas do Receptor A3 de Adenosina , Adenosina/análogos & derivados , Pulmão/patologia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle , Transportadores de Cassetes de Ligação de ATP , Adamantano/farmacologia , Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina , Animais , Gatos , Di-Hidropiridinas/farmacologia , Inibidores Enzimáticos/farmacologia , Hemodinâmica/efeitos dos fármacos , Histamina/sangue , Técnicas In Vitro , Canais KATP , Morfolinas/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III , Nitroarginina/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiologia
16.
FASEB J ; 18(11): 1240-2, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15180966

RESUMO

Hormone refractory metastatic prostate cancer remains an incurable disease. We found that high expression levels of the chemokine receptor CXCR4 correlated with the presence of metastatic disease in prostate cancer patients. Positive staining for CXCL12, the ligand for CXCR4, was mainly present in the tumor-associated blood vessels and basal cell hyperplasia. Subcutaneous xenografts of PC3 and 22Rv1 prostate tumors that overexpressed CXCR4 in NOD/SCID mice were two- to threefold larger in volume and weight vs. controls. Moreover, blood vessel density, functionality, invasiveness of tumors into the surrounding tissues, and metastasis to the lymph node and lung were significantly increased in these tumors. Neutralizing the interactions of CXCL12/CXCR4 in vivo with CXCR4 specific antibodies inhibited the CXCR4-dependent tumor growth and vascularization. In vitro, CXCL12 induced the proliferation and VEGF secretion but not migration of PC3 and 22Rv1 cells overexpressing CXCR4. Similar effects of CXCR4 overexpression on tumor growth in vivo were also noted in two breast cancer lines, suggesting that the observed effect of CXCR4 is not unique to prostate tumor cells. Thus high levels of the chemokine receptor CXCR4 induce a more aggressive phenotype in prostate cancer cells and identify CXCR4 as a potential therapeutic target in advanced cases of metastatic prostate cancer.


Assuntos
Adenocarcinoma/metabolismo , Metástase Neoplásica/genética , Proteínas de Neoplasias/fisiologia , Neovascularização Patológica/genética , Neoplasias da Próstata/metabolismo , Receptores CXCR4/fisiologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Medula Óssea/patologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/patologia , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12 , Quimiocinas CXC/análise , Quimiocinas CXC/farmacologia , Feminino , Humanos , Hiperplasia , Neoplasias Pulmonares/secundário , Metástase Linfática , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Especificidade de Órgãos , Neoplasias Ovarianas/patologia , Fenótipo , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Proteínas Recombinantes de Fusão/fisiologia , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Cancer Res ; 64(4): 1338-46, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14973073

RESUMO

Tumor microenvironment controls the selection of malignant cells capable of surviving in stressful and hypoxic conditions. The transcription factor, cyclic AMP-responsive element binding (CREB) protein, activated by multiple extracellular signals, modulates cellular response by regulating the expression of a multitude of genes. Previously, we have demonstrated that two cystein residues, at the DNA binding domain of CREB, mediate activation of CREB-dependent gene expression at normoxia and hypoxia. The construction of a dominant-positive CREB mutant, insensitive to hypoxia cue (substitution of two cystein residues at position 300 and 310 with serine in the DNA binding domain) and of a dominant negative CREB mutant (addition of a mutation in serine(133)), enabled a direct assessment, in vitro and in vivo, of the role of CREB in tumor progression. In this work, we demonstrate both in vitro and in vivo that CREB controls hepatocellular carcinoma growth, supports angiogenesis, and renders resistance to apoptosis. Along with the identification, by DNA microarray, of the CREB-regulated genes in normoxia and hypoxia, this work demonstrates for the first time that in parallel to other hypoxia responsive mechanisms, CREB plays an important role in hepatocellular carcinoma tumor progression.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Neoplasias Hepáticas Experimentais/patologia , Sequência de Aminoácidos , Animais , Apoptose , Divisão Celular , Hipóxia Celular , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Mol Ther ; 8(2): 342-50, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12907157

RESUMO

The major advantages of "naked DNA gene therapy" are its simplicity and a low or negligible immune response. Gene delivery by DNA electroporation (EP) involves injection of DNA and the application of a brief electric pulse to enhance cellular permeability. Although EP is an efficient gene transduction technique in rodents, it requires much higher voltages (>500 V) in larger animals, and hence, in practice it would be hazardous for human patients, as it would cause serious tissue damage. To overcome the obstacles associated with EP-mediated gene delivery in vivo, we developed a new method of gene transduction that uses laser energy. The femtosecond infrared titanium sapphire laser beam was developed specifically for enhancing in vivo gene delivery without risks of tissue damage. System optimization revealed that injection of 10 micro g naked DNA into the tibial muscle of mice followed by application of the laser beam for 5 s, focused to 2 mm depth upon an area of 95 x 95 micro m(2), resulted in the highest intensity and duration of gene expression with no histological or biochemical evidence of muscle damage. We assessed the potential clinical application of LBGT technology by using it to transfer the murine erythropoietin (mEpo) gene into mice. LBGT-mediated mEpo gene delivery resulted in elevated (>22%) hematocrit levels that were sustained for 8 weeks. Gene expression following LBGT was detected for >100 days. Hence, LBGT is a simple, safe, effective, and reproducible method for therapeutic gene delivery with significant clinical potential.


Assuntos
Terapia Genética/instrumentação , Terapia Genética/métodos , Raios Infravermelhos , Lasers , Animais , DNA/administração & dosagem , DNA/genética , Relação Dose-Resposta a Droga , Eletroporação/instrumentação , Eletroporação/métodos , Eritropoetina/genética , Expressão Gênica , Genes Reporter/genética , Terapia Genética/efeitos adversos , Camundongos , Fatores de Tempo , Transformação Genética
19.
Vaccine ; 21(23): 3169-78, 2003 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-12804845
20.
J Med Virol ; 69(4): 560-7, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12601765

RESUMO

Influenza and its complications account for substantial morbidity and mortality among young adults and especially among the elderly. In young adults, immunization provides 70-90% protection, while among the elderly the vaccine may be only 30-40% effective; hence the need for new, more immunogenic vaccines. We compared the safety and immunogenicity of a novel IL-2-supplemented liposomal influenza vaccine (designated INFLUSOME-VAC) with that of a commercial subunit vaccine and a commercial split virion vaccine in young adults (mean age 28 years) in the winter of 1999-2000. Seventy-three healthy young adults were randomly assigned to be vaccinated intramuscularly with the following: a commercial subunit vaccine (n = 17, group A), INFLUSOME-VAC (n = 36, group B), and a commercial split virion vaccine (n = 20, group C). The three vaccines contained equal amounts of hemagglutinin (approximately 15 microg each) from the strains A/Sydney (H3N2), A/Beijing (H1N1), and B/Yamanashi. INFLUSOME-VAC induced higher geometric mean HI titers and higher-fold increases in HI titers against all three strains, compared with the two commercial vaccines. In addition, seroconversion rates for the A/Sydney and B/Yamanashi strains were significantly higher (P < 0.05) compared with the split virion vaccine, and significantly higher for the three strains compared with the subunit vaccine (69-97% vs 35-65%, P < or = 0.02). Moreover, the anti-neuraminidase response was significantly greater (P = 0.05) in group B vs group A. INFLUSOME-VAC caused mild local pain at the injection site in a significantly higher proportion of the vaccinees (83%). Thus, INFLUSOME-VAC is an immunogenic and safe vaccine in young adults.


Assuntos
Vírus da Influenza A/imunologia , Vírus da Influenza B/imunologia , Vacinas contra Influenza , Influenza Humana/prevenção & controle , Adolescente , Adulto , Animais , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Lipossomos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neuraminidase/imunologia , Proteínas Recombinantes/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA