Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Vascul Pharmacol ; 154: 107276, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38242295

RESUMEN

INTRODUCTION: Cancer needs perfusion for its growth and metastasis. Cancer cell-derived extracellular vesicles (CA-EVs) alter the tumor microenvironment (TME), potentially promoting angiogenesis. We hypothesize that conditions in the tumor, e.g., hypoxia, and in the target cells of the TME, e.g., nutrient deprivation or extracellular matrix, can affect the angiogenic potential of CA-EVs, which would contribute to explaining the regulation of tumor vascularization and its influence on cancer growth and metastasis. METHODS: CA-EVs were isolated and characterized from cervical carcinoma cell lines HeLa and SiHa cultured under normoxia and hypoxia, and their angiogenic potential was evaluated in vitro in three endothelial cells (ECs) lines and aortic rings, cultured in basal (growth factor-reduced) or complete medium. RESULTS: Hypoxia increased EV production 10-100 times and protein content 2-4 times compared to normoxic CA-EVs. HeLa-EVs contained six times more RNA than SiHa-EVs, and this concentration was not affected by hypoxia. Treatment with CA-EVs increased tube formation and sprouting in ECs and aortic rings cultured in basal medium and long-term stabilized the stablished vascular networks formed by ECs cultured in complete medium. CONCLUSION: Hypoxia differentially affects CA-EVs in a cell line-dependent manner. The cellular environment (nutrient availability and extracellular matrix scaffold) influences the effect of CA-EV on the angiogenic potential of ECs.


Asunto(s)
Carcinoma , Vesículas Extracelulares , Humanos , Células Endoteliales/metabolismo , Angiogénesis , Vesículas Extracelulares/metabolismo , Carcinoma/metabolismo , Carcinoma/patología , Hipoxia/metabolismo , Microambiente Tumoral
2.
Transl Oncol ; 31: 101633, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36905871

RESUMEN

Vascularization is a multifactorial and spatiotemporally regulated process, essential for cell and tissue survival. Vascular alterations have repercussions on the development and progression of diseases such as cancer, cardiovascular diseases, and diabetes, which are the leading causes of death worldwide. Additionally, vascularization continues to be a challenge for tissue engineering and regenerative medicine. Hence, vascularization is the center of interest for physiology, pathophysiology, and therapeutic processes. Within vascularization, phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and Hippo signaling have pivotal roles in the development and homeostasis of the vascular system. Their suppression is related to several pathologies, including developmental defects and cancer. Non-coding RNAs (ncRNAs) are among the regulators of PTEN and/or Hippo pathways during development and disease. The purpose of this paper is to review and discuss the mechanisms by which exosome-derived ncRNAs modulate endothelial cell plasticity during physiological and pathological angiogenesis, through the regulation of PTEN and Hippo pathways, aiming to establish new perspectives on cellular communication during tumoral and regenerative vascularization.

3.
Sci Rep ; 11(1): 13493, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34188159

RESUMEN

The rate of the remodeling of the arterialized saphenous vein conduit limits the outcomes of coronary artery bypass graft surgery (CABG), which may be influenced by endothelial dysfunction. We tested the hypothesis that high stretch (HS) induces human saphenous vein endothelial cell (hSVEC) dysfunction and examined candidate underlying mechanisms. Our results showed that in vitro HS reduces NO bioavailability, increases inflammatory adhesion molecule expression (E-selectin and VCAM1) and THP-1 cell adhesion. HS decreases F-actin in hSVECs, but not in human arterial endothelial cells, and is accompanied by G-actin and cofilin's nuclear shuttling and increased reactive oxidative species (ROS). Pre-treatment with the broad-acting antioxidant N-acetylcysteine (NAC) supported this observation and diminished stretch-induced actin remodeling and inflammatory adhesive molecule expression. Altogether, we provide evidence that increased oxidative stress and actin cytoskeleton remodeling play a role in HS-induced saphenous vein endothelial cell dysfunction, which may contribute to predisposing saphenous vein graft to failure.


Asunto(s)
Actinas/metabolismo , Células Endoteliales/metabolismo , Estrés Oxidativo , Vena Safena/metabolismo , Estrés Mecánico , Humanos , Especies Reactivas de Oxígeno/metabolismo , Células THP-1
4.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L698-L704, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32048864

RESUMEN

Chronic lung diseases such as idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD) are associated with changes in extracellular matrix (ECM) composition and abundance affecting the mechanical properties of the lung. This study aimed to generate ECM hydrogels from control, severe COPD [Global Initiative for Chronic Obstructive Lung Disease (GOLD) IV], and fibrotic human lung tissue and evaluate whether their stiffness and viscoelastic properties were reflective of native tissue. For hydrogel generation, control, COPD GOLD IV, and fibrotic human lung tissues were decellularized, lyophilized, ground into powder, porcine pepsin solubilized, buffered with PBS, and gelled at 37°C. Rheological properties from tissues and hydrogels were assessed with a low-load compression tester measuring the stiffness and viscoelastic properties in terms of a generalized Maxwell model representing phases of viscoelastic relaxation. The ECM hydrogels had a greater stress relaxation than tissues. ECM hydrogels required three Maxwell elements with slightly faster relaxation times (τ) than that of native tissue, which required four elements. The relative importance (Ri) of the first Maxwell element contributed the most in ECM hydrogels, whereas for tissue the contribution was spread over all four elements. IPF tissue had a longer-lasting fourth element with a higher Ri than the other tissues, and IPF ECM hydrogels did require a fourth Maxwell element, in contrast to all other ECM hydrogels. This study shows that hydrogels composed of native human lung ECM can be generated. Stiffness of ECM hydrogels resembled that of whole tissue, while viscoelasticity differed.


Asunto(s)
Matriz Extracelular/metabolismo , Hidrogeles/metabolismo , Pulmón/metabolismo , Pulmón/fisiología , Rigidez Vascular/fisiología , Animales , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pepsina A/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Porcinos , Viscosidad
5.
J Biomed Mater Res A ; 106(9): 2494-2506, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29700983

RESUMEN

Abdominal aortic aneurysm (AAA) is the pathological dilation and weakening of the abdominal aorta wall. Inflammation, degradation of the extracellular matrix (ECM) and loss of smooth muscle cells and skewing of their function are pivotal in AAA pathology. We developed a recombinant collagen-based patch (RCP) to provide structural integrity and deliver Adipose tissue-Derived Stromal Cells (ASC) for repair. Patches supported adhesion and function as well as proliferation of ASC. ASC-loaded RCPs or bare patches, applied around the aorta after AAA induction in rats, both maintained structural integrity of the aortic wall at time of explant (2w). However, wall thinning, accompanied by loss of elastin fibers and loss of medial SMC, was only attenuated in ASC-loaded RCP-treated AAA rats. Interestingly, this coincided with migration of ASC into the media and a reduced influx of macrophages. We hypothesize that the medially-migrated ASC dampened or skewed the adverse innate immunity and thus suppressed SMC apoptosis, phenotypic skewing and elastin degradation. We conclude that the periadventitial delivery of ASC with RCP suppresses development and progression of AAA, which is has an expected future clinical benefit in combination with an appropriate early screening program of patients at risk for aneurysms. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A:2494-2506, 2018.


Asunto(s)
Tejido Adiposo/citología , Aneurisma de la Aorta Abdominal/terapia , Vasos Sanguíneos/fisiología , Andamios del Tejido/química , Animales , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/patología , Vasos Sanguíneos/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Dilatación , Elastina/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Proteolisis/efectos de los fármacos , Ratas Endogámicas F344 , Proteínas Recombinantes/farmacología , Células del Estroma/citología , Propiedades de Superficie , Cicatrización de Heridas/efectos de los fármacos
6.
J Tissue Eng Regen Med ; 7(2): 118-28, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22125235

RESUMEN

Cell therapy strategies that use adult peripheral blood-derived CD34⁺ progenitor cells are hampered by low cell numbers and the infrequent cellular incorporation into the neovasculature. Hence, the use of CD34⁺ cells to treat ischaemic diseases is under debate. Interaction between CD34⁺ cells and CD14⁺ cells results in superior endothelial differentiation of CD14⁺ cells in vitro, indicating that cell therapy approaches utilizing both CD34⁺ and CD14⁺ cells may be advantageous in therapeutic neovascularization. Here, human CD34⁺ and CD14⁺ cells were isolated from adult peripheral blood and implanted subcutaneously into nude mice, using matrigel as the carrier. Combined implantation of human CD34⁺ and CD14⁺ cells resulted in superior neovascularization, compared to either cell type alone, albeit incorporation of human cells into the murine vasculature was not observed. Human CD34⁺ and CD14⁺ cells produced and secreted a pentad of pro-angiogenic mediators, such as HGF, MCP-1 and IL-8, bFGF and VEGFa in monoculture. The production and secretion of pro-angiogenic mediators by CD14⁺ cells was highly amplified upon incubation with conditioned medium from CD34⁺ cells. In vivo, neovascularization of matrigel implants did not rely on the endothelial differentiation and incorporation of CD34⁺ or CD14⁺ cells, but depended on the paracrine effects of IL-8, MCP-1, HGF, bFGF and VEGFa secreted by implanted cells. Administration of this growth factor/cytokine pentad using matrigel as a carrier results in cell recruitment and microvessel formation equal to progenitor cell-induced neovascularization. These data provide new insights on neovascularization by cell therapy and may contribute to new strategies for the treatment of ischaemic diseases.


Asunto(s)
Antígenos CD34/metabolismo , Trasplante de Células , Receptores de Lipopolisacáridos/metabolismo , Neovascularización Fisiológica , Comunicación Paracrina , Animales , Colágeno/farmacología , Combinación de Medicamentos , Humanos , Laminina/farmacología , Masculino , Ratones , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Proteoglicanos/farmacología
7.
Eur Cell Mater ; 24: 320-330, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23070944

RESUMEN

An array of different types of hyaluronic acid (HA)- and collagen-based products is available for filling soft-tissue defects. A major drawback of the current soft-tissue fillers is their inability to induce cell infiltration and new tissue formation. Our aim is to develop novel biodegradable injectable gels which induce soft tissue regeneration, initially resulting in integration and finally replacement of the gel with new autologous tissue. Two reference gels of pure HA, monophasic HA-1 and micronised HA-2, were used. Furthermore, both gels were mixed with recombinant gelatin (RG) resulting in HA-1+RG and HA-2+RG. All gels were subcutaneously injected on the back of rats and explanted after 4 weeks. Addition of RG to HA-1 resulted in stroma formation (neovascularisation and ECM deposition) which was restricted to the outer rim of the HA-1+RG gel. In contrast, addition of RG to HA-2 induced stroma formation throughout the gel. The RG component of the gel was degraded by macrophages and giant cells and subsequently replaced by new vascularised tissue. Immunohistochemical staining showed that the extracellular matrix components collagen I and III were deposited throughout the gel. In conclusion, this study shows the proof of principle that addition of RG to HA-2 results in a novel injectable gel capable of inducing soft tissue regeneration. In this gel HA has a scaffold function whereas the RG component induces new tissue formation, resulting in proper vascularisation and integration of the HA-2+RG gel with the autologous tissue.


Asunto(s)
Gelatina/química , Ácido Hialurónico/química , Regeneración , Tejido Subcutáneo/fisiología , Andamios del Tejido/química , Animales , Gelatina/genética , Geles/química , Masculino , Ratas , Proteínas Recombinantes/química
8.
J Tissue Eng Regen Med ; 5(4): 264-74, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20661871

RESUMEN

We have previously shown that the foreign body reaction (FBR) against crosslinked collagen type I (Col-I) differs between subcutaneous and epicardial implantation sites; Col-I was quickly degraded epicardially, whereas degradation was attenuated subcutaneously. The current study set out to dissect the nature and regulation of the MMP-based degradation of implanted Col-I in mice during the FBR. Immunohistochemistry showed that MMP-2, MMP-8 and MMP-13 were present in subcutaneous and epicardial implants, whereas only MMP-9 was also present epicardially. Western blotting showed that MMP-8 and MMP-9 were mainly present in their inactive proform. In contrast, collagenase MMP-13 and gelatinase MMP-2 were the predominant active MMPs at both sites. Interestingly, the major MMP inhibitor TIMP-1 was solely observed in subcutaneous implants, which is why MMP-13 and MMP-2 are not able to degrade the collagen scaffold at the subcutaneous implantation site. Interleukin 10 (IL-10), a potent inducer of TIMP-1 expression, was also mainly detected subcutaneously; giant cells were the main source. Therefore, we surmise that IL-10, through regulation of the balance between MMPs and TIMP-1, suppresses the FBR against implanted biomaterials. Together, our findings would provide cues and clues to improve future therapies in regenerative medicine that are based on the tuned regulation of the degradation of biomaterial scaffolds.


