Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Eur J Cardiothorac Surg ; 61(3): 666-674, 2022 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-34528682

RESUMO

OBJECTIVES: The surgical treatment for diseases of the descending aorta is related to a high mortality rate because of the activation of a systemic inflammatory process due to ischaemia and reperfusion (I/R) injury. Activation of coagulation can contribute to the inflammatory process, resulting in microcirculatory damage and multiple organ failure. Our goal was to evaluate the role of prophylactic intravenous 17ß-oestradiol (E2) in coagulation, the inflammatory response and hepatic injury after occlusion of the descendent proximal aorta in male rats. METHODS: Wistar male rats were randomized and allocated to 3 groups (n = 8 per group): sham, surgically manipulated; IR, animals subjected to I/R; and E2, animals treated with E2 (280 µg/kg, intravenously) before I/R. I/R was induced by insertion of a 2-Fr Fogarty arterial embolectomy catheter in the descending aorta, which was occluded for 20 min, followed by a reperfusion period of 2 h. Serological markers, platelet aggregation, hepatic vascular flow, systemic and liver inflammatory response and apoptosis were analysed. The coagulation process was evaluated by thromboelastometry. RESULTS: The aortic occlusion led to a reduction in plasma fibrinogen concentration in parallel with increased clotting time, greater clot firmness and reduced lysis. E2 treatment was able to increase fibrinogen, prevent the increase in clotting time and normalize clot firmness, but it exerted only a mild effect on clot lysis. Platelet aggregation was increased by IR, and E2 treatment was able to reduce it. There was a reduction in flow percentage in the IR group that was not prevented by E2. In parallel, higher aggregate formation was observed in the vessels of the IR group of animals. There was increased systemic release of interleukin-1-ß, interleukin-6 and interleukin-10 in the IR group, which was reduced in the treated animals. CONCLUSIONS: The current results suggest that pretreatment with E2 before an ischaemic period induced by occlusion of the proximal descending aorta is effective in preventing alterations in coagulation and systemic inflammation due to I/R injury.


Assuntos
Aorta Torácica , Traumatismo por Reperfusão , Animais , Aorta Torácica/cirurgia , Estradiol/farmacologia , Estradiol/uso terapêutico , Humanos , Inflamação/prevenção & controle , Masculino , Microcirculação , Ratos , Ratos Wistar , Traumatismo por Reperfusão/prevenção & controle
2.
Clinics (Sao Paulo) ; 76: e3042, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34406272

RESUMO

OBJECTIVES: Lung transplantation is limited by the systemic repercussions of brain death (BD). Studies have shown the potential protective role of 17ß-estradiol on the lungs. Here, we aimed to investigate the effect of estradiol on the long-lasting lung inflammatory state to understand a possible therapeutic application in lung donors with BD. METHODS: Female Wistar rats were separated into 3 groups: BD, subjected to brain death (6h); E2-T0, treated with 17ß-estradiol (50 µg/mL, 2 mL/h) immediately after brain death; and E2-T3, treated with 17ß-estradiol (50 µg/ml, 2 ml/h) after 3h of BD. Complement system activity and macrophage presence were analyzed. TNF-α, IL-1ß, IL-10, and IL-6 gene expression (RT-PCR) and levels in 24h lung culture medium were quantified. Finally, analysis of caspase-3 gene and protein expression in the lung was performed. RESULTS: Estradiol reduced complement C3 protein and gene expression. The presence of lung macrophages was not modified by estradiol, but the release of inflammatory mediators was reduced and TNF-α and IL-1ß gene expression were reduced in the E2-T3 group. In addition, caspase-3 protein expression was reduced by estradiol in the same group. CONCLUSIONS: Brain death-induced lung inflammation in females is modulated by estradiol treatment. Study data suggest that estradiol can control the inflammatory response by modulating the release of mediators after brain death in the long term. These results strengthen the idea of estradiol as a therapy for donor lungs and improving transplant outcomes.


Assuntos
Morte Encefálica , Pneumonia , Animais , Estradiol/farmacologia , Estrogênios , Feminino , Ratos , Ratos Wistar
3.
Ann Transl Med ; 9(14): 1125, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34430566

RESUMO

BACKGROUND: Clinical and experimental data highlight the consequences of brain death on the quality of organs and demonstrate the importance of donor state to the results of transplantation. Female rats show higher cardio-pulmonary injury linked to decreased concentrations of female sex hormones after brain-dead (BD). This study evaluated the effect of 17ß-estradiol on brain death induced renal injury in female rats. METHODS: Female Wistar rats were randomically allocated into 4 groups: false-operation (Sham), BD, treatment with 17ß-estradiol (50 µg/mL, 2 mL/h) 3 h after brain death (E2-T3), or immediately after brain death confirmation (E2-T0). Creatinine, urea, cytokines, and complement system components were quantified. Renal injury markers, such as KIM-1, Caspase-3, BCL-2 and MMP2/9 were evaluated. RESULTS: Brain death leads to increased kidney KIM-1 expression and longer 17ß-estradiol treatment resulted in downregulation (P<0.0001). There was increase of neutrophil numbers in kidney from BD rats and E2 treatment was able to reduce it (P=0.018). Regarding complement elements, E2-T3 group evidenced E2 therapeutic effects, reducing C5b-9 (P=0.0004), C3aR (P=0.054) and C5aR (P=0.019). In parallel, there were 17ß-estradiol effects in reducing MMP2 (P=0.0043), MMP9 (P=0.011), and IL-6 (P=0.024). Moreover, E2-T3 group improved renal function in comparison to BD group (P=0.0938). CONCLUSIONS: 17ß-estradiol treatment was able to reduce acute kidney damage in BD female rats owing to its ability to prevent tissue damage, formation of C5b-9, and local synthesis of inflammatory mediators.

4.
Transplantation ; 105(4): 775-784, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33031230

RESUMO

BACKGROUND: Brain death (BD) affects the viability of lungs for transplantation. A correlation exists between high-lung inflammation after BD and the decrease in female sex hormones, especially estradiol. Therefore, we investigated the effects of 17ß-estradiol (E2) treatment on the lungs of female brain dead rats. METHODS: Female Wistar rats were divided into 4 groups: BD (submitted to BD for 6 h), sham (false operated), E2-T0 (treated with E2 immediately after BD; 50 µg/mL, 2 mL/h), and E2-T3 (treated with E2 after 3 h of BD; 50 µg/mL, 2 mL/h). Lung edema, hemorrhage, and leukocyte infiltration were analyzed. Adhesion molecules were evaluated, and analysis of NO synthase gene and protein expression was performed using real-time PCR and immunohistochemistry, respectively. Release of chemokines and matrix degradation in the lungs was analyzed. RESULTS: BD increased leukocyte infiltration, as shown by intravital microscopy (P = 0.017), bronchoalveolar lavage cell count (P = 0.016), the release of inflammatory mediators (P = 0.02), and expression of adhesion molecules. BD also increased microvascular permeability and the expression and activity of matrix metalloproteinase-9 in the lungs. E2 treatment reduced leukocyte infiltration, especially in the E2-T3 group, release of inflammatory mediators, adhesion molecules, and matrix metalloproteinase activity in the lungs. CONCLUSIONS: E2 treatment was successful in controlling the lung inflammatory response in females submitted to BD. Our results suggest that E2 directly decreases the release of chemokines, restraining cell traffic into the lungs. Thus, E2 has a therapeutic potential, and its role in improving donor lung quality should be explored further.


Assuntos
Anti-Inflamatórios/farmacologia , Morte Encefálica , Estradiol/farmacologia , Pulmão/efeitos dos fármacos , Pneumonia/prevenção & controle , Animais , Permeabilidade Capilar/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Citoproteção , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Leucócitos/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Metaloproteinase 9 da Matriz/metabolismo , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Edema Pulmonar/imunologia , Edema Pulmonar/metabolismo , Edema Pulmonar/patologia , Edema Pulmonar/prevenção & controle , Ratos Wistar , Técnicas de Cultura de Tecidos
5.
Clinics ; 76: e3042, 2021. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1286068

RESUMO

OBJECTIVES: Lung transplantation is limited by the systemic repercussions of brain death (BD). Studies have shown the potential protective role of 17β-estradiol on the lungs. Here, we aimed to investigate the effect of estradiol on the long-lasting lung inflammatory state to understand a possible therapeutic application in lung donors with BD. METHODS: Female Wistar rats were separated into 3 groups: BD, subjected to brain death (6h); E2-T0, treated with 17β-estradiol (50 μg/mL, 2 mL/h) immediately after brain death; and E2-T3, treated with 17β-estradiol (50 μg/ml, 2 ml/h) after 3h of BD. Complement system activity and macrophage presence were analyzed. TNF-α, IL-1β, IL-10, and IL-6 gene expression (RT-PCR) and levels in 24h lung culture medium were quantified. Finally, analysis of caspase-3 gene and protein expression in the lung was performed. RESULTS: Estradiol reduced complement C3 protein and gene expression. The presence of lung macrophages was not modified by estradiol, but the release of inflammatory mediators was reduced and TNF-α and IL-1β gene expression were reduced in the E2-T3 group. In addition, caspase-3 protein expression was reduced by estradiol in the same group. CONCLUSIONS: Brain death-induced lung inflammation in females is modulated by estradiol treatment. Study data suggest that estradiol can control the inflammatory response by modulating the release of mediators after brain death in the long term. These results strengthen the idea of estradiol as a therapy for donor lungs and improving transplant outcomes.


Assuntos
Animais , Feminino , Ratos , Pneumonia , Morte Encefálica , Ratos Wistar , Estradiol/farmacologia , Estrogênios
6.
Transpl Int ; 33(11): 1541-1550, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32890430

RESUMO

Brain death (BD) leads to a systemic inflammation associated with the activation of coagulation, which could be related to decreased microcirculatory perfusion. Evidence shows that females exhibit higher platelet aggregability than males. Thus, we investigated sex differences in platelets, coagulation and microcirculatory compromise after BD. BD was induced in male and female (proestrus) Wistar rats. After 3 h, we evaluated: (i) intravital microscopy to evaluate mesenteric perfusion and leucocyte infiltration; (ii) platelet aggregation assay; (iii) rotational thromboelastometry; and (iv) Serum NOx- . Female rats maintained the mesenteric perfusion, whereas male reduced percentage of perfused vessels. Male BD presented higher platelet aggregation than the controls. In contrast, female BD had lower platelet aggregation than the control. Thromboelastometry indicated a reduction in clot firmness with increased clotting time in the female group compared with the male group. Serum NOx- level in female BD was higher than that in the male BD and female control. There is sex dimorphism in platelet function and clotting process, which are altered in different ways by BD. Thus, it is possible to connect the reduction in microcirculatory perfusion in males to intravascular microthrombi formation and the maintenance of perfusion in females to a higher inflammatory response and NO synthesis.


Assuntos
Morte Encefálica , Caracteres Sexuais , Animais , Feminino , Masculino , Microcirculação , Perfusão , Ratos , Ratos Wistar
7.
Clinics (Sao Paulo) ; 75: e1980, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32490931

RESUMO

Considering that female sexual hormones may modulate the inflammatory response and also exhibit direct effects on the cells of the immune system, herein, we intend to discuss the sex differences and the role of estradiol in modulating the lung and systemic inflammatory response, focusing on its possible application as a treatment modality for SARS-CoV-2 patients. COVID-19 patients develop severe hypoxemia early in the course of the disease, which is silent most of the time. Small fibrinous thrombi in pulmonary arterioles and a tumefaction of endothelial were observed in the autopsies of fatal COVID-19 cases. Studies showed that the viral infection induces a vascular process in the lung, which included vasodilation and endothelial dysfunction. Further, the proportions of CD4+ T and CD8+ T lymphocytes were strongly reduced in patients with severe SARS-CoV-2 infection. Estradiol is connected with CD4+ T cell numbers and increases T-reg cell populations, affecting immune responses to infection. It is known that estradiol exerts a protective effect on endothelial function, activating the generation of nitric oxide (NO) via endothelial nitric oxide synthase. Estrogen attenuates the vasoconstrictor response to various stimuli and induces vasodilation in the pulmonary vasculature during stress situations like hypoxia. It exerts a variety of rapid actions, which are initiated after its coupling with membrane receptors, which in turn, may positively modulate vascular responses in pulmonary disease and help to maintain microvascular flow. Direct and indirect mechanisms underlying the effects of estradiol were investigated, and the results point to a possible protective effect of estradiol against COVID-19, indicating that it may be considered as an adjuvant therapeutic element for the treatment of patients affected by the novel coronavirus.


Assuntos
Betacoronavirus , Infecções por Coronavirus/terapia , Estradiol/uso terapêutico , Imunidade Inata , Inflamação/virologia , Pneumonia Viral/terapia , Animais , COVID-19 , Feminino , Humanos , Inflamação/tratamento farmacológico , Masculino , Pandemias , Ratos , SARS-CoV-2 , Fatores Sexuais
8.
Pulm Pharmacol Ther ; 61: 101901, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32044433

RESUMO

BACKGROUND: Lung transplantation is a treatment method for end stage lung disease, but the availability of donor lungs remains a major constraint. Brain death (BD) induces hemodynamic instability with microcirculatory hypoperfusion and increased inflammation, leading to pulmonary dysfunction. Hypertonic saline solution (HSS) is a volume expander possessing immunomodulatory effects. This study evaluated the influence of HSS on pulmonary dysfunction and inflammation in a rat model of BD. METHODS: BD was induced by inflation of an intracranial balloon catheter. Rats were divided into [1]: Sham, without BD [2]; NS, NaCl treatment (0.9%, 4 mL/kg, i.v.) immediately after BD [3]; HSS1, HSS treatment (NaCl 7.5%, 4 mL/kg, i.v.) immediately after BD; and [4] HSS60, HSS treatment 60 min post BD. All groups were analyzed after 360 min. RESULTS: Animals subjected to BD exhibited increased exhaled O2 and decreased CO2.The number of leukocytes in the lungs was significantly increased in the NS group (p = 0.002) and the HSS treatment was able to reduce it (HSS1, p = 0.018 and HSS60 = 0.030). In parallel, HSS-treated rats showed reduced levels of ICAM-1 expression, which was increased in the NS compared to Sham group. Lung edema was found increased in the NS group animals compared to Sham and no effect of the HSS treatment was observed. There were no differences among the groups in terms of TNF-α, VEGF, and CINC-1 lung concentrations. CONCLUSIONS: HSS is capable of reducing inflammatory cell infiltration into the lung after BD induction, which is associated with the reduction of ICAM-1 expression in organ vessels.


Assuntos
Morte Encefálica , Pulmão/fisiopatologia , Solução Salina Hipertônica/uso terapêutico , Animais , Pressão Arterial , Quimiocina CXCL1/metabolismo , Edema , Endotelina-1/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Transplante de Pulmão , Masculino , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
J Thorac Cardiovasc Surg ; 160(3): e135-e144, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31653422

RESUMO

OBJECTIVE: The study objective was to evaluate the effect of bilateral sympathectomy on ventricular remodeling and function in a rat model of dilated cardiomyopathy induced by doxorubicin. METHODS: Dilated cardiomyopathy was induced in male Wistar rats by weekly intraperitoneal injection of doxorubicin (2 mg/kg) for 9 weeks. Animals were divided into 4 groups: dilated cardiomyopathy; bilateral sympathectomy, submitted on day 15 of the protocol to bilateral sympathectomy; angiotensin-converting enzyme inhibitor, treated with enalapril through day 15 until the end of the experimental protocol; and sham, nonsubmitted through doxorubicin protocol, with weekly intraperitoneal injections of saline solution (0.9%). The left ventricular function was assessed, and the heart was collected for posterior analyses. RESULTS: The dilated cardiomyopathy group presented a significant decrease in the myocardial efficiency when compared with the sham group (33.4% vs 71.2%). Only the bilateral sympathectomy group was able to preserve it (57.5%; P = .0001). A significant dilatation in the left ventricular chamber was observed in the dilated cardiomyopathy group (15.9 µm2) compared with the sham group (10.2 µm2; P = .0053). Sympathectomy and enalapril prevented ventricular remodeling (9.5 and 9.6 µm2, respectively; P = .0034). There was a significant increase in interstitial myocardial fibrosis in the dilated cardiomyopathy group (14.8%) when compared with the sham group (2.4%; P = .0001). This process was significantly reduced with sympathectomy and enalapril (8.7 and 3.9%, respectively; P = .0001). CONCLUSIONS: Bilateral sympathectomy was effective in preventing remodeling and left ventricular dysfunction in a rat model of dilated cardiomyopathy induced by doxorubicin.


Assuntos
Cardiomiopatia Dilatada/induzido quimicamente , Cardiomiopatia Dilatada/cirurgia , Doxorrubicina/toxicidade , Simpatectomia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Modelos Animais de Doenças , Enalapril/administração & dosagem , Humanos , Masculino , Ratos , Ratos Wistar , Disfunção Ventricular Esquerda/prevenção & controle , Remodelação Ventricular
10.
Transpl Int ; 33(3): 279-287, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31701582

RESUMO

Organ donor's age negatively influences graft survival of organs, increasing risk of complications. Aging occurs in both men and women; however, the menopause marks a decrease in sex hormones and a sudden increase in the process of vascular aging. We investigated sex hormones' influence on the lung inflammatory process induced by BD in female rats. Wistar rats were grouped as: female rats from high estradiol to heat period (non-OVx) and ovariectomized (OVx) female rats. Ovariectomy was carried out 10 days before BD. BD was induced using intracranial balloon rapid inflation. Serum hormones and inflammatory mediators were quantified, leukocytes and platelets counted and lung samples were collected for RT-PCR, immunohistochemical, and histological analysis. Female sex hormones and corticosterone were reduced 6 h after BD in non-OVx group. The infiltration of leukocytes in female non-OVx lungs was higher compared to OVx. G-CSF, VEGF, and CINC-1 were found increased in non-OVx group serum in comparison to OVx. Lung mediators were increased in non-OVx rats compared to controls. The acute reduction of sex hormones induced by BD appears to have a worse effect on lung inflammation than a reduction that has happened over a prolonged period of time, allowing a physiological adaptation prior to BD.


Assuntos
Morte Encefálica , Pneumonia , Animais , Estradiol , Feminino , Hormônios Esteroides Gonadais , Pneumonia/etiologia , Ratos , Ratos Wistar
11.
Clinics ; 75: e1980, 2020. graf
Artigo em Inglês | LILACS | ID: biblio-1133360

RESUMO

Considering that female sexual hormones may modulate the inflammatory response and also exhibit direct effects on the cells of the immune system, herein, we intend to discuss the sex differences and the role of estradiol in modulating the lung and systemic inflammatory response, focusing on its possible application as a treatment modality for SARS-CoV-2 patients. COVID-19 patients develop severe hypoxemia early in the course of the disease, which is silent most of the time. Small fibrinous thrombi in pulmonary arterioles and a tumefaction of endothelial were observed in the autopsies of fatal COVID-19 cases. Studies showed that the viral infection induces a vascular process in the lung, which included vasodilation and endothelial dysfunction. Further, the proportions of CD4+ T and CD8+ T lymphocytes were strongly reduced in patients with severe SARS-CoV-2 infection. Estradiol is connected with CD4+ T cell numbers and increases T-reg cell populations, affecting immune responses to infection. It is known that estradiol exerts a protective effect on endothelial function, activating the generation of nitric oxide (NO) via endothelial nitric oxide synthase. Estrogen attenuates the vasoconstrictor response to various stimuli and induces vasodilation in the pulmonary vasculature during stress situations like hypoxia. It exerts a variety of rapid actions, which are initiated after its coupling with membrane receptors, which in turn, may positively modulate vascular responses in pulmonary disease and help to maintain microvascular flow. Direct and indirect mechanisms underlying the effects of estradiol were investigated, and the results point to a possible protective effect of estradiol against COVID-19, indicating that it may be considered as an adjuvant therapeutic element for the treatment of patients affected by the novel coronavirus.


Assuntos
Humanos , Animais , Masculino , Feminino , Ratos , Pneumonia Viral/terapia , Infecções por Coronavirus/terapia , Estradiol/uso terapêutico , Betacoronavirus , Imunidade Inata , Inflamação/virologia , Fatores Sexuais , Pandemias , SARS-CoV-2 , COVID-19 , Inflamação/tratamento farmacológico
12.
Shock ; 51(4): 495-501, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29688986

RESUMO

BACKGROUND: Brain death (BD) induces hemodynamic instability with microcirculatory hypoperfusion, leading to increased organ inflammation and dysfunction. This study investigated the effects of 7.5% hypertonic saline solution (HSS) on mesenteric microcirculatory dysfunction and inflammation in a rat model of BD. METHODS: Male Wistar rats were anesthetized and mechanically ventilated. BD was induced by rapidly inflating an intracranial balloon catheter. The rats were randomly divided into: SH, sham-operated rats subjected to trepanation; NS, rats treated with NaCl 0.9%, 4 mL/kg immediately after BD; T1, rats treated with HSS (NaCl 7.5%, 4 mL/kg) immediately or 60 min after BD, T60. All groups were analyzed 180 min after the start of the experiment. RESULTS: Rats in BD groups presented with a similar hypertensive peak, followed by hypotension. Proportion of perfused small vessels was decreased in the NS group (46%) compared with the SH group (74%, P = 0.0039). HSS restored the proportion of perfused vessels (T1 = 71%, P = 0.0018). The anti-endothelial nitric oxide synthase (eNOS) protein expression significantly increased in rats given HSS (T1, and T60, P = 0.0002). Similar results were observed regarding endothelin-1 (P < 0.0001). Increased numbers of rolling (P = 0.0015) and migrated (P = 0.0063) leukocytes were observed in the NS group compared with the SH group. Rats given HSS demonstrated an overall reduction in leukocyte-endothelial interactions. The ICAM-1 levels increased in the NS group compared with the SH group, and decreased in the HSS-treated groups (P = 0.0002). CONCLUSIONS: HSS may improve the density of mesenteric perfused small vessels due to its effects on eNOS and endothelin-1 protein expression, and reduces inflammation by decreasing leukocyte adhesion and migration in a rat model of BD.


Assuntos
Morte Encefálica/imunologia , Morte Encefálica/fisiopatologia , Solução Salina Hipertônica/farmacologia , Solução Salina Hipertônica/uso terapêutico , Animais , Eletrólitos , Endotelina-1/metabolismo , Hemodinâmica/efeitos dos fármacos , Imuno-Histoquímica , Inflamação/tratamento farmacológico , Inflamação/fisiopatologia , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Microcirculação/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Selectina-P/metabolismo , Ratos , Ratos Wistar
13.
J Heart Lung Transplant ; 37(11): 1381-1387, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30139547

RESUMO

BACKGROUND: Brain death elicits microvascular dysfunction and inflammation, and thereby compromises lung viability for transplantation. As 17ß-estradiol was shown to be anti-inflammatory and vascular protective, we investigated its effects on lung injury after brain death in male rats. METHODS: Wistar rats were assigned to: sham-operation by trepanation only (SH, n = 7); brain death (BD, n = 7); administration of 17ß-estradiol (280 µg/kg, iv) at 60 minutes after brain death (BD-E2, n = 7). Experiments were performed 180 minutes thereafter. Histopathological changes in the lung were evaluated by histomorphometry. Gene expression of inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), and endothelin-1 was measured by real-time polymerase chain reaction. Protein expression of NO synthases, endothelin-1, platelet endothelial cell adhesion molecule-1 (PECAM-1), vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), BCL-2, and caspase 3 was assessed by immunohistochemistry. Cytokines were quantified by enzyme-linked immunosorbent assay. RESULTS: Treatment with 17ß-estradiol after brain death decreased lung edema and hemorrhage (p < 0.0001), and serum levels of cytokine-induced neutrophil chemoattractant-1 (CINC-1; p = 0.0020). iNOS (p < 0.0001) and VCAM-1 (p < 0.0001) also diminished at protein levels, while eNOS accumulated (p = 0.0002). However, gene expression of iNOS, eNOS, and endothelin-1 was comparable among groups, as was protein expression of endothelin-1, ICAM-1, BCL-2, and caspase 3. CONCLUSIONS: 17ß-Estradiol effectively reduces lung injury in brain-dead rats mainly due to its ability to regulate NO synthases. Thus, the drug may improve lung viability for transplantation.


Assuntos
Anti-Inflamatórios/farmacologia , Morte Encefálica/patologia , Estradiol/farmacologia , Lesão Pulmonar/prevenção & controle , Transplante de Pulmão , Animais , Quimiocina CXCL1/sangue , Hemorragia/patologia , Hemorragia/prevenção & controle , Pneumopatias/patologia , Pneumopatias/prevenção & controle , Lesão Pulmonar/patologia , Masculino , Óxido Nítrico Sintase Tipo II/sangue , Óxido Nítrico Sintase Tipo III/sangue , Edema Pulmonar/patologia , Edema Pulmonar/prevenção & controle , Ratos , Ratos Wistar , Molécula 1 de Adesão de Célula Vascular/sangue
14.
Acta Cir Bras ; 27(7): 465-70, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22760831

RESUMO

PURPOSE: To evaluate histopathological alterations triggered by brain death and associated trauma on different solid organs in rats. METHODS: Male Wistar rats (n=37) were anesthetized with isoflurane, intubated and mechanically ventilated. A trepanation was performed and a balloon catheter inserted into intracraninal cavity and rapidly inflated with saline to induce brain death. After induction, rats were monitored for 30, 180, and 360 min for hemodynamic parameters and exsanguinated from abdominal aorta. Heart, lung, liver, and kidney were removed and fixed in paraffin to evaluation of histological alterations (H&E). Sham-operated rats were trepanned only and used as control group. RESULTS: Brain dead rats showed a hemodynamic instability with hypertensive episode in the first minute after the induction followed by hypotension for approximately 1 h. Histological analyses showed that brain death induces vascular congestion in heart (p<0.05), and lung (p<0.05); lung alveolar edema (p=0.001), kidney tubular edema (p<0.05); and leukocyte infiltration in liver (p<0.05). CONCLUSIONS: Brain death induces hemodynamic instability associated with vascular changes in solid organs and compromises most severely the lungs. However, brain death associated trauma triggers important pathophysiological alterations in these organs.


Assuntos
Morte Encefálica/patologia , Rim/patologia , Fígado/patologia , Pulmão/patologia , Animais , Morte Encefálica/fisiopatologia , Modelos Animais de Doenças , Coração/fisiopatologia , Hemodinâmica/fisiologia , Rim/fisiopatologia , Fígado/fisiopatologia , Pulmão/fisiopatologia , Masculino , Miocárdio/patologia , Distribuição Aleatória , Ratos , Ratos Wistar , Fatores de Tempo
15.
Acta cir. bras ; 27(7): 465-470, jul. 2012. ilus, tab
Artigo em Inglês | LILACS | ID: lil-640094

RESUMO

PURPOSE: To evaluate histopathological alterations triggered by brain death and associated trauma on different solid organs in rats. METHODS: Male Wistar rats (n=37) were anesthetized with isoflurane, intubated and mechanically ventilated. A trepanation was performed and a balloon catheter inserted into intracraninal cavity and rapidly inflated with saline to induce brain death. After induction, rats were monitored for 30, 180, and 360 min for hemodynamic parameters and exsanguinated from abdominal aorta. Heart, lung, liver, and kidney were removed and fixed in paraffin to evaluation of histological alterations (H&E). Sham-operated rats were trepanned only and used as control group. RESULTS: Brain dead rats showed a hemodynamic instability with hypertensive episode in the first minute after the induction followed by hypotension for approximately 1 h. Histological analyses showed that brain death induces vascular congestion in heart (p<0.05), and lung (p<0.05); lung alveolar edema (p=0.001), kidney tubular edema (p<0.05); and leukocyte infiltration in liver (p<0.05). CONCLUSIONS: Brain death induces hemodynamic instability associated with vascular changes in solid organs and compromises most severely the lungs. However, brain death associated trauma triggers important pathophysiological alterations in these organs.


OBJETIVO: Avaliar as alterações histopatológicas desencadeadas pela morte encefálica e pelo trauma associado em diferentes órgãos sólidos em ratos. MÉTODOS: Ratos Wistar machos (n=37) foram anestesiados com isoflurano, entubados e mecanicamente ventilados. Foi realizada trepanação e um cateter foi inserido na cavidade intracraniana e insuflado rapidamente para induzir morte encefálica. Após a indução, os ratos foram monitorados por 30, 180 e 360 min para parâmetros hemodinâmicos e exsanguinados pela aorta abdominal. Coração, pulmão, fígado e rim foram removidos e fixados em parafina para avaliação de alterações histológicas (H&E). Ratos falso-operados foram apenas trepanados e usados como grupo controle. RESULTADOS: Ratos com morte encefálica apresentaram instabilidade hemodinâmica com episódio hipertensivo no primeiro minuto após a indução seguido de hipotensão por aproximadamente 1 hora. Análises histológicas demonstraram que a morte encefálica induz congestão vascular no coração (p<0,05) e pulmão (p<0,05); edema alveolar (p=0,001); edema tubular (p<0,05); e infiltrado leucocitário no fígado (p<0,05). CONCLUSÕES: A morte encefálica induz instabilidade hemodinâmica associada com mudanças vasculares em órgãos sólidos e compromete mais severamente os pulmões. Contudo, o trauma associado à morte encefálica desencadeia importantes alterações fisiopatológicas naqueles órgãos.


Assuntos
Animais , Masculino , Ratos , Morte Encefálica/patologia , Rim/patologia , Fígado/patologia , Pulmão/patologia , Morte Encefálica/fisiopatologia , Modelos Animais de Doenças , Coração/fisiopatologia , Hemodinâmica/fisiologia , Rim/fisiopatologia , Fígado/fisiopatologia , Pulmão/fisiopatologia , Miocárdio/patologia , Distribuição Aleatória , Ratos Wistar , Fatores de Tempo
16.
Clinics (Sao Paulo) ; 67(1): 69-75, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22249483

RESUMO

OBJECTIVE: Experimental findings support clinical evidence that brain death impairs the viability of organs for transplantation, triggering hemodynamic, hormonal, and inflammatory responses. However, several of these events could be consequences of brain death-associated trauma. This study investigated microcirculatory alterations and systemic inflammatory markers in brain-dead rats and the influence of the associated trauma. METHOD: Brain death was induced using intracranial balloon inflation; sham-operated rats were trepanned only. After 30 or 180 min, the mesenteric microcirculation was observed using intravital microscopy. The expression of Pselectin and ICAM-1 on the endothelium was evaluated using immunohistochemistry. The serum cytokine, chemokine, and corticosterone levels were quantified using enzyme-linked immunosorbent assays. White blood cell counts were also determined. RESULTS: Brain death resulted in a decrease in the mesenteric perfusion to 30%, a 2.6-fold increase in the expression of ICAM-1 and leukocyte migration at the mesentery, a 70% reduction in the serum corticosterone level and pronounced leukopenia. Similar increases in the cytokine and chemokine levels were seen in the both the experimental and control animals. CONCLUSION: The data presented in this study suggest that brain death itself induces hypoperfusion in the mesenteric microcirculation that is associated with a pronounced reduction in the endogenous corticosterone level, thereby leading to increased local inflammation and organ dysfunction. These events are paradoxically associated with induced leukopenia after brain damage.


Assuntos
Morte Encefálica/fisiopatologia , Corticosterona/sangue , Hemodinâmica/fisiologia , Mediadores da Inflamação/sangue , Circulação Esplâncnica/fisiologia , Animais , Modelos Animais de Doenças , Molécula 1 de Adesão Intercelular/fisiologia , Leucopenia/sangue , Leucopenia/etiologia , Masculino , Microcirculação/fisiologia , Microscopia de Fluorescência , Selectina-P/fisiologia , Distribuição Aleatória , Ratos , Ratos Wistar
17.
Clinics ; 67(1): 69-75, 2012. ilus, tab
Artigo em Inglês | LILACS | ID: lil-610626

RESUMO

OBJECTIVE: Experimental findings support clinical evidence that brain death impairs the viability of organs for transplantation, triggering hemodynamic, hormonal, and inflammatory responses. However, several of these events could be consequences of brain death-associated trauma. This study investigated microcirculatory alterations and systemic inflammatory markers in brain-dead rats and the influence of the associated trauma. METHOD: Brain death was induced using intracranial balloon inflation; sham-operated rats were trepanned only. After 30 or 180 min, the mesenteric microcirculation was observed using intravital microscopy. The expression of Pselectin and ICAM-1 on the endothelium was evaluated using immunohistochemistry. The serum cytokine, chemokine, and corticosterone levels were quantified using enzyme-linked immunosorbent assays. White blood cell counts were also determined. RESULTS: Brain death resulted in a decrease in the mesenteric perfusion to 30 percent, a 2.6-fold increase in the expression of ICAM-1 and leukocyte migration at the mesentery, a 70 percent reduction in the serum corticosterone level and pronounced leukopenia. Similar increases in the cytokine and chemokine levels were seen in the both the experimental and control animals. CONCLUSION: The data presented in this study suggest that brain death itself induces hypoperfusion in the mesenteric microcirculation that is associated with a pronounced reduction in the endogenous corticosterone level, thereby leading to increased local inflammation and organ dysfunction. These events are paradoxically associated with induced leukopenia after brain damage.


Assuntos
Animais , Masculino , Ratos , Morte Encefálica/fisiopatologia , Corticosterona/sangue , Hemodinâmica/fisiologia , Mediadores da Inflamação/sangue , Circulação Esplâncnica/fisiologia , Modelos Animais de Doenças , Molécula 1 de Adesão Intercelular/fisiologia , Leucopenia/sangue , Leucopenia/etiologia , Microscopia de Fluorescência , Microcirculação/fisiologia , Selectina-P/fisiologia , Distribuição Aleatória , Ratos Wistar
18.
Acta cir. bras ; 19(4): 337-343, July-Aug. 2004. ilus, tab
Artigo em Português | LILACS | ID: lil-363442

RESUMO

O transplante intestinal tem sido utilizado como uma alternativa promissora no tratamento da síndrome do intestino curto em pacientes adultos e pediátricos. Vários modelos experimentais foram desenvolvidos com a finalidade de testar diferentes soluções de preservação. No entanto são modelos complexos que levam, muitas vezes, um tempo prolongado para a sua realização. OBJETIVO: Neste estudo, desenvolvemos um modelo alternativo de autotransplante intestinal in situ analisando os efeitos hemodinâmicos e metabólicos iniciais da perfusão hipotérmica com solução de Ringer lactato. MÉTODOS: Foram utilizados seis cães machos sem raça definida (22,8±1,4 Kg); as variáveis hemodinâmicas sistêmicas foram obtidas por meio de cateter arterial e pelo Swan-Ganz. A perfusão do trato gastrintestinal foi avaliada pela medida do fluxo sangüíneo da veia mesentérica superior (FSVMS, fluxômetro ultra-sônico), e através da medida do pCO2 intestinal (pCO2-int e pCO2-gap, tonometria a gás). Inicialmente realizamos a secção do jejuno proximal e íleo distal e isolamento dos vasos mesentéricos com fitas cardíacas, todo o tecido nervoso e linfático em torno do eixo vascular intestinal foi seccionado. O território mesentérico foi perfundido através da artéria mesentérica superior por 30 minutos com 1000 ml de Ringer lactato a 4ºC, e o efluente drenado através de uma pequena incisão na veia mesentérica superior. Os animais foram observados por 120 min após o inicio do período de reperfusão. Amostras de sangue foram obtidas da aorta abdominal, para análise gasométrica. RESULTADOS: A perfusão intestinal hipotérmica induziu uma redução do FSMVS apenas nos primeiros 30 min de reperfusão (587±70,9 para 398±102,8 ml/min) e um aumento do pCO2-gap (2±2,7 para 29,8±6 mmHg). Não foram observadas alterações significativas em relação a parâmetros hemodinâmicos e metabólicos sistêmicos (PAM, DC, pH, excesso de bases, hemoglobina) assim como na temperatura central. CONCLUSAO: O modelo de autotransplante intestinal é extremamente útil e de fácil execução, para a avaliação inicial de soluções de preservação e/ou drogas antioxidantes, comumente utilizadas no transplante de intestino.


Assuntos
Animais , Masculino , Cães , Hipotermia Induzida , Intestinos , Preservação de Órgãos , Perfusão , Reperfusão , Gasometria , Veias Mesentéricas , Transplante Autólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...