Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Allergol Immunopathol (Madr) ; 51(1): 110-115, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36617829

RESUMO

Asthma is a chronic airway disease. Allergic reactions and T helper (h)2 immune response play a key role in asthma occurrence. Cell therapy can control inflammation and remodeling responses in allergic asthma, and cytokines can change this effect. Therefore, in this study, the effect of treated cell therapy with IL-2 to control allergic asthma was studied. Bone marrow cells were extracted and co-cultured with IL-2 and the cells were used via intra-tracheal administration in allergic asthma mice. Levels of IL-4, IL-5, IL-13, Leukotriene B4 and C4, and remodeling factors were measured. At least, a histopathology test of lung tissue was done. Type2 cytokines, leukotrienes, remodeling factors, mucus secretion, goblet cell hyperplasia, peri-bronchial and peri-vascular inflammation were significantly (p˂0.05) decreased by treating with bone marrow-derived mononuclear cells (BMDMCs) and IL-2-BMDMCs. Treatment with IL-2-BMDMCs could significantly decrease IL-13, transforming growth factor (TGF)-ß, HP levels, and mucus secretion (p˂0.05) compared to BMDMCs treatment. In this study, BMDMCs and IL-2-BMDMCs therapy could decrease inflammation, allergic, and remodeling factors in allergic asthma. Cell therapy with BMDMCs had a strong and notable effect on the control of allergic asthma pathophysiology when co-cultured and used with IL-2.


Assuntos
Asma , Hipersensibilidade , Interleucina-2 , Leucócitos Mononucleares , Animais , Camundongos , Asma/patologia , Medula Óssea , Líquido da Lavagem Broncoalveolar , Citocinas/metabolismo , Modelos Animais de Doenças , Hipersensibilidade/patologia , Inflamação/patologia , Interleucina-13 , Interleucina-2/farmacologia , Pulmão/patologia , Camundongos Endogâmicos BALB C , Ovalbumina , Fator de Crescimento Transformador beta
2.
Allergol. immunopatol ; 51(1): 110-115, ene. 2023. graf, ilus
Artigo em Inglês | IBECS | ID: ibc-214026

RESUMO

Asthma is a chronic airway disease. Allergic reactions and T helper (h)2 immune response play a key role in asthma occurrence. Cell therapy can control inflammation and remodeling responses in allergic asthma, and cytokines can change this effect. Therefore, in this study, the effect of treated cell therapy with IL-2 to control allergic asthma was studied. Bone marrow cells were extracted and co-cultured with IL-2 and the cells were used via intra-tracheal administration in allergic asthma mice. Levels of IL-4, IL-5, IL-13, Leukotriene B4 and C4, and remodeling factors were measured. At least, a histopathology test of lung tissue was done. Type2 cytokines, leukotrienes, remodeling factors, mucus secretion, goblet cell hyperplasia, peri-bronchial and peri-vascular inflammation were significantly (p˂0.05) decreased by treating with bone marrow-derived mononuclear cells (BMDMCs) and IL-2-BMDMCs. Treatment with IL-2-BMDMCs could significantly decrease IL-13, transforming growth factor (TGF)-β, HP levels, and mucus secretion (p˂0.05) compared to BMDMCs treatment. In this study, BMDMCs and IL-2-BMDMCs therapy could decrease inflammation, allergic, and remodeling factors in allergic asthma. Cell therapy with BMDMCs had a strong and notable effect on the control of allergic asthma pathophysiology when co-cultured and used with IL-2 (AU)


Assuntos
Animais , Masculino , Camundongos , Asma/imunologia , Asma/terapia , Imunomodulação , Interleucina-2/uso terapêutico , Células da Medula Óssea , Modelos Animais de Doenças , Camundongos Endogâmicos BALB C
3.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 34(4): 362-366, 2022 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-35692199

RESUMO

OBJECTIVE: To study the effect of different crystalloid resuscitation on renal function in septic shock rabbits, and to provide a theoretical basis for the choice of crystalloid for clinical fluid resuscitation. METHODS: Thirty-six healthy male New Zealand white rabbits were divided into six groups by random number table: control group, model group, and four crystalloid groups including normal saline (NS) group, lactate Ringer solution (LR) group, acetate Ringer solution (AR) group, and sodium potassium magnesium calcium glucose injection (SPMCG) group, with 6 rabbits in each group. Rabbits were infused with Escherichia coli lipopolysaccharide (LPS) 500 µg/kg via the marginal ear vein (infused at a constant speed within 20 minutes), and then continued to infuse in an increase of 300 µg/kg every 10 minutes, the maximum dose was 2 mg/kg, until the mean arterial pressure (MAP) dropped to 60% of the basal value, the septic shock model was considered to be successfully reproduced. The rabbits in the control group were not injected with LPS, and other operations were the same as in the model group. Different crystalloid groups were given crystal solution immediately after modeling for resuscitation (predetermined fluid volume 60 mL/kg, transfusion within 3 hours). The volume stress test was performed every hour to guide the fluid volume, and the stroke volume index increase rate (ΔSVI) < 15% was the end point of resuscitation. The control group and the model group were given NS 4 mL×kg-1×h-1 to maintain the physiological requirement. All groups were given tracheotomy and mechanical ventilation, and the hemodynamic changes were monitored by pulse-indicated continuous cardiac output (PiCCO). The dynamic changes of hemodynamic indexes, arterial blood gas analysis, electrolytes, blood glucose and renal function biomarkers were monitored before modeling, immediately after modeling and 3, 6, and 12 hours after resuscitation. RESULTS: (1) Hemodynamic indicators: after modeling, the MAP in the model group and the four fluid resuscitation groups decreased significantly, the cardiac index (CI) increased, and the systemic vascular resistance index (SVRI), global end-diastolic volumn index (GEDVI) decreased. After different crystalloid resuscitation at different time points, MAP, SVRI, and GEDVI increased in the four crystalloid groups. (2) Arterial blood gas analysis, electrolytes, blood glucose: blood lactic acid (Lac) in the model group and the four fluid resuscitation groups increased after model success. After fluid resuscitation, the Lac of each crystalloid group began to decrease and reached to the lowest at 12 hours. Compared with the LR, AR and SPMCG groups, the pH value decreased in the NS group at 6 hours and 12 hours of fluid resuscitation (6 hours: 7.29±0.00 vs. 7.40±0.02, 7.35±0.02, 7.37±0.02; 12 hours: 7.27±0.02 vs. 7.38±0.02, 7.39±0.02, 7.35±0.01; all P < 0.05). After fluid resuscitation, blood Cl- levels at 3, 6, and 12 hours in the NS group were significantly higher than those in the LR, AR and SPMCG groups (mmol/L: 113.4±0.6 vs. 101.4±3.6, 108.0±1.1, 106.0±0.8 at 3 hours; 115.1±2.0 vs. 101.1±2.7, 109.0±2.2, 105.3±0.6 at 6 hours; 116.9±0.1 vs. 104.2±4.4, 107.6±1.7, 108.7±0.6 at 12 hours; all P < 0.05). There was no significant difference in blood glucose at each time point among the four crystalloid groups. (3) Biomarkers of renal function: blood and urine neutrophil gelatinase associated lipocalin (NGAL) and cystatin C (Cys C) were significantly increased in the model group and four fluid resuscitation groups. After fluid resuscitation, blood, urine NGAL and Cys C decreased. There was no significant difference in blood, urine NGAL and Cys C at all the time points among the different fluid resuscitation groups. CONCLUSIONS: In the rabbit model of septic shock induced by Escherichia coli LPS, hyperchloremia and acidosis occurred after NS resucitation, but did not occur during the recovery of LR, AR and SPMCG. There was no difference in the effects of different crystalloid resuscitation on renal function in septic shock rabbits.


Assuntos
Choque Séptico , Animais , Glicemia , Débito Cardíaco , Soluções Cristaloides/uso terapêutico , Escherichia coli , Hidratação , Rim/fisiologia , Lipocalina-2 , Lipopolissacarídeos , Masculino , Coelhos , Ressuscitação , Solução de Ringer/uso terapêutico , Choque Séptico/tratamento farmacológico
4.
Heart Lung Circ ; 23(1): 75-81, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23891309

RESUMO

BACKGROUND: c-kit-positive cardiac progenitor cells (CPCs) have been proven suitable for stem cell therapy. CPCs marker c-kit and its ligand, the stem cell factor (SCF), are associated with the functions of proliferation and differentiation. In our previous study, we found that stromal cell-derived factor-1α (SDF-1α) could enhance the expression of c-kit. However, the mechanism is unknown. METHODS AND RESULTS: CPCs were isolated from adult mouse hearts, and c-kit-positive CPCs were purified by magnetic-activated c-kit cell sorting magnetic beads. The cells were cultured with SDF-1α, c-kit expression was measured by western blotting and qPCR, the proliferation and migration of cells were measured by CCK-8 and transwell assay, DNA methyltransferase (DNMT) mRNA were measured by qPCR, global DNMT activity was measured by DNMT activity assay kit, and DNA methylation was analysed using Sequenom's MassARRAY platform. Results showed that SDF-1α could enhance the expression of c-kit, which results in the promoting of c-kit-positive CPCs proliferation and migration. SDF-1α stimulation inhibited the expression of DNMT1, DNMT3ß, and global DNMT activity, which led to significant demethylation in c-kit-positive CPCs. CONCLUSIONS: SDF-1α signalling, via CXCR4 activation, up-regulated c-kit expression by inhibiting DNMT1 and DNMT3ß expression and global DNMT activity, and by subsequent demethylation of the c-kit gene.


Assuntos
Quimiocina CXCL12/metabolismo , Regulação da Expressão Gênica/fisiologia , Miocárdio/metabolismo , Proteínas Proto-Oncogênicas c-kit/biossíntese , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Animais , Movimento Celular/fisiologia , Proliferação de Células , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/fisiologia , Camundongos , Miocárdio/citologia , Células-Tronco/citologia
5.
PLoS One ; 8(12): e81505, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24312554

RESUMO

OBJECTIVES: Stem cell preconditioning (PC) is a powerful approach in reducing cell death after transplantation. We hypothesized that PC human endothelial progenitor cells (hEPCs) with bradykinin (BK) enhance cell survival, inhibit apoptosis and repair the infarcted myocardium. METHODS: The hEPCs were preconditioned with or without BK. The hEPCs apoptosis induced by hypoxia along with serum deprivation was determined by annexin V-fluorescein isothiocyanate/ propidium iodide staining. Cleaved caspase-3, Akt and eNOS expressions were determined by Western blots. Caspase-3 activity and vascular endothelial growth factor (VEGF) levels were assessed in hEPCs. For in vivo studies, the survival and cardiomyocytes apoptosis of transplanted hEPCs were assessed using 1,1'-dioctadecyl-3,3,3',3'-tetramethylindodi- carbocyanine,4-chlorobenzenesul-fonate salt labeled hEPCs and TUNEL staining. Infarct size and cardiac function were measured at 10 days after transplantation, and the survival of transplanted hEPCs were visualized using near-infrared optical imaging. RESULTS: In vitro data showed a marked suppression in cell apoptosis following BK PC. The PC reduced caspase-3 activation, increased the Akt, eNOS phosphorylation and VEGF levels. In vivo data in preconditioned group showed a robust cell anti-apoptosis, reduction in infarct size, and significant improvement in cardiac function. The effects of BK PC were abrogated by the B2 receptor antagonist HOE140, the Akt and eNOS antagonists LY294002 and L-NAME, respectively. CONCLUSIONS: The activation of B2 receptor-dependent PI3K/Akt/eNOS pathway by BK PC promotes VEGF secretion, hEPC survival and inhibits apoptosis, thereby improving cardiac function in vivo. The BK PC hEPC transplantation for stem cell-based therapies is a novel approach that has potential for clinical used.


Assuntos
Bradicinina/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/citologia , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
PLoS One ; 8(7): e69134, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894420

RESUMO

BACKGROUND: Cardiac progenitor cells (CPCs) have been proven suitable for stem cell therapy after myocardial infarction, especially c-kit(+)CPCs. CPCs marker c-kit and its ligand, the stem cell factor (SCF), are linked as c-kit/SCF axis, which is associated with the functions of proliferation and differentiation. In our previous study, we found that stromal cell-derived factor-1α (SDF-1α) could enhance the expression of c-kit. However, the mechanism is unknown. METHODS AND RESULTS: CPCs were isolated from adult mouse hearts, c-kit(+) and c-kit(-) CPCs were separated by magnetic beads. The cells were cultured with SDF-1α and CXCR4-selective antagonist AMD3100, and c-kit expression was measured by qPCR and Western blotting. Results showed that SDF-1α could enhance c-kit expression of c-kit(+)CPCs, made c-kit(-)CPCs expressing c-kit, and AMD3100 could inhibit the function of SDF-1α. After the intervention of SDF-1α and AMD3100, proliferation and migration of CPCs were measured by CCK-8 and transwell assay. Results showed that SDF-1α could enhance the proliferation and migration of both c-kit(+) and c-kit(-) CPCs, and AMD3100 could inhibit these functions. DNA methyltransferase (DNMT) mRNA were measured by qPCR, DNMT activity was measured using the DNMT activity assay kit, and DNA methylation was analyzed using Sequenom's MassARRAY platform, after the CPCs were cultured with SDF-1α. The results showed that SDF-1α stimulation inhibited the expression of DNMT1 and DNMT3ß, which are critical for the maintenance of regional DNA methylation. Global DNMT activity was also inhibited by SDF-1α. Lastly, SDF-1α treatment led to significant demethylation in both c-kit(+) and c-kit(-) CPCs. CONCLUSIONS: SDF-1α combined with CXCR4 could up-regulate c-kit expression of c-kit(+)CPCs and make c-kit(-)CPCs expressing c-kit, which result in the CPCs proliferation and migration ability improvement, through the inhibition of DNMT1 and DNMT3ß expression and global DNMT activity, as well as the subsequent demethylation of the c-kit gene.


Assuntos
Quimiocina CXCL12/farmacologia , Epigênese Genética/efeitos dos fármacos , Miocárdio/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Animais , Western Blotting , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Epigênese Genética/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Contraste de Fase , Reação em Cadeia da Polimerase em Tempo Real , Células-Tronco/citologia
7.
PLoS One ; 7(7): e37948, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22815687

RESUMO

BACKGROUND: Cardiac progenitor cells (CPCs) have been shown to be suitable in stem cell therapy for resurrecting damaged myocardium, but poor retention of transplanted cells in the ischemic myocardium causes ineffective cell therapy. Hypoxic preconditioning of cells can increase the expression of CXCR4 and pro-survival genes to promote better cell survival; however, it is unknown whether hypoxia preconditioning will influence the survival and retention of CPCs via the SDF-1α/CXCR4 axis. METHODS AND RESULTS: CPCs were isolated from adult mouse hearts and purified by magnetic activated cell sorting using c-kit magnetic beads. These cells were cultured at various times in either normoxic or hypoxic conditions, and cell survival was analyzed using flow cytometry and the expression of hypoxia-inducible factor-1α (HIF-1α), CXCR4, phosphorylated Akt and Bcl-2 were measured by Western blot. Results showed that the expression of pro-survival genes significantly increased after hypoxia treatment, especially in cells cultured in hypoxic conditions for six hours. Upon completion of hypoxia preconditioning from c-kit+ CPCs for six hours, the anti-apoptosis, migration and cardiac repair potential were evaluated. Results showed a significant enhancement in anti-apoptosis and migration in vitro, and better survival and cardiac function after being transplanted into acute myocardial infarction (MI) mice in vivo. The beneficial effects induced by hypoxia preconditioning of c-kit+ CPCs could largely be blocked by the addition of CXCR4 selective antagonist AMD3100. CONCLUSIONS: Hypoxic preconditioning may improve the survival and retention of c-kit+ CPCs in the ischemic heart tissue through activating the SDF-1α/CXCR4 axis and the downstream anti-apoptosis pathway. Strategies targeting this aspect may enhance the effectiveness of cell-based cardiac regenerative therapy.


Assuntos
Quimiocina CXCL12/metabolismo , Precondicionamento Isquêmico Miocárdico/métodos , Miocárdio/patologia , Receptores CXCR4/metabolismo , Células-Tronco/metabolismo , Células-Tronco/patologia , Animais , Apoptose , Hipóxia Celular , Movimento Celular , Sobrevivência Celular , Terapia Baseada em Transplante de Células e Tecidos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Fenótipo
8.
Hum Gene Ther ; 23(8): 859-70, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22435954

RESUMO

Tissue kallikrein (TK) has been demonstrated to improve neovasculogenesis after myocardial infarction (MI). In the present study, we examined the role and underlying mechanisms of TK in peripheral endothelial progenitor cell (EPC) function. Peripheral blood-derived mononuclear cells containing EPCs were isolated from rat. The in vitro effects of TK on EPC differentiation, apoptosis, migration, and vascular tube formation capacity were studied in the presence or absence of TK, kinin B(2) receptor antagonist (icatibant), and phosphatidylinositol-3 kinase inhibitor (LY294002). Apoptosis was evaluated by flow-cytometry analysis using Annexin V-FITC/PI staining, as well as western-blot analysis of Akt phosphorylation and cleaved caspase-3. Using an MI mouse model, we then examined the in vivo effects of human TK gene adenoviral vector (Ad.hTK) administration on the number of CD34(+)Flk-1(+) progenitors in the peripheral circulation, heart tissue, extent of vasculogenesis, and heart function. Administration of TK significantly increased the number of Dil-LDL/UEA-lectin double-positive early EPCs, as well as their migration and tube formation properties in vitro. Transduction of TK in cultured EPCs attenuated apoptosis induced by hypoxia and led to an increase in Akt phosphorylation and a decrease in cleaved caspase-3 levels. The beneficial effects of TK were blocked by pretreatment with icatibant and LY294002. The expression of recombinant human TK in the ischemic mouse heart significantly improved cardiac contractility and reduced infarct size 7 days after gene delivery. Compared with the Ad.Null group, Ad.hTK reduced mortality and preserved left ventricular function by increasing the number of CD34(+)Flk-1(+) EPCs and promoting the growth of capillaries and arterioles in the peri-infarct myocardium. These data provide direct evidence that TK promotes vessel growth by increasing the number of EPCs and enhancing their functional properties through the kinin B(2) receptor-Akt signaling pathway.


Assuntos
Células Endoteliais/patologia , Miócitos Cardíacos/patologia , Neovascularização Fisiológica , Células-Tronco/patologia , Calicreínas Teciduais/metabolismo , Adenoviridae/genética , Animais , Complexo CD3/genética , Complexo CD3/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Diferenciação Celular , Movimento Celular , Células Cultivadas , Células Endoteliais/metabolismo , Vetores Genéticos , Humanos , Masculino , Camundongos , Contração Miocárdica , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , Células-Tronco/metabolismo , Calicreínas Teciduais/genética
9.
Int J Nanomedicine ; 7: 641-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22334786

RESUMO

OBJECTIVE: Mural inflammation has been shown to contribute to the development of plaque, with the α(V)ß(3) integrin highly expressed in atherosclerotic plaques. We herein examined α(V)ß(3) integrin expression as a function of carotid atherosclerosis formation in the apolipoprotein E-deficient (apoE(-/-)) mouse. METHODS AND RESULTS: Constrictive collars were placed around the left common carotid arteries of apo E(-/-) mice maintained on a high-fat diet (n = 14). Before and 21 days following collar placement, in vivo serial magnetic resonance imaging (MRI) measurements of the carotid aortic diameter were performed using a 7T magnetic resonance (MR) scanner. Near- infrared fluorescence (NIRF) imaging was performed (n = 6) using an in vivo imaging system 0-24 hours following administration of 1.0 nmol c(RGDyK)-Cy5.5 via the tail vein. A competition experiment was performed by the co-injection of a saturating dose of bicyclic RGD peptide H-Glu[cyclo(Arg-Gly-Asp-D-Tyr-Lys)]2 (n = 3). Following image acquisition and sacrifice at 24 hours after injection, carotid arteries were harvested for histological analyses. Neointima formation and arterial remodeling in the carotid arteries of apoE(-/-) mice were induced by the placement of a constrictive collar. Significantly greater fluorescent signals were obtained from constrictive collar left common carotid arteries as compared to uninvolved aortic segments in constrictive collar mice. Binding to stenotic lesions was efficiently blocked in competition experiments. Immunostaining confirmed the presence of mural α(V)ß(3) integrin expression in macrophages in the neointima. Signal intensity increased in a macrophage density-dependent fashion in the stenotic segments. CONCLUSION: Mural α(V)ß(3) integrin expression, as determined using RGD-Cy5.5 near-infrared optical imaging, was increased in carotid arteries with constrictive collars in experimental mice. This expression can estimate the macrophage-bound inflammatory activity of atherosclerotic lesions.


Assuntos
Doenças das Artérias Carótidas/metabolismo , Integrina alfaVbeta3/biossíntese , Análise de Variância , Animais , Estenose da Valva Aórtica/metabolismo , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/patologia , Estenose das Carótidas/metabolismo , Estenose das Carótidas/patologia , Dieta Hiperlipídica , Histocitoquímica , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neointima/metabolismo , Espectroscopia de Luz Próxima ao Infravermelho
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...