Asunto(s)
Colágeno/metabolismo , Reactivos de Enlaces Cruzados/metabolismo , Interleucina-10/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Procesamiento Proteico-Postraduccional , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Andamios del Tejido/química , Animales , Western Blotting , Bovinos , Colagenasas/metabolismo , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Gelatinasas/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Tejido Subcutáneo/efectos de los fármacos , Tejido Subcutáneo/metabolismo , Factores de Tiempo
9.
Eur Cell Mater ; 20: 197-209, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-20931490

RESUMEN

Subcutaneously implanted disks of hexamethylenediisocyanate or glutaraldehyde cross-linked sheep collagen (referred to as HDSC and GDSC, respectively) in mice show large differences in degradation rate. Although comparable numbers of macrophages are seen in HDSC and GDSC, phagocytosis of collagen by macrophages occurred only in GDSC. The molecular mechanisms involved in the phagocytosis of collagen by macrophages are essentially unknown. Immunofluorescence and RT-PCR showed that Endo180 was expressed in GDSC only. TissueFaxs showed that Endo180 co-localized with MT1-MMP on F4÷80 positive cells, which is likely responsible for the phagocytosis in GDSC. RT-PCR further showed that Endo180 expression correlated with high levels of IFN-γ mRNA. In vitro, IFN-γ induced the expression Endo180 and MT1-MMP in murine macrophages cultured on collagen type I (although too high levels of IFN-γ dampened the expression of Endo180 and MT1-MMP). Moreover, the expression of Endo180 and MT1-MMP induced by IFN-γ can be inhibited through IL-10. The differences in microenvironment between GDSC and HDSC (high IFN-γ and low IL-10 levels in GDSC, low IFN-γ and high IL-10 levels in HDSC) provide an explanation why phagocytosis of collagen by macrophages is only seen in GDSC. In summary, we show for the first time that the IFN-γ dependent co-expression of Endo180 and MT1-MMP on macrophages coincides with collagen phagocytosis, thus providing evidence that the mechanism of collagen phagocytosis operating in the foreign body reaction by macrophages is comparable with the mechanism of intracellular collagen degradation by fibroblasts seen under physiological conditions.


Asunto(s)
Colágeno/metabolismo , Interferón gamma/metabolismo , Macrófagos/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Fagocitosis/fisiología , Receptores Mitogénicos/metabolismo , Andamios del Tejido , Animales , Interleucina-10/metabolismo , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Ovinos
10.
J Biomed Mater Res A ; 95(1): 198-208, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20574980

RESUMEN

Tissue engineering and regenerative medicine have furnished a vast range of modalities to treat either damaged tissue or loss of soft tissue or its function. In most approaches, a temporary porous scaffold is required to support tissue regeneration. The scaffold should be designed such that the turnover synchronizes with tissue remodeling and regeneration at the implant site. Segmented polyester urethanes (PUs) used in this study were based on epsilon-caprolactone (CL) and co-monomers D,L-lactide (D,L-L) and gamma-butyrolactone (BL), and 1,4-butanediisocyanate (BDI). In vitro, the PUs were nontoxic and haemocompatible. To test in vivo biocompatibility, the PUs were further processed into porous structures and subcutaneously implanted in rats for a period up to 21 days. Tissue remodeling and scaffold turnover was associated with a mild tissue response. The tissue response was characterized by extensive vascularization through the interconnected pores, with low numbers of macrophages on the edges and stroma formation inside the pores of the implants. The tissue ingrowth appeared to be related to the extent of microphase separation of the PUs and foam morphology. By day 21, all of the PU implants were highly vascularized, confirming the pores were interconnected. Degradation of P(CL/D,L-L)-PU was observed at this time, whereas the other two PU types remained intact. The robust method reported here of manufacturing and processing, good mechanical properties, and in vivo tissue response of the porous P(CL/D,L-L)-PU and PBCL-PU makes them excellent candidates as biomaterials with an application for soft tissue remodeling, for example, for cardiovascular regeneration.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Poliuretanos/química , Poliuretanos/farmacología , Ingeniería de Tejidos/métodos , Animales , Muerte Celular/efectos de los fármacos , Cristalización , Endotoxinas/metabolismo , Masculino , Ensayo de Materiales , Microscopía Electrónica de Rastreo , Poliuretanos/síntesis química , Porosidad/efectos de los fármacos , Implantación de Prótesis , Ratas , Ratas Wistar , Sus scrofa
11.
J Cell Mol Med ; 13(8B): 2521-2533, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18752636

RESUMEN

Neovascularization by endothelial progenitor cells (EPC) for the treatment of ischaemic diseases has been a topic of intense research. The CD34(+) cell is often designated as EPC, because it contributes to repair of ischaemic injuries through neovascularization. However, incorporation of CD34(+) cells into the neovasculature is limited, suggesting another role which could be paracrine. CD14(+) cells can also differentiate into endothelial cells and contribute to neovascularization. However, the low proliferative capacity of CD14(+) cell-derived endothelial cells hampers their use as therapeutic cells. We made the assumption that an interaction between CD34(+) and CD14(+) cells augments endothelial differentiation of the CD14(+) cells. In vitro, the influence of CD34(+) cells on the endothelial differentiation capacity of CD14(+) cells was investigated. Endothelial differentiation was analysed by expression of endothelial cell markers CD31, CD144, von Willebrand Factor and endothelial Nitric Oxide Synthase. Furthermore, we assessed proliferative capacity and endothelial cell function of the cells in culture. In monocultures, 63% of the CD14(+)-derived cells adopted an endothelial cell phenotype, whereas in CD34(+)/CD14(+) co-cultures 95% of the cells showed endothelial cell differentiation. Proliferation increased up to 12% in the CD34(+)/CD14(+) co-cultures compared to both monocultures. CD34-conditioned medium also increased endothelial differentiation of CD14(+) cells. This effect was abrogated by hepatocyte growth factor neutralizing antibodies, but not by interleukin-8 and monocyte chemoattractant protein-1 neutralizing antibodies. We show that co-culturing of CD34(+) and CD14(+) cells results in a proliferating population of functional endothelial cells, which may be suitable for treatment of ischaemic diseases such as myocardial infarction.


Asunto(s)
Antígenos CD34/inmunología , Diferenciación Celular/inmunología , Endotelio Vascular/inmunología , Receptores de Lipopolisacáridos/inmunología , Células Madre/inmunología , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Endotelio Vascular/citología , Humanos , Células Madre/citología
12.
Biomaterials ; 30(5): 730-5, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19038442

RESUMEN

The foreign body reaction (FBR) is of great importance for the function and turnover of biomaterial scaffolds. The development of biological tools that modulate the FBR will augment scaffold functionality and benefit regenerative medicine. The human cytomegalovirus encodes a functional homolog of the potent anti-inflammatory human cytokine interleukin-10 (cmvIL-10). We hypothesized that cmvIL-10 downmodulates the FBR, impairing degradation of biomaterial. We studied the effect of cmvIL-10 on the FBR to subcutaneously implanted hexamethylenediisocyanate-crosslinked dermal sheep collagen (HDSC) discs in rats. CmvIL-10 impaired macrophage influx, vascularization and ingrowth into the discs up to 21 days. It also impaired the formation of giant cells and the degradation of HDSC. At day 10, deposited fibrin fibers were still present in cmvIL-10 discs. Impaired collagenase activity coincided with the impaired HDSC degradation. These results indicate that cmvIL-10 downmodulates the FBR, impairing the progression of the FBR. This study demonstrates the feasibility of interleukin-10 as a biomolecular tool in biomaterials for regenerative medicine.


Asunto(s)
Implantes Absorbibles/efectos adversos , Citomegalovirus/metabolismo , Reacción a Cuerpo Extraño/inmunología , Reacción a Cuerpo Extraño/prevención & control , Interleucina-10/farmacología , Animales , Colágeno , Reactivos de Enlaces Cruzados/química , Cianatos , Células Gigantes de Cuerpo Extraño/inmunología , Inmunohistoquímica , Isocianatos , Macrófagos/metabolismo , Masculino , Ratas
13.
J Pathol ; 216(2): 201-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18702175

RESUMEN

The human epithelial cell adhesion molecule (hEpCAM) is involved in epithelial morphogenesis and repair of epithelial tissues. We hypothesized that changes in hEpCAM expression in vivo correlate with regeneration of renal epithelia after ischaemia/reperfusion injury (IRi). Unilateral IRi was performed on kidneys of hEpCAM transgenic mice. Changes in hEpCAM expression were investigated by quantitative RT-PCR in renal cortex and medulla dissected by laser dissection microscopy and expression patterns of hEpCAM in regenerating kidneys were assessed by immunohistochemistry. The mechanism of hEpCAM promoter activation was investigated in vitro, by real-time bioluminescent imaging in HK-2 cells and in primary tubular epithelial cells (PTECs) subjected to hypoxia and reoxygenation. In vivo, the transcription of the human epcam gene significantly increased in the renal cortex during tubular re-epithelialization (p < 0.01). Moreover, the number of tubuli that expressed hEpCAM protein more than doubled in the renal cortex during regeneration. De novo expression of hEpCAM was detected in the S1 segments of proximal tubuli. Under hypoxic conditions in vitro, activity of the hEpCAM promoter was up-regulated two-fold in the HK-2 proximal epithelial cell line. Moreover, both in primary proximal epithelial cells and in HK-2 cells, hEpCAM protein expression was increased after hypoxia and reoxygenation. The significant up-regulation of hEpCAM during post-ischaemic renal regeneration in vivo and during in vitro hypoxia indicates that hEpCAM expression is associated with renal regeneration.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/patología , Riñón/patología , Riñón/fisiología , Regeneración , Regulación hacia Arriba , Animales , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/genética , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/genética , Línea Celular , Molécula de Adhesión Celular Epitelial , Células Epiteliales/metabolismo , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Inmunohistoquímica , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
14.
J Pathol ; 214(3): 377-86, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18095257

RESUMEN

Myofibroblasts play a major role in scar formation during wound healing after myocardial infarction (MI). Their origin has been thought to be interstitial cardiac fibroblasts. However, the bone marrow (BM) can be a source of myofibroblasts in a number of organs after injury. We have studied the temporal, quantitative and functional role of BM-derived (BMD) myofibroblasts in myocardial scar formation. MI was induced by permanent coronary artery ligation in mice reconstituted with EGFP or pro-Col1A2 transgenic BM. In the latter, luciferase and beta-galactosidase transgene expression mirrors that of the endogenous pro-collagen 1A2 gene, which allows for functional assessment of the recruited cells. After MI, alpha-SMA-positive myofibroblasts and collagen I gradually increased in the infarct area until day 14 and remained constant afterwards. Numerous EGFP-positive BMD cells were present during the first week post-MI, and gradually decreased afterwards until day 28. Peak numbers of BMD myofibroblasts, co-expressing EGFP and alpha-SMA, were found on day 7 post-MI. An average of 21% of the BMD cells in the infarct area were myofibroblasts. These cells constituted up to 24% of all myofibroblasts present. By in vivo IVIS imaging, BMD myofibroblasts were found to be active for collagen I production and their presence was confined to the infarct area. These results show that BMD myofibroblasts participate actively in scar formation after MI.


Asunto(s)
Células de la Médula Ósea/patología , Cicatriz/patología , Infarto del Miocardio/patología , Miocardio/patología , Cicatrización de Heridas , Actinas/análisis , Actinas/genética , Animales , Cicatriz/enzimología , Colágeno/genética , Colágeno/metabolismo , Colágeno Tipo I , Fibroblastos/patología , Expresión Génica , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Ligadura , Luciferasas/análisis , Luciferasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Infarto del Miocardio/enzimología , Miocardio/enzimología , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
15.
J Biomed Mater Res A ; 83(3): 879-86, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17567860

RESUMEN

Multinucleated giant cells (GCs) are often observed in the foreign body reaction against implanted materials. The in vivo function of GCs in this inflammatory process remains to be elucidated. GCs degrade collagen implants in rats and may also orchestrate the inflammatory process via the expression and secretion of modulators, such as cytokines and chemokines. In this study, we show that the gene expression of PMN chemoattractants, CXCL1/KC and CXCL2/MIP-2, is high in GCs micro-dissected from explanted Dacron, cross-linked collagen (HDSC), and bioactive ureido-pyrimidinone functionalized oligocaprolactone (bioactive PCLdiUPy). Conversely, the gene expression levels of TGFbeta and pro-angiogenic mediators VEGF and FGF were found to be low in these GCs as compared with the expression levels in total explants. GCs in bioactive PCLdiUPy displayed high cytokine and angiogenic mediator expression compared with GCs isolated from the two other studied materials, whereas chemokine gene expression in GCs isolated form HDSC was low. Thus, GCs adopt their expression profile in response to the material that is encountered.


Asunto(s)
Materiales Biocompatibles/efectos adversos , Citocinas/biosíntesis , Reacción a Cuerpo Extraño/metabolismo , Células Gigantes de Cuerpo Extraño/metabolismo , Mediadores de Inflamación/metabolismo , Prótesis e Implantes/efectos adversos , Animales , Reacción a Cuerpo Extraño/patología , Regulación de la Expresión Génica , Células Gigantes de Cuerpo Extraño/patología , Ensayo de Materiales , Ratas
16.
J Mol Cell Cardiol ; 42(6): 1086-97, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17490680

RESUMEN

CD34+ progenitor cells hold promise for therapeutic neovascularization in various settings. In this study, the role of human peripheral blood CD34+ cells in neovascularization and inflammatory cell recruitment was longitudinally studied in vivo. Human CD34+ cells were incorporated in Matrigel, implanted subcutaneously in nude mice, and explanted after 2, 4, 7, or 14 days. Cell-free Matrigels served as controls. Histochemical analyses demonstrated that neovascularization occurred almost exclusively in CD34+ implants. Cellular and capillary density were increased in cell-loaded Matrigels after 2 days and further increased at 14 days. Human CD34+ cells did not incorporate in neovessels, but formed vWF+/CD31+/VEGF+ cell clusters that were present up to day 14. However, CD34+ cells induced host neovascularization, as demonstrated by increased presence of murine CD31+ and vWF+ vasculature from day 7 to 14. Moreover, recruitment of murine monocytes/macrophages was significantly enhanced in CD34+ implants at all time points. Gene expression of chemotactic cytokines MCP-1 and IL-8 was detected on CD34+ cells in vitro and confirmed immunohistochemically in cell-loaded explants at all time points. Our data indicate that human CD34+ cells, implanted in a hypoxic environment, generate an angiogenic niche by secreting chemotactic and angiogenic factors, enabling rapid neovascularization, possibly via recruitment of monocytes/macrophages.


Asunto(s)
Antígenos CD34/metabolismo , Células Madre Hematopoyéticas/inmunología , Inflamación/inmunología , Neovascularización Fisiológica/inmunología , Animales , Antígenos CD34/administración & dosificación , Antígenos CD34/genética , Células Cultivadas , Quimiocina CCL2/metabolismo , Colágeno , Modelos Animales de Enfermedad , Combinación de Medicamentos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Inmunohistoquímica , Inyecciones Subcutáneas , Interleucina-8/metabolismo , Laminina , Macrófagos/citología , Masculino , Ratones , Ratones Desnudos , Monocitos/citología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteoglicanos , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Cancer Gene Ther ; 13(2): 150-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16096650

RESUMEN

Adenoviral vectors are widely used in cancer gene therapy. After systemic administration however, the majority of the virus homes to the liver and the expressed transgene may cause hepatotoxicity. To restrict transgene expression to tumor cells, tumor- or tissue-specific promoters are utilized. The tumor antigen epithelial glycoprotein-2 (EGP-2), also known as Ep-CAM, is expressed in many cancers from different epithelial origins. In this study, the EGP-2 promoter was shown to restrict the expression of luciferase and thymidine kinase in an adenoviral context in different cell lines. In vivo, the EGP-2 promoter mediated efficient expression of luciferase in tumors but showed a 3-log lower activity in liver tissue when compared with the cytomegalovirus (CMV) promoter. Similarly, the EGP-2 promoter mediated specific cell killing after ganciclovir treatment in EGP-2-positive cells. Moreover, in vivo, this treatment regiment did not cause any rise in the liver enzymes aspartate aminotransferase (ASAT) and alanine aminotransferase (ALAT), demonstrating absence of liver toxicity. In contrast, CMV-mediated expression of thymidine kinase in combination with ganciclovir treatment resulted in high ASAT and ALAT values. This study demonstrates the value of the EGP-2 promoter to restrict transgene expression to a broad range of tumor types, thereby preventing liver toxicity.


Asunto(s)
Antígenos de Neoplasias/genética , Moléculas de Adhesión Celular/genética , Regulación Neoplásica de la Expresión Génica , Terapia Genética/métodos , Neoplasias/terapia , Regiones Promotoras Genéticas/genética , Adenoviridae , Alanina Transaminasa/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Línea Celular Tumoral , Cartilla de ADN , Molécula de Adhesión Celular Epitelial , Ganciclovir/toxicidad , Vectores Genéticos/genética , Humanos , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timidina Quinasa/metabolismo , Timidina Quinasa/toxicidad , Pruebas de Toxicidad , Transgenes/genética
18.
Antiviral Res ; 69(2): 124-7, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16368154

RESUMEN

The human cytomegalovirus has found smart ways to exploit the chemokine network in order to subvert immune attack. Chemokines trigger the arrest and firm adhesion of inflammatory cells to the vascular wall. Scavenging of chemokines by viral decoy receptors, such as US28, might prevent arrest of leukocytes to the vascular wall and impair an antiviral immune response. We determined the effect of chemokine scavenging by endothelium-expressed signaling mute US28 (US28R129A) on static monocyte adhesion. Despite the chemokine scavenging capacity of US28R129A, expression of this construct by endothelial cells was insufficient to disrupt leukocyte adhesion to cytokine-activated monolayers. Our results suggest that the concentrations of chemokines that trigger firm leukocyte adhesion are too high to be efficiently scavenged by viral chemokine decoy receptors like US28. From the results of this experimental model a role for US28 in viral immune evasion by chemokine scavenging would appear therefore unlikely.


Asunto(s)
Adhesión Celular/inmunología , Quimiocinas/metabolismo , Células Endoteliales/fisiología , Endotelio Vascular/inmunología , Monocitos/fisiología , Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Células Endoteliales/inmunología , Humanos , Monocitos/inmunología , Venas Umbilicales
19.
J Mol Cell Cardiol ; 39(2): 363-76, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15992820

RESUMEN

Myocardial infarction leads to scar formation and subsequent reduced cardiac performance. The ultimate therapy after myocardial infarction would pursue stem cell-based regeneration. The aim of stem cell-mediated cardiac repair embodies restoration of cardiac function by regeneration of healthy myocardial tissue, which is accomplished by neo-angiogenesis and cardiogenesis. A major reservoir of adult autologous stem cells distal from the heart is the bone marrow. Adequate regulation of signaling between the bone marrow, the peripheral circulation and the infarcted myocardium is important in orchestrating the process of mobilization, homing, incorporation, survival, proliferation and differentiation of stem cells, that leads to myocardial regeneration. In this review, we discuss key signaling factors, including cytokines, chemokines and growth factors, which are involved in orchestrating the stem cell driven repair process. We focus on signaling factors known for their mobilizing and chemotactic abilities (SDF-1, G-CSF, SCF, IL-8, VEGF), signaling factors that are expressed after myocardial infarction involved in the patho-physiological healing process (TNF-alpha, IL-8, IL-10, HIF-1alpha, VEGF, G-CSF) and signaling factors that are involved in cardiogenesis and neo-angiogenesis (VEGF, EPO, TGF-beta, HGF, HIF-1alpha, IL-8). The future therapeutic application and capacity of secreted factors to modulate tissue repair after myocardial infarction relies on the intrinsic potency of factors and on the optimal localization and timing of a combination of signaling factors to stimulate stem cells in their niche to regenerate the infarcted heart.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Sustancias de Crecimiento/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Transducción de Señal , Células Madre/metabolismo , Animales , Humanos , Infarto del Miocardio/patología , Trasplante de Células Madre
20.
Rev Med Virol ; 15(4): 269-82, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15861487

RESUMEN

Mimicking host proteins is a strategy adopted by several herpesviruses to exploit the host cell for their own benefit. In this respect the human cytomegalovirus (HCMV) chemokine receptor homologue US28, has been extensively studied. Molecular pirates such as US28 can teach us about crucial events in HCMV infection and may either offer a potential target for antiviral therapy or provide an alternative strategy to immune suppression. Despite elaborate research into the chemokine binding affinity, signalling properties, intracellular trafficking and expression kinetics of US28, a solid hypothesis about the role of US28 in HCMV infection has not yet been proposed. It appears that US28 may behave as a molecular pirate that employs smart strategies for cell entry, host gene regulation and immune evasion. This review will elaborate on these aspects of US28 biology and discuss possible implications for HCMV infection.


Asunto(s)
Infecciones por Citomegalovirus/fisiopatología , Citomegalovirus/fisiología , Receptores de Quimiocina/fisiología , Proteínas Virales/fisiología , Arteriosclerosis/etiología , Quimiocina CX3CL1 , Quimiocinas CX3C/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/fisiología , Miocitos del Músculo Liso/virología , Transducción de Señal , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA