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










Base de dados
Intervalo de ano de publicação
1.
Langenbecks Arch Surg ; 395(1): 33-40, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19851781

RESUMO

PURPOSE: The purpose of this study is to determine gender-specific differences in the development of necrosis in persistent ischemic tissue and to analyze whether differences are due to gender-specific loss of vascular reactivity or change in ischemic tolerance. METHODS: Hairless mice (skh-1) of both genders were assigned to three groups of adolescent, adult, and senescent age. Critical ischemia was induced by transection of the two distal pedicles of the animal's ear. Microcirculation was assessed over a 5-day period using intravital epifluorescence microscopy. Tissue necrosis, blood flow, functional capillary density (FCD), red blood cell (RBC) velocity, and capillary diameter were analyzed. RESULTS: Induction of persistent ischemia caused an age-dependent demarcation of nonperfused flap tissue. Adult and senescent females developed markedly more necrosis than age-matched males (49 +/-1% vs. 37 +/-3% and 53 +/- 3% vs. 44 +/- 2%, respectively; p<0.05), whereas no gender-specific difference in flap necrosis was observed in adolescent animals (31 +/- 2% vs. 33 +/- 3%). Gender did not affect the amount of microcirculatory dysfunction in the flap. Thus, age-matched females and males exhibited a comparable decrease of FCD, RBC velocity, and capillary dilatory response. CONCLUSIONS: Both age and female gender may predispose for an increased susceptibility to develop ischemic tissue necrosis. The increased necrosis in female animals does not apply to an aggravated microvascular dysfunction, but rather to a reduced ischemic tissue tolerance.


Assuntos
Envelhecimento/fisiologia , Isquemia/patologia , Pele/irrigação sanguínea , Retalhos Cirúrgicos/irrigação sanguínea , Análise de Variância , Animais , Modelos Animais de Doenças , Feminino , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Masculino , Camundongos , Camundongos Pelados , Microcirculação/fisiologia , Microscopia de Fluorescência , Necrose/patologia , Necrose/fisiopatologia , Probabilidade , Distribuição Aleatória , Fluxo Sanguíneo Regional , Fatores Sexuais , Estatísticas não Paramétricas
2.
Surgery ; 145(4): 372-83, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19303985

RESUMO

BACKGROUND: Erythropoietin (Epo), the primary regulator of erythropoiesis, has recently been shown to exert antiinflammatory and antiapoptotic properties in neuronal and myocardial tissue. We herein studied whether Epo pretreatment can reduce cell death and ischemic necrosis in a chronic in vivo model. METHODS: C57BL/6 mice were treated daily for 3 consecutive days with either 500 IU EPO/kg body weight (bw) (group Epo 500, n = 8) or 5000 IU EPO/kg bw (group Epo 5000, n = 8) administered intraperitoneally 24 hours before surgery. Thereafter, a random pattern myocutaneous flap subjected to acute persistent ischemia was elevated and fixed into a dorsal skinfold chamber. Flap elevation in animals receiving the water-soluble vitamin E analog Trolox (6-hydroxy-2, 5, 7, 8-tetramethylchroman-2-carboxylic acid) served as a nonspecific antiinflammatory agent control group (Tro); untreated control animals (Con) received saline only. Capillary perfusion, leukocyte-endothelial cell interaction, apoptotic cell death, and tissue necrosis were determined over a 10-day observation period using intravital multifluorescence microscopy. RESULTS: Epo 5000 (44 +/- 26 cm/cm(2)) but, more noticeably, Epo 500 (116 +/- 32 cm/cm(2)) improved capillary perfusion compared with the two control groups, particularly the Con group (9 +/- 7 cm/cm(2); P < .05). The ischemia-associated leukocytic inflammation was found drastically attenuated in both Epo-pretreatment groups. Epo 500 further decreased apoptotic cell death and was effective in significantly reducing tissue necrosis (16% +/- 4% vs Tro: 48% +/- 7% and Con: 52% +/- 4%; P < .001). No angiogenic blood vessel formation could be observed in either of the Epo groups. Of interest, Epo 5000-but not Epo 500-increased systemic hematocrit. CONCLUSION: Despite the lack of neovascularization, Epo pretreatment was capable of reducing ischemic tissue necrosis by protecting capillary perfusion, ie, nutrition of the tissue. Low-dose pretreatment was more effective, a result that was most likely due to the better perfusion conditions without an increase of the hematocrit values. Thus, low-dose Epo pretreatment might represent a promising strategy to protect critically perfused ischemic tissue.


Assuntos
Apoptose/efeitos dos fármacos , Eritropoetina/uso terapêutico , Isquemia/tratamento farmacológico , Microcirculação/efeitos dos fármacos , Retalhos Cirúrgicos/patologia , Animais , Adesão Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/imunologia , Eritropoetina/farmacologia , Hematócrito , Isquemia/imunologia , Leucócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Necrose/prevenção & controle , Neovascularização Fisiológica/efeitos dos fármacos , Receptores da Eritropoetina/metabolismo , Retalhos Cirúrgicos/irrigação sanguínea
3.
Ann Surg ; 248(6): 919-29, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19092336

RESUMO

OBJECTIVE: The objective of this study was to analyze whether erythropoietin (EPO) protects from necrosis of critically perfused musculocutaneous tissue and the mechanisms by which this protection is achieved. BACKGROUND: EPO is the regulator of erythropoiesis and is used to treat patients with anemia of different causes. Recent studies suggest that EPO has also other tissue-protective effects, irrespective of its erythropoietic properties. MATERIAL AND METHODS: C57BL/6-mice were treated with 3 doses of EPO at 500 IU/kg intraperitoneally. EPO was given either before (preconditioning, n = 7), before and after (overlapping treatment, n = 7), or after (treatment, n = 7) surgery. Animals receiving only saline served as controls (CON). Acute persistent ischemia was induced by elevating a randomly perfused flap in the back of the animals. This critically perfused tissue demonstrates an initial microvascular failure of approximately 40%, resulting in approximately 50% tissue necrosis if kept untreated. Repetitive fluorescence microscopy was performed over 10 days, assessing angiogenesis, functional capillary density, inflammatory leukocyte-endothelial cell interaction, apoptotic cell death, and tissue necrosis. Additional molecular tissue analyses included the determination of inducible nitric oxide synthase, erythropoietin receptor (EPO-R), and vascular endothelial growth factor (VEGF). RESULTS: EPO preconditioning did not affect hematocrit and EPO-R expression, but increased inducible nitric oxide synthase in the critically perfused tissue. This correlated with a significant arteriolar dilation, which resulted in a maintained functional capillary density (CON: 0 +/- 0 cm/cm(2); preconditioning: 37 +/- 21 cm/cm(2); overlapping treatment: 72 +/- 26 cm/cm(2); P < 0.05). EPO pretreatment further significantly reduced microvascular leukocyte adhesion and apoptotic cell death. Moreover, EPO pretreatment induced an early VEGF upregulation, which resulted in new capillary network formation (CON: 0 +/- 0 cm/cm(2); preconditioning: 40 +/- 3 cm/cm(2); overlapping treatment: 33 +/- 3 cm/cm(2); P < 0.05). Accordingly, EPO pretreatment significantly reduced tissue necrosis (CON: 48% +/- 2%; preconditioning: 26% +/- 3%; overlapping treatment: 20% +/- 3%; P < 0.05). Of interest, EPO treatment was only able to alleviate ischemia-induced inflammation but could not improve microvascular perfusion and tissue survival. CONCLUSIONS: EPO pretreatment improves survival of critically perfused tissue by nitric oxide -mediated arteriolar dilation, protection of capillary perfusion, and VEGF-initiated new blood vessel formation.


Assuntos
Eritropoetina/farmacologia , Retalhos Cirúrgicos/irrigação sanguínea , Animais , Arteríolas/patologia , Western Blotting , Capilares/fisiologia , Dilatação Patológica , Eritropoetina/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação/fisiologia , Microscopia de Fluorescência/métodos , Necrose , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/fisiologia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Retalhos Cirúrgicos/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
J Surg Res ; 150(2): 293-303, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18262556

RESUMO

BACKGROUND: Tissues are endowed with protective mechanisms to counteract chronic ischemia. Previous studies have demonstrated that endogenous heme oxygenase (HO)-1 may protect parenchymal tissue from inflammation- and reoxygenation-induced injury. Nothing is known, however, on whether endogenous HO-1 also plays a role in chronic ischemia to protect from development of tissue necrosis. The aim of this study is, therefore, to evaluate in vivo whether endogenous HO-1 exerts protection on chronically ischemic musculocutaneous tissue, and whether this protection is mediated by an attenuation of the microcirculatory dysfunction. MATERIALS AND METHODS: In C57BL/6-mice, a chronically ischemic flap was elevated and fixed into a dorsal skinfold chamber. In a second group, tin-protoporphyrin-IX was administrated to competitively block the action of HO-1. Animals without flap elevation served as controls. With the use of intravital fluorescence microscopy, microcirculation, apoptotic cell death, and tissue necrosis were analyzed over a 10-day observation period. The time course of HO-1 expression was determined by Western blotting. RESULTS: Chronic ischemia induced an increase of HO-1 expression, particularly at day 1 and 3. This was associated with arteriolar dilation and hyperperfusion, which was capable of maintaining an adequate capillary perfusion density in the critically perfused central part of the flap, demarcating the distal necrosis. Inhibition of endogenous HO-1 by tin-protoporphyrin-IX completely abrogated arteriolar dilation (44.6 +/- 6.2 microm versus untreated flaps: 71.3 +/- 7.3 microm; P < 0.05) and hyperperfusion (3.13 +/- 1.29 nL/s versus 8.55 +/- 3.56 nL/s; P < 0.05). This resulted in a dramatic decrease of functional capillary density (16 +/- 16 cm/cm(2)versus 84 +/- 31 cm/cm(2); P < 0.05) and a significant increase of apoptotic cell death (585 +/- 51 cells/mm(2)versus 365 +/- 53 cells/mm(2); P < 0.05), and tissue necrosis (73% +/- 5% versus 51% +/- 5%; P < 0.001). CONCLUSION: Thus, our results suggest that chronic ischemia-induced endogenous HO-1 protects ischemically endangered tissue, probably by the vasodilatory action of the HO-1-associated carbon monoxide.


Assuntos
Apoptose , Heme Oxigenase-1/metabolismo , Isquemia/enzimologia , Proteínas de Membrana/metabolismo , Necrose/enzimologia , Retalhos Cirúrgicos/fisiologia , Animais , Arteríolas/fisiopatologia , Capilares/fisiopatologia , Isquemia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Regulação para Cima , Sistema Vasomotor/fisiopatologia
5.
Intensive Care Med ; 33(8): 1434-43, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17576536

RESUMO

OBJECTIVE: To investigate whether microdialysis is capable of assessing metabolic derangements during intra-abdominal hypertension (IAH), and whether monitoring of the rectus abdominis muscle (RAM) by microdialysis represents a reliable approach in the early detection of organ dysfunctions in abdominal compartment syndrome (ACS). DESIGN: Prospective, randomized, controlled animal study. SETTING: University animal research facility. SUBJECTS: Fifteen isoflurane-anesthetized and mechanically ventilated Sprague-Dawley rats. INTERVENTIONS: IAH of 20 mmHg was induced for 3 h and followed by decompression and reperfusion for another 3-h period (n = 10). Five sham-operated animals served as controls. Microdialysis was performed in the anterior gastric wall, liver, kidney, and RAM. The anterior cervical muscles served as distant reference. Glucose, lactate, pyruvate, and glycerol was analyzed throughout the 6-h experiment. MEASUREMENTS AND MAIN RESULTS: Prolonged IAH induced significant cardiopulmonary dysfunction and persistent abdominal organ injury. Microdialysis revealed a significant increase of lactate/pyruvate and glycerol in kidney, intestine and liver, indicating ischemia, energy failure, and cell membrane damage. In addition, at 3 h IAH glucose was significantly decreased in all organs studied. The distant reference did not show any alteration of lactate/pyruvate, glycerol, and glucose over the entire 6-h observation period. In contrast to the other organs, microdialysis of the RAM showed an early and more pronounced increase of lactate, lactate/pyruvate and glycerol already at 1 h IAH. It is noteworthy that lactate, glycerol, and glucose did not completely recover upon decompression of IAH. CONCLUSIONS: Our data suggest that continuous microdialysis in the RAM may represent a promising tool for early detecting IAH-induced metabolic derangements.


Assuntos
Abdome/fisiopatologia , Síndromes Compartimentais/diagnóstico , Diagnóstico Precoce , Microdissecção , Reto do Abdome , Animais , Artérias , Síndromes Compartimentais/metabolismo , Síndromes Compartimentais/fisiopatologia , Modelos Animais de Doenças , Alemanha , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão
6.
Langenbecks Arch Surg ; 392(3): 331-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17380348

RESUMO

BACKGROUND AND AIMS: Insufficient perfusion of distal flap areas, which may lead to partial necrosis, still represents a challenge in reconstructive surgery. In the process of microvascular and endothelial dysfunction, endothelins (ETs) and their receptors may play an important role. Therefore, the aim of the study was to investigate in a chronic in vivo model the effect of various ET-receptor antagonists in critically perfused flap tissue. MATERIALS AND METHODS: A random pattern musculocutaneous flap was elevated in the back of 25 C57BL/6 mice and fixed into a dorsal skinfold chamber. Repetitive intravital fluorescence microscopy was performed over a 10-day observation period, assessing arteriolar diameter, arteriolar blood flow (aBF), functional capillary density (FCD), the area of tissue necrosis, and the development of newly formed blood vessels. ET-receptor blockers were administrated intraperitoneally 30 min before induction of ischemia, as well as daily during the subsequent 4-day period, including (1) BQ-123, a specific ET-A-receptor antagonist (ET-A = 1 mg/kg), (2) BQ-788, a selective ET-B-receptor antagonist (ET-B = 1 mg/kg), and (3) PD-142893, a nonselective ET-AB-receptor antagonist (ET-AB = 0.5 mg/kg). Animals receiving saline only served as controls (n = 7). RESULTS: Despite an increase in aBF during the 10-day observation period (day 1 = 1.92 +/- 0.29 nl/s; day 10 = 4.70 +/- 1.64 nl/s), the flaps of saline-treated controls showed a distinct decrease in FCD (94 +/- 12 cm/cm(2)). This perfusion failure resulted in flap necrosis of 52 +/- 3%. Selective blockade of the ET-B receptor caused a further increase in aBF already at day 1 (2.97 +/- 0.42 nl/s), which persisted during the following 10-day observation period (day 10 = 5.74 +/- 0.69 nl/s). Accordingly, adequate FCD could be maintained (day 10 = 215 +/- 8 cm/cm(2); p < 0.05 vs control), resulting in a significant reduction in flap necrosis (day 10 = 25 +/- 4%; p < 0,05). In contrast, neither selective blockade of the ET-A receptor nor nonselective ET-A- and ET-B-receptor blockade were able to significantly affect aBF when compared to controls (day 1 = ET-A = 1.39 +/- 0.10 nl/s; ET-AB = 1.53 +/- 0.80 nl/s; n.s.). Accordingly, flap necrosis after ET-A- and ET-AB-receptor inhibition did not differ from that of controls (day 10 = ET-A: 46 +/- 10%; ET-AB = 51 +/- 7%). CONCLUSION: Our data show that only selective ET-B-receptor inhibition is capable of maintaining nutritive perfusion and, hence, reducing necrosis in critically perfused flap tissue. Accordingly, administration of ET-B-receptor antagonists may be considered in the treatment of critically perfused flaps.


Assuntos
Antagonistas do Receptor de Endotelina A , Antagonistas do Receptor de Endotelina B , Isquemia/fisiopatologia , Retalhos Cirúrgicos/irrigação sanguínea , Animais , Arteríolas/patologia , Arteríolas/fisiopatologia , Sobrevivência de Enxerto , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Microscopia de Fluorescência , Modelos Animais , Necrose , Oligopeptídeos/farmacologia , Peptídeos Cíclicos/farmacologia , Piperidinas/farmacologia , Fluxo Sanguíneo Regional , Retalhos Cirúrgicos/patologia , Fatores de Tempo , Vênulas/patologia , Vênulas/fisiopatologia
7.
J Surg Res ; 139(2): 209-16, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17254610

RESUMO

BACKGROUND: The purpose of the present study was to develop of a rodent model of abdominal compartment syndrome (ACS), which allows detailed analysis of intra-abdominal hypertension (IAH)- and decompression-associated reperfusion injury. METHODS: In 20 anesthetized and ventilated Sprague-Dawley rats an IAH of 20 mmHg was induced for 3 h by intraperitoneal infusion of gelatin polysuccinate. After decompression, an additional 3-h period of reperfusion was studied. Sham-operated animals, undergoing identical procedures without IAH induction, served as controls. Controlled hyperventilation and intravenous fluid substitution were adapted to keep PCO(2) <60 mmHg and mean arterial pressure (MAP) >100 mmHg. RESULTS: IAH of 20 mmHg could successfully be maintained for the entire 3-h period. MAP was not affected during IAH, however, decreased upon decompression despite forced fluid resuscitation. CVP was markedly elevated during IAH, but returned to baseline after decompression. Of interest, the IAH-induced reduction of PaO(2) did not recover to baseline after decompression, indicating a persistent deterioration of gas exchange. In contrast, IAH-associated elevation of PaCO(2) normalized during reperfusion. IAH was further accompanied by metabolic acidosis, which persisted after decompression, indicating reperfusion injury. IAH was further associated with a significant increase of serum potassium, lactate, AST, LDH, bilirubin, urea, and creatinine as well as creatine kinase (CK) and CK-MB. Histomorphological analysis revealed parenchymal injury in liver, lung, intestine, and myocardium. CONCLUSION: We established an easily reproducible ACS model in the rat, demonstrating hemodynamic deteriorations and organ dysfunctions similar as known from patients with IAH. Decompression did not restore functional deteriorations, indicating persistent post-ACS reperfusion injury. The model may be suitable to study mechanisms and novel treatment strategies in ACS.


Assuntos
Abdome , Síndromes Compartimentais , Modelos Animais de Doenças , Ratos , Abdome/irrigação sanguínea , Animais , Artérias , Pressão Sanguínea , Líquidos Corporais/metabolismo , Peso Corporal , Dióxido de Carbono/sangue , Pressão Venosa Central , Síndromes Compartimentais/complicações , Síndromes Compartimentais/patologia , Síndromes Compartimentais/fisiopatologia , Síndromes Compartimentais/cirurgia , Descompressão Cirúrgica , Feminino , Hipertensão/etiologia , Fígado/patologia , Masculino , Oxigênio/sangue , Ratos Sprague-Dawley , Traumatismo por Reperfusão , Reprodutibilidade dos Testes
8.
Clin Sci (Lond) ; 112(8): 429-40, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17147518

RESUMO

In the present study in a murine model of chronic ischaemia, we analysed: (i) whether aging was associated with an increased susceptibility to ischaemic necrosis, and (ii) whether this was based on microvascular dysfunction or reduced ischaemic tolerance. An ischaemic pedicled skin flap was created in the ear of homozygous hairless mice. The animals were assigned to three age groups, including adolescent (2+/-1 months), adult (10+/-2 months) and senescent (19+/-3 months). Microvascular perfusion of the ischaemic flap was assessed over 5 days by intravital microscopy, evaluating FCD (functional capillary density), capillary dilation response and the area of tissue necrosis. Expression of the stress-protein HO (haem oxygenase)-1 was determined by immunohistochemistry and Western blotting. Induction of chronic ischaemia stimulated a significant expression of HO-1 without a significant difference between the three age groups. This was associated with capillary dilation, which, however, was more pronounced in adolescent (10.5+/-2.8 microm compared with 3.95+/-0.79 microm at baseline) and adult (12.1+/-3.1 microm compared with 3.36+/-0.45 microm at baseline) animals compared with senescent animals (8.5+/-1.7 microm compared with 3.28+/-0.69 microm at baseline; P value not significant). In senescent animals, flap creation further resulted in complete cessation of capillary flow in the distal area of the flap (FCD, 0+/-0 cm/cm(2)), whereas adult (11.9+/-13.5 cm/cm(2)) and, in particular, adolescent animals (58.4+/-33.6 cm/cm(2); P<0.05) were capable of maintaining residual capillary perfusion. The age-associated microcirculatory dysfunction resulted in a significantly increased flap necrosis of 49+/-8% (P<0.05) and 42+/-8% (P<0.05) in senescent and adult animals respectively, compared with 31+/-6% in adolescent mice. Of interest, functional inhibition of HO-1 by SnPP-IX (tin protoporphyrin-IX) in adolescent mice abrogated capillary dilation, decreased functional capillary density and aggravated tissue necrosis comparably with that observed in senescent mice. Thus aging is associated with an increased susceptibility to tissue necrosis, which is due to a loss of vascular reactivity to endogenous HO-1 expression, rather than a reduction in ischaemic tolerance.


Assuntos
Envelhecimento/fisiologia , Isquemia/fisiopatologia , Retalhos Cirúrgicos/irrigação sanguínea , Animais , Western Blotting/métodos , Capilares , Orelha Externa , Feminino , Sobrevivência de Enxerto , Heme Oxigenase (Desciclizante)/análise , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Pelados , Microscopia de Fluorescência , Modelos Animais , Necrose , Fluxo Sanguíneo Regional , Pele/irrigação sanguínea
9.
J Trauma ; 61(4): 924-32, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17033564

RESUMO

BACKGROUND: The present study aimed at quantitatively evaluating the impact of severity of local trauma on manifestation of soft-tissue injury-associated microcirculatory and microlymphatic dysfunctions in a chronic model that allowed repeated analyses by intravital fluorescence microscopy. METHODS: C57BL/6 mice were chronically instrumented with dorsal skinfold chambers and subjected to mild (180 J/m2, n = 6), moderate (270 J/m2, n = 6), or severe trauma (450 J/m2, n = 6; 540 J/m2, n = 6). Nontraumatized animals served as controls (sham; n = 8). Intravital microscopy was performed before and at 5 minutes, 1 hour, 8 hours, 24 hours, 3 days, and 5 days after trauma, and included the analysis of (1) blood and lymph microvessel rupture, (2) hematoma formation and lymph leakage, (3) arteriolar and venular constriction, (4) capillary perfusion failure, (5) arteriolar and venular leukocyte adhesion, and (6) interstitial edema formation. RESULTS: Mild trauma did not induce any changes of microcirculatory and microlymphatic functions. Moderate trauma did not affect lymphatics but provoked arteriolar constriction, capillary perfusion failure, leukocyte-endothelial cell interactions, and minor blood vessel ruptures with hematoma formation. These alterations, however, recovered within the first 24 hours after trauma. Severe trauma also did not affect the lymphatic microvasculature, but resulted in massive hematoma formation, arteriolar constriction, and capillary perfusion failure, which was associated with marked arteriolar and venular leukocyte recruitment and edema formation, and which did not recover to normal over a 5-day observation period. CONCLUSION: Only severe trauma of > 450 J/m2 provokes irreversible microcirculatory dysfunction in soft tissue, however, without affecting the integrity of lymphatic microvessels. Of interest, trauma-induced microcirculatory alterations are neither dominated solely by microcirculatory dysfunction nor by leukocytic inflammation. Instead, both pathologies develop in parallel, generating a vicious circle, which may be responsible for the compromised healing of severely traumatized soft tissue frequently observed in clinical practice.


Assuntos
Doenças Linfáticas/etiologia , Microcirculação/fisiopatologia , Lesões dos Tecidos Moles/fisiopatologia , Animais , Feminino , Escala de Gravidade do Ferimento , Doenças Linfáticas/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Lesões dos Tecidos Moles/classificação
10.
Tissue Eng ; 12(8): 2093-104, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16968151

RESUMO

Long-term function of three-dimensional (3D) tissue constructs depends on adequate vascularization after implantation. Accordingly, research in tissue engineering has focused on the analysis of angiogenesis. For this purpose, 2 sophisticated in vivo models (the chorioallantoic membrane and the dorsal skinfold chamber) have recently been introduced in tissue engineering research, allowing a more detailed analysis of angiogenic dysfunction and engraftment failure. To achieve vascularization of tissue constructs, several approaches are currently under investigation. These include the modification of biomaterial properties of scaffolds and the stimulation of blood vessel development and maturation by different growth factors using slow-release devices through pre-encapsulated microspheres. Moreover, new microvascular networks in tissue substitutes can be engineered by using endothelial cells and stem cells or by creating arteriovenous shunt loops. Nonetheless, the currently used techniques are not sufficient to induce the rapid vascularization necessary for an adequate cellular oxygen supply. Thus, future directions of research should focus on the creation of microvascular networks within 3D tissue constructs in vitro before implantation or by co-stimulation of angiogenesis and parenchymal cell proliferation to engineer the vascularized tissue substitute in situ.


Assuntos
Membrana Corioalantoide/fisiologia , Neovascularização Fisiológica , Fenômenos Fisiológicos da Pele , Engenharia Tecidual , Animais , Embrião de Galinha , Cricetinae
11.
Ann Surg ; 242(6): 869-78, discussion 878-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16327497

RESUMO

INTRODUCTION: Supraphysiologic stress induces a heat shock response, which may exert protection against ischemic necrosis. Herein we analyzed in vivo whether the induction of heat shock protein (HSP) 32 improves survival of chronically ischemic myocutaneous tissue, and whether this is based on amelioration of microvascular perfusion or induction of ischemic tolerance. METHODS: The dorsal skin of mice was subjected to local heat preconditioning (n = 8) 24 hours before surgery. In additional heat-preconditioned animals (n = 8), HSP-32 was inhibited by tin-protoporphyrin-IX. Unconditioned animals served as controls (n = 8). A random-pattern myocutaneous flap was elevated in the back of the animals and fixed into a dorsal skinfold chamber. The microcirculation, edema formation, apoptotic cell death, and tissue necrosis were analyzed over a 10-day period using intravital fluorescence microscopy. RESULTS: HSP-32 protein expression was observed only in heat-preconditioned but not in unconditioned flaps. Heat preconditioning induced arteriolar dilation, which was associated with a significant improvement of both arteriolar blood flow and capillary perfusion in the distal part of the flap. Further, heat shock reduced interstitial edema formation, attenuated apoptotic cell death, and almost completely abrogated the development of flap necrosis (4% +/- 1% versus controls: 53% +/- 5%; P[r] < 0.001). Most strikingly, inhibition of HSP-32 by tin-protoporphyrin-IX completely blunted the preconditioning-induced improvement of microcirculation and resulted in manifestation of 72% +/- 4% necrosis. CONCLUSION: Local heat preconditioning of myocutaneous tissue markedly increases flap survival by maintaining adequate nutritive perfusion rather than inducing ischemic tolerance. The protection is caused by the increased arteriolar blood flow due to significant arteriolar dilation, which is mediated through the carbon monoxide-associated vasoactive properties of HSP-32.


Assuntos
Proteínas de Choque Térmico/fisiologia , Resposta ao Choque Térmico/fisiologia , Isquemia/fisiopatologia , Retalhos Cirúrgicos , Análise de Variância , Animais , Técnicas Imunoenzimáticas , Metaloporfirinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação/fisiologia , Microscopia de Fluorescência , Necrose , Protoporfirinas/farmacologia
12.
Crit Care Med ; 33(8): 1805-13, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16096459

RESUMO

OBJECTIVE: Changes in body temperature occur as a systemic reaction to severe trauma; however, its role in the manifestation of injury remains unclear. Thermoregulatory responses vary considerably from fever to hypothermia. Although hypothermic trauma patients seem to have a worse prognosis, there is the question whether hypothermia per se or the severity of trauma producing the hypothermia is responsible for aggravated injury and increased mortality rate. The present study unravels how moderate to severe systemic hypothermia modulates local microcirculatory dysfunction and cellular injury in local soft tissue trauma. DESIGN: Prospective, experimental study. SETTING: Research laboratory at a university. SUBJECTS: C57BL/6J mice. INTERVENTIONS: A model involving standardized drop weight device-induced tissue trauma and high-resolution multifluorescence microscopy in the dorsal skinfold chamber was used to show arteriolar vasoconstriction, reduction of blood flow, nutritive perfusion failure, and apoptotic cell death at 1 hr after trauma. MEASUREMENTS AND MAIN RESULTS: During the 8-hr posttrauma observation period, microcirculation, but not apoptosis, restituted to almost baseline level. Concomitant systemic hypothermia of either 34 degrees C or 30 degrees C did not affect late manifestation of apoptotic cell death but aggravated initial microcirculatory dysfunction and inhibited recovery during the 8-hr follow-up period. CONCLUSIONS: Our study provides evidence that systemic hypothermia may aggravate soft tissue trauma-associated microcirculatory dysfunction. These experimental results clearly support clinical efforts to prevent hypothermia in the acutely traumatized patient.


Assuntos
Hipotermia/fisiopatologia , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/lesões , Animais , Apoptose , Velocidade do Fluxo Sanguíneo , Adesão Celular , Hipotermia/etiologia , Leucócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação , Microscopia de Fluorescência , Músculo Esquelético/patologia , Lesões dos Tecidos Moles/complicações , Lesões dos Tecidos Moles/fisiopatologia
14.
Am J Physiol Heart Circ Physiol ; 288(3): H1224-32, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15513962

RESUMO

Using intravital microscopy in a chronic in vivo mouse model, we studied the demarcation of myocutaneous flaps and evaluated microvascular determinants for tissue survival and necrosis. Chronic ischemia resulted in a transition zone, characterized by a red fringe and a distally adjacent white falx, which defined the demarcation by dividing the proximally normal from the distally necrotic tissue. Tissue survival in the red zone was determined by hyperemia, as indicated by recovery of the transiently reduced functional capillary density, and capillary remodeling, including dilation, hyperperfusion, and increased tortuosity. Angiogenesis and neovascularization were not observed over the 10-day observation period. The white rim distal to the red zone, appearing as "falx lunatica," showed a progressive decrease of functional capillary density similar to that of the necrotic distal area but without desiccation, and thus transparency, of the tissue. Development of the distinct zones of the critically ischemic tissue could be predicted by partial tissue oxygen tension (Pt(O(2))) analysis by the time of flap elevation. The falx lunatica evolved at a Pt(O(2)) between 6.2 +/- 1.3 and 3.8 +/- 0.7 mmHg, whereas tissue necrosis developed at <3.8 +/- 0.7 mmHg. Histological analysis within the falx lunatica revealed interstitial edema formation and muscle fiber nuclear rarefaction but an absence of necrosis. We have thus demonstrated that ischemia-induced necrosis does not demarcate sharply from normal tissue but develops beside a fringe of tissue with capillary remodeling an adjacent falx lunatica that survives despite nutritive capillary perfusion failure, probably by direct oxygen diffusion.


Assuntos
Isquemia/fisiopatologia , Neovascularização Fisiológica/fisiologia , Retalhos Cirúrgicos/irrigação sanguínea , Retalhos Cirúrgicos/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Microcirculação/fisiologia , Microscopia de Fluorescência , Oxigênio/metabolismo
15.
Am J Physiol Heart Circ Physiol ; 287(5): H2332-40, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15217802

RESUMO

Induction of the "delay phenomenon" by chronic ischemia is an established clinical procedure, but the mechanisms conferring tissue protection are still incompletely understood. To elucidate the role of heme oxygenase-1 [HO-1 or heat shock protein-32 (HSP-32)] in delay, we examined in the skin-flap model of the ear of the hairless mouse, 1) whether chronic ischemia (delay) is capable to induce expression of HO-1, and 2) whether delay-induced HO-1 affects skin-flap microcirculation and survival by either its carbon monoxide-associated vasodilatory action or its biliverdin-associated anti-oxidative mechanism. Chronic ischemia was induced by transsection of the central feeding vessel of the ear 7 days before flap creation. The flap was finally raised by an incision through four-fifths of the base of the ear. Microcirculatory dysfunction and tissue necrosis were studied with the use of laser Doppler fluxmetry and intravital fluorescence microscopy. HO-1 protein expression was determined with Western blot analysis. Seven days of chronic ischemia (delay) induced a marked expression of HO-1. This was paralleled by a significant improvement (P <0.05) of microvascular perfusion and a reduction (P <0.05) of flap necrosis when compared with nondelayed controls. Importantly, blockade of HO-1 activity by tin protoporhyrin-IX completely blunted the protection of microcirculation and the improvement of tissue survival. Additional administration of the vitamin E analog trolox after blockade of HO-1 to mimic exclusively the anti-oxidative action of the heat shock protein did not restore the HO-1-associated microcirculatory improvement and only transiently attenuated the manifestation of flap necrosis. Thus our data indicate that the delay-induced protection from tissue necrosis is mediated by HO-1, predominantly through its carbon monoxide-associated action of adequately maintaining nutritive capillary perfusion.


Assuntos
Citoproteção , Orelha , Heme Oxigenase (Desciclizante)/metabolismo , Pele/patologia , Retalhos Cirúrgicos , Animais , Antioxidantes , Biliverdina , Monóxido de Carbono/metabolismo , Condicionamento Psicológico , Feminino , Expressão Gênica , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1 , Imuno-Histoquímica , Isquemia/enzimologia , Isquemia/patologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Nus , Microcirculação , Necrose , Pele/irrigação sanguínea , Fatores de Tempo , Vasodilatação
16.
Eur J Nucl Med Mol Imaging ; 31(4): 532-41, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14722685

RESUMO

Pancreatic cancer is associated with the worst 5-year survival rate of any human cancer. This high mortality is due, in part, to difficulties in establishing early and accurate diagnosis. Because most tumours share the ability to accumulate amino acids more effectively than normal tissues and any other pathology, assessment of amino acid transport in tumour cells using radiolabelled amino acids has become one of the most promising tools for tumour imaging. This study investigated the potential of p-[(123)I]iodo-L-phenylalanine (IPA) for detection of pancreatic cancer by single-photon emission tomography. IPA affinity for pancreatic tumour was investigated in human pancreatic adenocarcinoma PaCa44 and PanC1 cells, followed by analysis of the underlying mechanisms of tracer accumulation in neoplastic cells. Thereafter, IPA was evaluated for targeting of pancreatic tumours using SCID mice engrafted with primary human pancreatic adenocarcinoma cells, as well as in acute inflammation models in immunocompetent mice and rats. IPA accumulated intensively in human pancreatic tumour cells. Radioactivity accumulation in tumour cells following a 30-min incubation at 37 degrees C/pH 7.4 varied from 41% to 58% of the total loaded activity per 10(6) cells. The cellular uptake was temperature and pH dependent and predominantly mediated by specific carriers for neutral amino acids, namely the sodium-independent and L-leucine-preferring (L-system) transporter and the alanine-, serine- and cysteine-preferring (ASC-system) transporter. Protein incorporation was less than 8%. Biodistribution studies showed rapid localization of the tracer to tumours, reaching 10%+/-2.5% to 15%+/-3% of the injected dose per gram (I.D./g) in heterotopic tumours compared with 17%+/-3.5% to 22%+/-4.3% I.D./g in the orthotopic tumours, at 60 and 240 min post injection of IPA, respectively. In contrast, IPA uptake in the gastrointestinal tract and areas of inflammation remained moderate and decreased with time. Excellent tumour detection was obtained by gamma camera imaging. The specific and high-level targeting of IPA to tumour and the negligible uptake in the gastrointestinal tract and areas of inflammation indicate that p-[(123)I]iodo-L-phenylalanine is a promising tracer for differential diagnosis of pancreatic cancer.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/metabolismo , Fenilalanina/análogos & derivados , Fenilalanina/farmacocinética , Adenocarcinoma/diagnóstico , Animais , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Humanos , Taxa de Depuração Metabólica , Camundongos , Camundongos SCID , Especificidade de Órgãos , Neoplasias Pancreáticas , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual
17.
Langenbecks Arch Surg ; 388(5): 281-90, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14530978

RESUMO

BACKGROUND: During the past decade, experimental studies have provided convincing evidence that microcirculatory dysfunction plays a pivotal role in the manifestation of tissue injury in ischemia-reperfusion and osteomyocutaneous flap transfer. The study of the mechanisms of injury, however, requires sophisticated experimental in vivo models. With the use of microsurgical techniques, osteomyocutaneous flap transfer can successfully be performed in rat hind limbs, allowing in vivo fluorescent microscopic analysis of post-ischemic microcirculatory dysfunction in all tissues involved, including periosteum, striated muscle, subcutis and skin. The drawback of this "acute" model is that the period of analysis is restricted to a few hours only. METHOD: To overcome this limitation, the "chronic" dorsal skinfold chamber preparation, containing striated muscle and subcutis, can be used. This model allows one to study microcirculatory dysfunction after both tourniquet-induced and pressure-induced ischemia-reperfusion-induced tissue injury over a period of up to 3 weeks. RESULTS: With the use of these models, recent investigations have demonstrated that ischemia-reperfusion and osteomyocutaneous flap transfer are associated with capillary perfusion failure (no-reflow), mediated by intravascular hemoconcentration, endothelial swelling and endothelin (ET)-1-mediated microvascular constriction. In addition, post-ischemic reperfusion provokes an inflammatory response (reflow paradox) in post-capillary venules, which is characterized by beta2-integrin-mediated and intercellular adhesion molecule (ICAM)-1-mediated leukocyte adhesion and vascular hyperpermeability, which results in interstitial edema formation. Treatment studies have produced evidence that isovolemic hemodilution and heat shock protein induction are successful in ameliorating capillary no-reflow, while blockade of adhesion molecules, inactivation of oxygen radicals and, also, induction of heat shock proteins, are capable of reducing the post-ischemic inflammatory response. CONCLUSION: These experimental results not only demonstrate the importance of the use of advanced in vivo methods to delineate pathophysiological mechanisms in complex disease models, but may also provide a basis for potential prospective randomized trials to test the benefit for the patient in the daily clinical routine.


Assuntos
Músculo Esquelético/irrigação sanguínea , Traumatismo por Reperfusão/fisiopatologia , Retalhos Cirúrgicos/irrigação sanguínea , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Microcirculação , Microscopia de Fluorescência , Pele/irrigação sanguínea , Retalhos Cirúrgicos/fisiologia
18.
FASEB J ; 17(2): 175-85, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12554696

RESUMO

Local cooling protects against TNF-alpha-induced injury by attenuating inflammation-associated microcirculatory dysfunction and leukocytic response. Mechanisms of protection, however, are not fully understood. We studied whether the metabolites of the HO and NOS pathway, exerting potent vasodilatory, antioxidant, and anti-apoptotic properties, are involved in tissue cryoprotection. In animals pretreated with L-NAME or SnPP-IX, cooling-associated abrogation of TNF-alpha-induced microcirculatory dysfunction was abolished. Combined L-NAME/SnPP-IX pretreatment did not cause greater blunting than seen when each mediator system was inhibited separately. In SnPP-IX- but not L-NAME-pretreated animals, transient hypothermia failed to reduce TNF-alpha-mediated leukocyte adherence. Vice versa, treatment of TNF-alpha-exposed animals with either the NO donor l-arginine or the HO-1 inductor hemin mimicked cooling-associated tissue protection except for failure of l-arginine to abrogate the inflammatory leukocyte response. The efficiency of cooling to inhibit TNF-alpha-induced apoptotic cell death was blunted in SnPP-IX-, L-NAME-, and SnPP-IX/L-NAME-pretreated animals. Coadministration of Trolox in SnPP-IX-treated animals partly attenuated leukocyte adherence and cell apoptosis, implying that the HO pathway metabolite biliverdin contributes to the salutary effects of cooling. Thus, our study provides evidence that metabolites of the HO and the NOS pathway mediate the cooling-associated protection of inflamed tissue. Biliverdin rather than CO and NO mediates the anti-inflammatory action, whereas a coordinated function of the gaseous monoxides prevents microcirculatory dysfunction and apoptotic cell death.


Assuntos
Apoptose/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/metabolismo , Óxido Nítrico/metabolismo , Pele/irrigação sanguínea , Fator de Necrose Tumoral alfa/administração & dosagem , Animais , Arginina/farmacologia , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Crioterapia/métodos , Inibidores Enzimáticos/farmacologia , Feminino , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Hemina/farmacologia , Inflamação/induzido quimicamente , Inflamação/enzimologia , Inflamação/terapia , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Masculino , Metaloporfirinas/farmacologia , Camundongos , Microcirculação/efeitos dos fármacos , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/antagonistas & inibidores , Protoporfirinas/farmacologia , Pele/efeitos dos fármacos , Pele/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...