Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Am J Pathol ; 188(2): 450-460, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29154961

RESUMEN

The incorporation of endothelial progenitor cells (EPCs) into newly developing blood vessels contributes to the vascularization of endometriotic lesions. We analyzed whether cyclooxygenase (COX)-2 signaling regulates this vasculogenic process. Endometriotic lesions were surgically induced in irradiated FVB/N mice, which were reconstituted with bone marrow from FVB/N-TgN [Tie2/green fluorescent protein (GFP)] 287 Sato mice. The animals received ß-estradiol 17-valerate once a week and were treated daily with the selective COX-2 inhibitor parecoxib (25 mg/kg) or vehicle (control) for 7 and 28 days. Analyses involved the determination of lesion growth, cyst formation, homing of GFP+/Tie2+ EPCs, numbers of circulating EPCs, vascularization, cell proliferation, apoptosis, and immune cell infiltration by means of high-resolution ultrasonography, caliper measurements, flow cytometry, histologic analysis, and immunohistochemical analysis. In parecoxib-treated mice, blood circulating EPCs were higher, but numbers of recruited EPCs in endometriotic lesions were significantly lower when compared with controls. This finding was associated with an impaired early vascularization and stromal tissue growth as well as reduced glandular secretory activity of the lesions. Parecoxib-treated lesions further contained less proliferating and more apoptotic cells and exhibited lower numbers of infiltrating macrophages and neutrophilic granulocytes. These findings demonstrate that the inhibition of COX-2 suppresses vasculogenesis in endometriotic lesions, which may contribute to an impaired lesion vascularization and growth.


Asunto(s)
Ciclooxigenasa 2/fisiología , Endometriosis/patología , Células Progenitoras Endoteliales/patología , Neovascularización Patológica/prevención & control , Animales , Apoptosis/fisiología , Proliferación Celular/fisiología , Inhibidores de la Ciclooxigenasa 2/farmacología , Endometriosis/diagnóstico por imagen , Endometriosis/enzimología , Endometriosis/inmunología , Endometrio/irrigación sanguínea , Endometrio/patología , Endotelio Vascular/patología , Femenino , Isoxazoles/farmacología , Ratones Endogámicos , Microvasos/patología , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Transducción de Señal/fisiología , Ultrasonografía
2.
Am J Pathol ; 186(8): 2129-2142, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27315780

RESUMEN

The incorporation of endothelial progenitor cells (EPCs) into microvessels contributes to the vascularization of endometriotic lesions. Herein, we analyzed whether this vasculogenic process is regulated by estrogen. Estrogen- and vehicle-treated human EPCs were analyzed for migration and tube formation. Endometriotic lesions were induced in irradiated FVB/N mice, which were reconstituted with bone marrow from FVB/N-TgN (Tie2/green fluorescent protein) 287 Sato mice. The animals were treated with 100 µg/kg ß-estradiol 17-valerate or vehicle (control) over 7 and 28 days. Lesion growth, cyst formation, homing of green fluorescent protein(+)/Tie2(+) EPCs, vascularization, cell proliferation, and apoptosis were analyzed by high-resolution ultrasonography, caliper measurements, histology, and immunohistochemistry. Numbers of blood circulating EPCs were assessed by flow cytometry. In vitro, estrogen-treated EPCs exhibited a higher migratory and tube-forming capacity when compared with controls. In vivo, numbers of circulating EPCs were not affected by estrogen. However, estrogen significantly increased the number of EPCs incorporated into the lesions' microvasculature, resulting in an improved early vascularization. Estrogen further stimulated the growth of lesions, which exhibited massively dilated glands with a flattened layer of stroma. This was mainly because of an increased glandular secretory activity, whereas cell proliferation and apoptosis were not markedly affected. These findings indicate that vasculogenesis in endometriotic lesions is dependent on estrogen, which adds a novel hormonally regulated mechanism to the complex pathophysiology of endometriosis.


Asunto(s)
Endometriosis/patología , Células Progenitoras Endoteliales/patología , Estradiol/análogos & derivados , Neovascularización Patológica/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/efectos de los fármacos , Estradiol/metabolismo , Estradiol/farmacología , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Neovascularización Patológica/patología , Reacción en Cadena de la Polimerasa , Transcriptoma
3.
Artículo en Inglés | MEDLINE | ID: mdl-38299944

RESUMEN

Objective: Chronic wounds represent a considerable burden for the affected patients and the health care system. To overcome this problem, effective treatment strategies are urgently required. In this study, we tested a novel approach by combining platelet-rich plasma (PRP) and microvascular fragments (MVF) to create a prevascularized gel dressing. Approach: MVF were enzymatically isolated from the epididymal fat pads of transgenic green fluorescent protein (GFP)+ C57BL/6J donor mice. Subsequently, 5,000 MVF were suspended in 10 µL murine PRP as carrier and transferred into full-thickness skin wounds within dorsal skinfold chambers of C57BL/6J wild-type mice (PRP+MVF). Wound healing in comparison to empty wounds (control) and wounds filled with PRP alone was repeatedly analyzed throughout 14 days by means of stereomicroscopy, histology, and immunohistochemistry. Results: Planimetric assessment of the wound size over time revealed a significantly accelerated and improved healing of PRP+MVF-treated wounds when compared with PRP-treated and empty control wounds. These wounds also exhibited a significantly higher density of blood and lymph vessels, which originated from the GFP+ MVF isolates and effectively promoted granulation tissue formation inside the skin defects. Innovation: This study is the first to combine PRP and MVF for the improvement of wound healing. Conclusion: The combination of PRP and MVF represents a promising approach for the future treatment of wounds that do not heal spontaneously due to poor wound-healing conditions.

4.
Microcirculation ; 19(8): 739-48, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22827532

RESUMEN

OBJECTIVE: We developed a model for direct assessment of BMC sequestration in the postischemic murine myocardium after direct antegrade intracoronary injection. METHODS: Modified syngeneic heterotopic heart transplantation was used as a basic model for global myocardial I/R injury in a total of n = 29 animals. IVM was employed to analyze the right ventricular subepicardial coronary microcirculation and for tracking fluorescently labeled BMCs. RESULTS: IVM allowed monitoring all segments of the coronary microcirculation including feeding arterioles, nutritive capillaries, and postcapillary venules. WI and generalized atherosclerosis induced profound reperfusion failure, particularly in nutritive myocardial capillaries. BMCs were found to exclusively sequester in myocardial capillaries, but not in coronary arterioles or postcapillary venules. The sequestration of BMCs in coronary capillaries occurred independent of WI, generalized atherosclerosis, or adhesion molecule function. CONCLUSIONS: This is the first study allowing direct assessment of BMC homing to the postischemic myocardium. Heterotopic heart transplantation and IVM are proper means to study the myocardial sequestration of BMCs after direct antegrade intracoronary injection in vivo. We show for the first time that intracoronarily injected BMCs sequester exclusively in nutritive myocardial capillaries.


Asunto(s)
Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Circulación Coronaria , Microcirculación , Daño por Reperfusión Miocárdica , Células Madre/metabolismo , Animales , Células de la Médula Ósea/patología , Trasplante de Corazón , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Células Madre/patología , Trasplante Heterotópico , Trasplante Isogénico
5.
Am J Pathol ; 178(1): 442-50, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224081

RESUMEN

Endometriosis is a frequent gynecological disease that is characterized by the development of vascularized endometriotic lesions inside the peritoneal cavity. Herein, we analyzed whether circulating endothelial progenitor cells (EPCs) are recruited and incorporated into the microvasculature of these lesions. Intraperitoneal endometriotic lesions were surgically induced in irradiated FVB/N mice, which were reconstituted with bone marrow from FVB/N-TgN (Tie2/green fluorescent protein [GFP]) 287 Sato mice. Vascularization and recruitment of GFP-positive EPCs in the lesions was analyzed by intravital fluorescence microscopy and immunohistochemistry over 4 weeks. The numbers of stem cell antigen-1 (Sca-1)/vascular endothelial growth factor receptor-2-positive EPCs in blood and hematopoietic organs of additional endometriotic and control mice were assessed by flow cytometry. We found that approximately 15% of the microvascular endothelium in engrafting endometriotic lesions consisted of incorporated GFP-positive EPCs. Recruitment of EPCs into the lesions coincided with the establishment of own blood supply and the expression of stromal cell-derived factor-1. Accordingly, treatment with the stromal cell-derived factor-1/chemokine receptor type 4 axis antagonist AMD3100 significantly decreased the number of recruited EPCs and the vascularization of endometriotic lesions. However, endometriosis did not induce increased levels of EPCs in the blood, bone marrow, and spleen of C57BL/6 mice. To our knowledge, our findings indicate for the first time that vasculogenesis (ie, de novo generation of blood vessels from EPCs) may represent an integral mechanism in the pathogenesis of endometriosis.


Asunto(s)
Endometriosis/patología , Endotelio Vascular/patología , Neovascularización Patológica/patología , Células Madre/patología , Animales , Antígenos Ly/sangre , Antígenos Ly/metabolismo , Bencilaminas , Ciclamas , Endometriosis/sangre , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Compuestos Heterocíclicos/farmacología , Proteínas de la Membrana/sangre , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Tissue Eng Regen Med ; 19(1): 161-175, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34536211

RESUMEN

BACKGROUND: Adipose tissue-derived microvascular fragments (MVF) represent effective vascularization units for tissue engineering. Most experimental studies in rodents exclusively use epididymal adipose tissue as a visceral fat source for MVF isolation. However, in future clinical practice, MVF may be rather isolated from liposuctioned subcutaneous fat tissue of patients. Therefore, we herein compared the vascularization characteristics of MVF isolates from visceral and subcutaneous fat tissue of murine origin. METHODS: MVF isolates were generated from visceral and subcutaneous fat tissue of donor mice using two different enzymatic procedures. For in vivo analyses, the MVF isolates were seeded onto collagen-glycosaminoglycan scaffolds and implanted into full-thickness skin defects within dorsal skinfold chambers of recipient mice. RESULTS: By means of the two isolation procedures, we isolated a higher number of MVF from visceral fat tissue when compared to subcutaneous fat tissue, while their length distribution, viability and cellular composition were comparable in both groups. Intravital fluorescence microscopy as well as histological and immunohistochemical analyses revealed a significantly reduced vascularization of implanted scaffolds seeded with subcutaneous MVF isolates when compared to implants seeded with visceral MVF isolates. Light and scanning electron microscopy showed that this was due to high amounts of undigested connective tissue within the subcutaneous MVF isolates, which clogged the scaffold pores and prevented the interconnection of individual MVF into new microvascular networks. CONCLUSION: These findings indicate the need for improved protocols to generate connective tissue-free MVF isolates from subcutaneous fat tissue for future translational studies.


Asunto(s)
Microvasos , Neovascularización Fisiológica , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Grasa Subcutánea , Ingeniería de Tejidos/métodos
7.
Front Bioeng Biotechnol ; 9: 777687, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34778238

RESUMEN

Adipose tissue-derived microvascular fragments (MVF) represent effective vascularization units for tissue engineering. Most experimental studies exclusively use epididymal fat tissue of male donor mice as a source for MVF isolation. However, in future clinical practice, MVF-based approaches may be applied in both male and female patients. Therefore, we herein compared the vascularization capacity of MVF isolated from the epididymal and peri-ovarian fat tissue of male and female donor mice. Freshly isolated MVF from male and female donors did not differ in their number, length distribution, viability and cellular composition. After their assembly into spheroids, they also exhibited a comparable in vitro sprouting activity. Moreover, they could be seeded onto collagen-glycosaminoglycan matrices, which were implanted into full-thickness skin defects within mouse dorsal skinfold chambers. Repetitive intravital fluorescence microscopy as well as histological and immunohistochemical analyses revealed a comparable vascularization and incorporation of implants seeded with MVF of male and female origin. Taken together, these findings demonstrate that the vascularization capacity of MVF is not gender-specific.

8.
Int J Cancer ; 126(6): 1302-15, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19821487

RESUMEN

Recent studies have shown that the chemokine stromal cell-derived factor (SDF)-1 and its receptor CXCR4 are involved in the metastatic process of colorectal cancer. The impact of SDF-1 on the stimulated metastatic growth during hepatectomy-associated liver regeneration is unknown. With the use of a heterotopic murine colon cancer model, we analyzed whether blockade of SDF-1 inhibits angiogenesis and extrahepatic growth of colorectal cancer after liver resection. Functional neutralization of SDF-1 by 1 mg/kg body weight anti-SDF-1 antibody only slightly delayed the initial tumor cell engraftment but also did not reduce the size of established extrahepatic tumors compared with controls. Tumor cell apoptosis was increased by anti-SDF-1 treatment only during the early 5-9-day period of tumor cell engraftment, but was found significantly decreased during the late phase of tumor growth. The initial delay of tumor cell engraftment was associated with an increase of tumor capillary density and microvascular permeability. This was associated with an increased vascular endothelial growth factor (VEGF) expression and an enhanced tumor cell invasion of the neighboring tissue. In contrast to the neutralization of SDF-1, blockade of the SDF-1 receptors CXCR4 and CXCR7 significantly reduced tumor capillary density and tumor growth. Thus, our study indicates that neutralization of SDF-1 after hepatectomy is not capable of inhibiting angiogenesis and growth of extrahepatic colorectal tumors, because it is counteracted by the compensatory actions through an alternative VEGF-dependent pathway.


Asunto(s)
Quimiocina CXCL12/metabolismo , Neoplasias Colorrectales/metabolismo , Neovascularización Patológica/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Femenino , Citometría de Flujo , Hepatectomía , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Receptores CXCR/inmunología , Receptores CXCR4/inmunología , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Ann Surg ; 252(1): 131-41, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20531003

RESUMEN

BACKGROUND: Liver insufficiency after major hepatectomy still represents a serious challenge in liver surgery. Although some previous studies indicate that erythropoietin (EPO) and its analogue darbepoetin-alpha (DPO) may improve liver function and liver regeneration, little is known on their effect on tumor growth after hepatectomy. Because EPO may promote tumor progression, we herein studied the effect of DPO on tumor growth after major hepatectomy. METHODS: CT26.WT colorectal cancer cells were implanted into the left liver lobe of BALB/c mice. Animals underwent 50% hepatectomy (Phx) and received 10 microg/kg DPO-treatment. Additional Phx animals received only saline treatment. Nonhepatectomized animals with DPO-treatment or saline treatment served as controls. One week after hepatectomy angiogenic blood vessel formation, leukocyte-endothelial cell interaction, tumor cell proliferation, apoptotic cell death, and tumor growth were studied using intravital fluorescence microscopy, histology, Immunohistochemistry, and Western blot analysis. RESULTS: Phx significantly enhanced the growth of liver metastases. This was associated with an increase of tumor capillary density and tumor cell proliferation. In nonhepatectomized animals, DPO only slightly affected metastatic growth. In hepatectomized animals, however, DPO significantly enhanced the Phx-induced stimulation of tumor growth. This was associated with an increased tumor capillary density, a decreased leukocyte-endothelial cell interaction, and a reduced cleaved caspase-3 expression of the CT26.WT cells. CONCLUSIONS: : Our data indicate that DPO significantly enhances the hepatectomy-induced stimulation of colorectal liver metastatic growth by increasing neovascularization, suppressing intratumoral leukocyte recruitment, and reducing tumor cell apoptosis. Thus, EPOs may not be used in patients undergoing hepatectomy for malignant tumor resection.


Asunto(s)
Neoplasias Colorrectales/patología , Eritropoyetina/análogos & derivados , Hematínicos/efectos adversos , Hepatectomía/efectos adversos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/análisis , Darbepoetina alfa , Eritropoyetina/efectos adversos , Eritropoyetina/farmacología , Femenino , Citometría de Flujo , Hematínicos/farmacología , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica , Infiltración Neutrófila/efectos de los fármacos , Células Tumorales Cultivadas
10.
J Tissue Eng ; 11: 2041731420911816, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32313616

RESUMEN

Adipose tissue-derived microvascular fragments rapidly reassemble into microvascular networks within implanted scaffolds. Herein, we analyzed the contribution of macrophages to this process. C57BL/6 mice received clodronate (clo)-containing liposomes for macrophage depletion, whereas animals treated with phosphate-buffered-saline-containing liposomes served as controls. Microvascular fragments were isolated from clo- and phosphate-buffered-saline-treated donor mice and seeded onto collagen-glycosaminoglycan matrices, which were implanted into dorsal skinfold chambers of clo- and phosphate-buffered-saline-treated recipient mice. The implants' vascularization and incorporation were analyzed by stereomicroscopy, intravital fluorescence microscopy, histology, and immunohistochemistry. Compared to controls, matrices within clo-treated animals exhibited a significantly reduced functional microvessel density. Moreover, they contained a lower fraction of microvessels with an α-smooth muscle actin (SMA)+ cell layer, indicating impaired vessel maturation. This was associated with a deteriorated implant incorporation. These findings demonstrate that macrophages not only promote the reassembly of microvascular fragments into microvascular networks, but also improve their maturation during this process.

11.
Acta Biomater ; 108: 194-206, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32194259

RESUMEN

Porous polyethylene (pPE) is a commonly used biomaterial in craniofacial reconstructive surgery. However, implant failure due to insufficient vascularization represents a major issue. To overcome this problem, we herein introduce an effective strategy to improve the vascularization and incorporation of pPE. Adipose tissue-derived microvascular fragments (MVF) from transgenic green fluorescent protein (GFP)+ mice were suspended in platelet-rich plasma (PRP) for the coating of pPE. PRP/MVF-coated pPE as well as PRP-coated and uncoated controls were subsequently implanted into the dorsal skinfold chamber and the flanks of GFP- wild-type mice to analyze their in vivo performance throughout 2, 4 and 8 weeks by means of intravital fluorescence microscopy, histology and immunohistochemistry. The GFP+/GFP- cross-over design allowed the identification of GFP+ MVF within the implants. Shortly after implantation, they rapidly reassembled into new blood-perfused microvascular networks, resulting in a significantly accelerated vascularization of PRP/MVF-coated pPE when compared to both controls. The overall numbers of rolling and adherent leukocytes within the microcirculation as well as macrophages, multi-nucleated giant cells and mast cells around the implants did not differ between the three groups. However, in contrast to uncoated controls, PRP/MVF-coated and PRP-coated pPE promoted pro-angiogenic M2 macrophage polarization at the implantation site. These findings demonstrate that PRP/MVF-coating represents a highly effective strategy to enhance the vascularization, biocompatibility and tissue incorporation of pPE. STATEMENT OF SIGNIFICANCE: The clinical in vivo performance of implanted biomaterials is crucially dependent on their adequate incorporation into the body. To achieve this, we herein introduce an effective biological coating strategy. Our results demonstrate that coating with PRP and MVF accelerates and enhances the vascularization, biocompatibility and tissue incorporation of porous polyethylene. Because this type of biological coating is easily applicable on any type of biomaterial, our approach may rapidly be translated into clinical practice to improve the outcome of various regenerative approaches.


Asunto(s)
Plasma Rico en Plaquetas , Polietileno , Tejido Adiposo , Animales , Ratones , Neovascularización Fisiológica , Porosidad
12.
J Biol Eng ; 12: 24, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30473729

RESUMEN

BACKGROUND: The seeding of scaffolds with the stromal vascular fraction (SVF) of adipose tissue is a common prevascularization strategy in tissue engineering. Alternatively, adipose tissue-derived microvascular fragments (ad-MVF) may serve as vascularization units. In contrast to SVF single cells, they represent a mixture of intact arteriolar, capillary and venular vessel segments. Therefore, we herein hypothesized that the ad-MVF-based prevascularization of scaffolds is superior to the conventional SVF single cells-based approach. RESULTS: SVF single cells and ad-MVF were enzymatically isolated from epididymal fat pads of green fluorescent protein (GFP)+ donor mice to assess their viability and cellular composition using fluorescence microscopy and flow cytometry. Moreover, collagen-glycosaminoglycan matrices (Integra®) were seeded with identical amounts of the isolates and implanted into full-thickness skin defects within dorsal skinfold chambers of GFP- recipient mice for the intravital fluorescent microscopic, histological and immunohistochemical analysis of implant vascularization and incorporation throughout an observation period of 2 weeks. Non-seeded matrices served as controls. While both isolates contained a comparable fraction of endothelial cells, perivascular cells, adipocytes and stem cells, ad-MVF exhibited a significantly higher viability. After in vivo implantation, the vascularization of ad-MVF-seeded scaffolds was improved when compared to SVF-seeded ones, as indicated by a significantly higher functional microvessel density. This was associated with an enhanced cellular infiltration, collagen content and density of CD31+/GFP+ microvessels particularly in the center of the implants, demonstrating a better incorporation into the surrounding host tissue. In contrast, non-seeded matrices exhibited a poor vascularization, incorporation and epithelialization over time. CONCLUSIONS: The present study demonstrates that ad-MVF are highly potent vascularization units that markedly accelerate and improve scaffold vascularization when compared to the SVF.

13.
Shock ; 18(4): 348-54, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12392279

RESUMEN

Elderly patients have a higher incidence of morbidity and mortality due to infectious diseases. Because most immune functions in the elderly differ compared with those in younger subjects, we studied the effect of the immunomodulating agent G-CSF on endotoxic liver injury and cytokine release in an aging animal model of acute sepsis. Young (3-month-old), mature (12-month-old), and senescent (24-month-old) male Sprague-Dawley rats (n = 6 each) were treated with bacterial endotoxin for 6 h. Age-matched groups of animals (n = 6 each) received G-CSF (200 microg/kg i.v.) 1 h prior to endotoxin. LPS-induced hepatic toxicity, as assessed in vivo by nutritive perfusion failure, intravascular leukocyte accumulation, biliary excretion dysfunction, and liver enzyme release, was significantly more pronounced in mature and old animals when compared with young animals. Concomitantly, mature but, in particular, senescent endotoxic animals exhibited 2- to 14-fold higher plasma levels of TNF-alpha, IL-1beta, IL-6, IL-10, and Rantes than young endotoxic rats. The percentage of G-CSF receptor surface expression on neutrophils did not significantly differ between young, mature, and senescent animals (36%-46%) and was found comparably down-regulated at 6 h after LPS exposure (15%-19%). Kupffer cell activation, i.e., clearance of intravascularly applied fluorescent latex beads, did not differ between the LPS-exposed age groups. G-CSF dampened LPS-induced Kupffer cell activation, and significantly reduced plasma cytokine levels in young and mature, but not in senescent animals. Thereby, G-CSF caused attenuation of hepatic tissue injury in all except the senescent endotoxic animals. In summary, our results show an age-dependent increase in hepatic LPS toxicity. Because flow cytometric analysis of G-CSF receptor expression disproved that cytokinemia-induced down-regulation of G-CSF receptors might account for the unresponsiveness to G-CSF in the senescent animals, other homeostatic regulatory mechanisms appear to undergo changes with age that bring out a disrupted balance between inflammatory cytokines with unresponsiveness to G-CSF and, thus, compromised host defense mechanisms in the elderly.


Asunto(s)
Envejecimiento/inmunología , Endotoxemia/inmunología , Factor Estimulante de Colonias de Granulocitos/inmunología , Animales , Aspartato Aminotransferasas/efectos de los fármacos , Aspartato Aminotransferasas/metabolismo , Bilis/fisiología , Citocinas/sangre , Endotoxemia/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/farmacología , Macrófagos del Hígado/efectos de los fármacos , Lipopolisacáridos , Hígado/patología , Circulación Hepática/efectos de los fármacos , Masculino , Neutrófilos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo
14.
Clin Exp Metastasis ; 31(4): 447-59, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24493023

RESUMEN

The chemokine CXCL12 has a decisive role in tumor progression by mediating pro-angiogenic and pro-metastatic effects through its receptor CXCR4. The CXCL12 pathway is connected with another chemokine, CXCL11, through its second receptor CXCR7. CXCL11 also binds to the CXCR3 receptor. CXCL11 function in tumor angiogenesis is likely receptor dependent because CXCR3 predominantly mediates angiostatic signals whereas CXCR7 mediated signaling is rather angiogenic. We therefore studied the interaction of CXCL12 and CXCL11 in an in vivo model of colorectal cancer metastasis. GFP-transfected CT26.WT colorectal cancer cells were implanted into the dorsal skinfold chamber of syngeneic BALB/c mice. The animals received either peritumoral application of CXCL11 or intraperitoneal injections with neutralizing antibodies against CXCL11, CXCL12 or both. Tumor growth characteristics, angiogenesis, cell migration, invasive tumor growth, tumor cell proliferation and apoptosis were studied by intravital fluorescence microscopy and immunohistochemistry during an observation period of 14 days. Local exposure to CXCL11 significantly stimulated tumor growth compared to controls and enhanced invasive growth characteristics without affecting tumor angiogenesis and tumor cell migration. Neither CXCL11 nor CXCL12-blockade had a significant impact on tumor growth and angiogenesis, whereas the combined neutralization of CXCL11 and CXCL12 almost completely abrogated tumor vessel formation. As a consequence, tumor growth and invasive growth characteristics were reduced compared to the other groups. The results of the present study underline the interaction of CXCL12 and CXCL11 during tumor angiogenesis. The combined blockade of both signaling pathways may provide an interesting anti-angiogenic approach for anti-tumor therapy.


Asunto(s)
Movimiento Celular , Quimiocina CXCL11/metabolismo , Quimiocina CXCL12/metabolismo , Neoplasias Colorrectales/irrigación sanguínea , Neovascularización Patológica/patología , Animales , Apoptosis , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Fluorescente , Neovascularización Patológica/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Microcirculation ; 10(2): 143-52, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12700583

RESUMEN

OBJECTIVE: In vitro and in vivo studies using isolated platelets require that the cells used for testing are not activated by the isolation procedure. This ensures that the effects measured by the test are the result of the environment or the applied stimulus, but is not an artifact resulting from activation by cell isolation. METHODS: Herein, we analyzed two different platelet isolation procedures (i.e., a Sepharose column versus density gradient centrifugation) with special emphasis on cell activation, including flow cytometric analysis of P-selectin expression, functional quantification of mechanical platelet retention, light microscopic assessment of platelet aggregation, and fluorescence microscopic determination of in vivo rat liver platelet-endothelium cell interaction. RESULTS: Under resting conditions, Sepharose column-isolated platelets showed a negligible fraction of only 2.7 +/- 3.3% cells (mean +/- SEM) with P-selectin expression, and an appropriate response (i.e., a 33-fold increase) upon activation with thrombin receptor-activating peptide (TRAP). In contrast, density gradient centrifugation resulted in P-selectin expression under resting conditions of approximately 50% of the isolated cells and only a 1.6-fold increase on further TRAP stimulation. In addition, density gradient-isolated platelets, but not Sepharose column-isolated platelets, showed increased mechanical retention and agglutination/aggregation in vitro, as well as pronounced adhesion to hepatic venular endothelium in vivo. Interestingly, density gradient-isolated platelets additionally induced in vivo an increase of colocalization of platelets with adherent leukocytes, indicating a generalized microvascular inflammatory response that is comparable to that observed after a 60-minute ischemia/30-minute reperfusion insult. CONCLUSION: Density gradient centrifugation-isolated platelets, but not Sepharose column-isolated platelets, are activated already under resting conditions and induce in vivo a platelet-leukocyte-endothelial cell-associated inflammatory response. Thus, we propose that the method of platelet isolation using the Sepharose column is superior to the density gradient centrifugation technique and might therefore be preferred for in vitro and in vivo assays to study platelet function.


Asunto(s)
Plaquetas/citología , Separación Celular/métodos , Separación Celular/normas , Centrifugación por Gradiente de Densidad/métodos , Cromatografía en Agarosa/métodos , Endotelio Vascular/citología , Venas Hepáticas/citología , Humanos , Microcirculación/citología , Fragmentos de Péptidos/farmacología , Activación Plaquetaria , Adhesividad Plaquetaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA