Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 30(3): 703-11, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20392987

ABSTRACT

Tumor invasion requires intense interactions with stromal cells and a profound extracellular matrix remodelling by matrix metalloproteinases (MMPs). Here, we assessed the specific contribution of fibroblasts to tumor invasion, MMPs, tissue inhibitors of MMPs and angiogenesis-related cytokine expression in organotypic cultures of highly malignant HaCaT-ras A-5RT3 cells, with and without MMP inhibition. Collagen degradation, the hallmark of tumor invasion, was dependent on fibroblasts and active MMP-2. Additionally, MMP blockade down-regulated VEGF-A and up-regulated PDGF-BB. These results were paralleled in xenotransplants in vivo, demonstrating strong inhibitory effects of MMP blockade on tumor invasion and vascularization, as shown by the almost complete absence of VEGF-A and MMP-14 and by the decrease in relative blood volume. MMP blockade also increased the fraction of mature vessels, as demonstrated by an increased mean tumor vessel diameter and a higher ratio of Ng2-positive vessels. Thus, this study highlights the importance of targeting the tumor stroma to defeat cancer.


Subject(s)
Matrix Metalloproteinases, Secreted/antagonists & inhibitors , Platelet-Derived Growth Factor/biosynthesis , Skin Neoplasms/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Becaplermin , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Collagen/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Matrix Metalloproteinases, Secreted/biosynthesis , Matrix Metalloproteinases, Secreted/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Organic Chemicals/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-sis , Pyrimidines/pharmacology , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/metabolism , Xenograft Model Antitumor Assays
2.
Am J Pathol ; 176(2): 981-94, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20042679

ABSTRACT

Vascular endothelial growth factor (VEGF), which is a key regulator of angiogenesis, often induces formation of immature vessels with increased permeability and reduced vessel functionality. Here, we demonstrate that de novo expression of murine (m)VEGF-164 induces malignant and invasive tumor growth of HaCaT keratinocytes. However, the mVEGF-164-induced tumors are ulcerated with a disorganized epithelium that is interrupted by lacunae with limited basement membrane and endothelial cell coverage. Vessel maturation is strongly impaired. Tumor and vessel micromorphology are markedly improved by the combined expression of human platelet-derived growth factor (hPDGF)-B and mVEGF-164. Although tumor size and malignancy are comparable with either mVEGF-164 alone or combined human PDGF-B and mVEGF-164 expression, combined hPDGF-B and mVEGF-164 expression leads to a more solid and compact tumor tissue with a mature functional tumor vasculature and a higher microvessel density, as demonstrated histologically and by dynamic contrast-enhanced magnetic resonance imaging. Treatment of the hPDGF-B- and mVEGF-164-expressing tumors with imatinib mesylate to block PDGF-B signaling reverses this effect. In addition, tumor cell invasion of mVEGF-164 transfectants and mVEGF-164 plus hPDGF-B transfectants in vivo is associated with a marked induction of tumor-derived matrix metalloproteinase-1 and stromal matrix metalloproteinase-9 and -13, as was confirmed in three-dimensional organotypic co-cultures with fibroblasts in vitro. These data clearly demonstrate the need for a concerted action of different growth factors in the establishment of solid tumors with functional vasculature and emphasize the need for a multifactorial therapy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Neovascularization, Pathologic/physiopathology , Proto-Oncogene Proteins c-sis/physiology , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics , Animals , Benzamides , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/ultrastructure , Cell Proliferation , Cells, Cultured , Humans , Imatinib Mesylate , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-sis/genetics , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Skin Neoplasms/blood supply , Skin Neoplasms/genetics , Skin Neoplasms/ultrastructure , Transfection , Transplantation, Heterologous , Tumor Burden/genetics
3.
Radiology ; 253(2): 462-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19789239

ABSTRACT

PURPOSE: To investigate the biologic effect of arginine-glycine-aspartic acid (RGD)-labeled ultrasmall superparamagnetic iron oxide (USPIO) (referred to as RGD-USPIO) on human umbilical vein endothelial cells (HUVECs), ovarian carcinoma (MLS) cells, and glioblastoma (U87MG) cells and on U87MG xenografts in vivo. MATERIALS AND METHODS: All experiments were approved by the governmental review committee on animal care.USPIOs were coated with integrin-specific (RGD) or unspecific (arginine-alanine-aspartic acid [RAD]) peptides. USPIO uptake in HUVECs, MLS cells, and U87MG cells and in U87MG tumor xenografts was determined with T2 magnetic resonance (MR) relaxometry in 16 nude mice. Cells and tumors were characterized by using immunofluorescence microscopy. Trypan blue staining and lactate dehydrogenase assay were used to assess cytotoxicity. Statistical evaluation was performed by using a Mann-Whitney test or a linear mixed model with random intercept for the comparison of data from different experiments. Post hoc pairwise comparisons were adjusted according to a Tukey test. RESULTS: HUVECs and MLS cells internalized RGD-USPIOs significantly more than unspecific probes. Controversially, U87MG cells accumulated RGD-USPIOs to a lesser extent than USPIO. Furthermore, only in U87MG cells, free RGD and alpha(v)beta(3) integrin-blocking antibodies strongly reduced endocytosis of nonspecific USPIOs. This was accompanied by a loss of cadherin-dependent intercellular contacts, which could not be attributed to cell damage. In U87MG tumors, RGD-USPIO accumulated exclusively at the neovasculature but not within tumor cells. The vascular accumulation of RGD-USPIO caused significantly higher changes of the R2 relaxation rate of tumors than observed for USPIO. CONCLUSION: In glioma cells with unstable intercellular contacts, inhibition of alpha(v)beta(3) integrins by antibodies and RGD and RGD-USPIO disintegrated intercellular contacts and reduced endocytotic activity, illustrating the risk of inducing biologic effects by using molecular MR probes.


Subject(s)
Cell Adhesion/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/physiology , Contrast Media/pharmacology , Dextrans/pharmacology , Endocytosis/drug effects , Endothelium, Vascular/metabolism , Ferrosoferric Oxide/pharmacology , Glioma/metabolism , Integrin alphaVbeta3/metabolism , Oligopeptides/pharmacology , Animals , Cell Survival , Cells, Cultured , Contrast Media/pharmacokinetics , Dextrans/pharmacokinetics , Female , Ferrosoferric Oxide/pharmacokinetics , Fluorescent Antibody Technique , Glioblastoma , Glioma/blood supply , Glioma/physiopathology , Humans , Magnetic Resonance Imaging , Magnetite Nanoparticles , Mice , Mice, Nude , Neoplasm Transplantation , Oligopeptides/pharmacokinetics , Ovarian Neoplasms , Umbilical Veins
4.
J Magn Reson Imaging ; 30(5): 1163-70, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19856450

ABSTRACT

PURPOSE: To measure longitudinal relaxation rate (R1) changes during contrast agent studies using a driven equilibrium single pulse observation of T1 (DESPOT) method with a sliding window (sw) acquisition. MATERIALS AND METHODS: A sw-DESPOT technique was implemented that uses several three-dimensional (3D) image data sets to calculate R1 with a temporal resolution of only a single data set. Different sources of systematic errors were studied in simulations, and the technique was tested in a tumor-bearing mouse using an intravascular contrast agent. RESULTS: Consistent concentration distributions of the CA were calculated with a temporal resolution of 10 s. CONCLUSION: Sw-DESPOT offers a precise and fast method to monitor the CA dynamics in 3D volumes.


Subject(s)
Contrast Media/pharmacology , Magnetic Resonance Imaging/methods , Algorithms , Animals , Humans , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Mice , Models, Statistical , Phantoms, Imaging , Software , Time Factors
5.
J Magn Reson Imaging ; 29(5): 1125-33, 2009 May.
Article in English | MEDLINE | ID: mdl-19388117

ABSTRACT

PURPOSE: To assess vascular remodeling in tumors during two different antiangiogenic therapies with dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and vessel size imaging and to evaluate the vessel size index (VSI) as a novel biomarker of therapy response. MATERIALS AND METHODS: In two independent experiments, nude mice bearing human skin squamous cell carcinoma xenografts were treated with a vascular endothelial growth factor (VEGF) inhibitor (bevacizumab) or a multitargeted tyrosine kinase inhibitor (SU11248). Changes in tumor vascularity were assessed by DCE-MRI and vessel size imaging. DCE-MRI data were analyzed applying a two-compartment model (Brix), calculating the parameters Amplitude and k(ep). RESULTS: For both experiments Amplitude decreased significantly in treated tumors while k(ep) did not change significantly. VSI showed controversial results. VSI was significantly increased in SU11248-treated A431 tumors, whereas no changes were found in bevacizumab-treated HaCaT-ras-A-5RT3 tumors. Immunohistology confirmed these results and suggest differences in the maturation of tumor vascularization as a possible explanation. CONCLUSION: DCE-MRI and vessel size imaging provide reliable and supplementing biomarkers of antiangiogenic therapy response. The results of both methods are in excellent agreement with histology. Nevertheless, our results also indicate that vascular remodeling is complex and that a uniform response cannot be expected for different tumors and therapies.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Blood Vessels/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Magnetic Resonance Imaging/methods , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Animals , Antineoplastic Agents/administration & dosage , Blood Vessels/drug effects , Carcinoma, Squamous Cell/blood supply , Contrast Media , Female , Indoles/administration & dosage , Mice , Mice, Nude , Pyrroles/administration & dosage , Sunitinib , Treatment Outcome
6.
Invest Radiol ; 43(3): 162-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18301312

ABSTRACT

OBJECTIVES: To assess the pharmacodynamic behavior of cyanoacrylate, streptavidin-coated microbubbles (MBs) and to investigate their suitability for molecular ultrasound imaging. MATERIALS AND METHODS: Biodistribution of MBs was analyzed in tumor-bearing mice using gamma-counting, immunohistochemistry, flow cytometry, and ultrasound. Further, vascular endothelial growth factor receptor 2-antibody coupled MBs were used to image tumor neovasculature. RESULTS: After 1 minute >90% of MBs were cleared from the blood and pooled in the lungs, liver, and spleen. Subsequently, within 1 hour a decent reincrease of MB-concentration was observed in the blood. The remaining MBs were removed by liver and spleen macrophages. About 30% of the phagocytosed MBs were intact after 48 hours. Shell fragments were found in the kidneys only. No relevant MB-accumulation was observed in tumors. In contrast, vascular endothelial growth factor receptor 2-specific MBs accumulated significantly within the tumor vasculature (P < 0.05). CONCLUSIONS: The pharmacokinetic behavior of streptavidin-coated cyanoacrylate MBs has been studied. In this context, the low amount of MBs in tumors after >5 minutes is beneficial for specific targeting of angiogenesis.


Subject(s)
Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/metabolism , Contrast Media/pharmacokinetics , Cyanoacrylates/chemistry , Microbubbles , Streptavidin/pharmacokinetics , Animals , Coated Materials, Biocompatible/chemistry , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Streptavidin/chemistry , Tissue Distribution , Ultrasonography
7.
Mol Cancer Ther ; 7(1): 101-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18202013

ABSTRACT

Molecular ultrasound is capable of elucidating the expression of angiogenic markers in vivo. However, the capability of the method for volumetric "multitarget quantification" and for the assessment of antiangiogenic therapy response has rather been investigated. Therefore, we generated cyanoacrylate microbubbles linked to vascular endothelial growth factor receptor 2 (VEGFR2) and alphavbeta3 integrin binding ligands and quantified their accumulation in squamous cell carcinoma xenografts (HaCaT-ras-A-5RT3) in mice with the quantitative volumetric ultrasound scanning technique, sensitive particle acoustic quantification. Specificity of VEGFR2 and alphavbeta3 integrin binding microbubbles was shown, and changes in marker expression during matrix metalloproteinase inhibitor treatment were investigated. In tumors, accumulation of targeted microbubbles was significantly higher compared with nonspecific ones and could be inhibited competitively by addition of the free ligand in excess. Also, multimarker imaging could successfully be done during the same imaging session. Molecular ultrasound further indicated a significant increase of VEGFR2 and alphavbeta3 integrin expression during tumor growth and a considerable decrease in both marker densities after matrix metalloproteinase inhibitor treatment. Histologic data suggested that the increasing VEGFR2 and alphavbeta3 integrin concentrations in tumors during growth are related to an up-regulation of its expression by the endothelial cells, whereas its decrease under therapy is more related to the decreasing relative vessel density. In conclusion, targeted ultrasound appears feasible for the longitudinal molecular profiling of tumor angiogenesis and for the sensitive assessment of therapy effects in vivo.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/blood supply , Neoplasms/diagnostic imaging , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/drug therapy , Animals , Cell Line, Tumor , Humans , Immunohistochemistry , Integrin alphaVbeta3/metabolism , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Microtubules/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Ultrasonography , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
8.
Eur Radiol ; 18(4): 753-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18084768

ABSTRACT

The sensitivity of Doppler ultrasound below 10 MHz to assess antiangiogenic therapy effects in tumor xenografts has been shown to be limited. Thus, our aim was to evaluate high-frequency volumetric power-Doppler ultrasound (HF-VPDU) for monitoring antiangiogenic treatments. Squamous cell carcinoma xenografts grown in nude mice were scanned with HF-VPDU at a center frequency of 30 MHz. Images with 200-microm slice thicknesses were recorded and merged into a three-dimensional dataset, of which the relative color pixel density was determined. Animals received either VEGFR2 antibodies or 0.9% NaCl and were examined at days 0, 3 and 6 of treatment. After the last examination, tumors were resected and their vascularization characterized by immunohistology. At day 6, the volumes of treated and untreated tumors were not significantly different yet. In contrast, mean tumor vascularization in treated animals had decreased to 44%, while in the control group it had increased to 152% (P < 0.01). In correspondence, vessel density, as determined by CD31 staining, was 0.19 +/- 0.10% in treated and 0.92 +/- 0.41% in untreated tumors (P < 0.01). Additionally, the fraction of mature (SMA-positive) vessels increased under therapy. Thus, HF-VPDU can be considered as an easily applicable and fast method to screen high animal numbers for antiangiogenic therapy effects.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/diagnostic imaging , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/drug therapy , Ultrasonography, Doppler/methods , Animals , Immunohistochemistry , Mice , Mice, Nude , Sensitivity and Specificity , Statistics, Nonparametric , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
9.
Cancer Res ; 67(4): 1555-62, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17308094

ABSTRACT

Angiogenesis is essential for the development of malignant tumors and provides important targets for tumor diagnosis and therapy. To noninvasively assess the angiogenic profile of tumors, novel alpha(v)beta(3) integrin-targeted ultrasmall superparamagnetic iron oxide particles (USPIOs) were designed and their specific uptake by endothelial cells was evaluated in vitro and in vivo. USPIOs were coated with 3-aminopropyltrimethoxysilane (APTMS) and conjugated with Arg-Gly-Asp (RGD) peptides. Accumulation in human umbilical vein endothelial cells (HUVECs) was evaluated using Prussian blue staining, transmission electron microscopy, magnetic resonance (MR) imaging, and inductively coupled plasma mass spectrometry. Uptake of RGD-USPIO by HUVECs was significantly increased when compared with unlabeled USPIO and could be competitively inhibited by addition of unbound RGD. The ability of the RGD-USPIO to noninvasively distinguish tumors with high (HaCaT-ras-A-5RT3) and lower (A431) area fractions of alpha(v)beta(3) integrin-positive vessels was evaluated using a 1.5-T MR scanner. Indeed, after RGD-USPIO injection, there was a more pronounced decrease in T(2) relaxation times in HaCaT-ras-A-5RT3 tumors than in A431 tumors. Furthermore, T(2)*-weighted images clearly identified the heterogeneous arrangement of vessels with alpha(v)beta(3) integrins in HaCaT-ras-A-5RT3 tumors by an irregular signal intensity decrease. In contrast, in A431 tumors with predominantly small and uniformly distributed vessels, the signal intensity decreased more homogeneously. In summary, RGD-coupled, APTMS-coated USPIOs efficiently label alpha(v)beta(3) integrins expressed on endothelial cells. Furthermore, these molecular MR imaging probes are capable of distinguishing tumors differing in the degree of alpha(v)beta(3) integrin expression and in their angiogenesis profile even when using a clinical 1.5-T MR scanner.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Endothelial Cells/metabolism , Ferric Compounds/pharmacokinetics , Integrin alphaVbeta3/metabolism , Magnetic Resonance Imaging/methods , Neoplasms, Experimental/blood supply , Oligopeptides/pharmacokinetics , Animals , Carcinoma, Squamous Cell/metabolism , Cells, Cultured , Humans , Integrin alphaVbeta3/biosynthesis , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/diagnosis , Neovascularization, Pathologic/metabolism , Particle Size , Propylamines/pharmacokinetics , Silanes/pharmacokinetics
10.
Eur Radiol ; 17(8): 2136-48, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17308924

ABSTRACT

The switch to an angiogenic phenotype is an important precondition for tumor growth, invasion and spread. Since newly formed vessels are characterized by structural, functional and molecular abnormalities, they offer promising targets for tumor diagnosis and therapy. Previous studies indicate that MRI is valuable to assess vessel morphology and function. It can be used to distinguish between benign and malignant lesions and to improve delineation of proliferating areas within heterogeneous tumors. In addition, tracer kinetic analysis of contrast-enhanced image series allows the estimation of well-defined physiological parameters such as blood volume, blood flow and vessel permeability. Frequently, changes of these parameters during cytostatic, anti-angiogenic and radiation therapy precede tumor volume reduction. Moreover, target-specific MRI techniques can be used to elucidate the expression of angiogenic markers at the molecular level. This review summarizes strategies for non-invasive characterization of tumor vascularization by functional and molecular MRI, hereby introducing representative preclinical and clinical applications.


Subject(s)
Magnetic Resonance Angiography/methods , Neoplasms/blood supply , Neovascularization, Pathologic/diagnosis , Animals , Contrast Media , Humans
11.
Neoplasia ; 9(1): 57-67, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17325744

ABSTRACT

In prostate cancers (PCa), the formation of malignant stroma may substantially influence tumor phenotype and aggressiveness. Thus, the impact of the orthotopic and subcutaneous implantations of hormone-sensitive (H), hormone-insensitive (HI), and anaplastic (AT1) Dunning PCa in rats on growth, microcirculation, and metabolism was investigated. For this purpose, dynamic contrast-enhanced magnetic resonance imaging and (1)H magnetic resonance spectroscopy ([(1)H]MRS) were applied in combination with histology. Consistent observations revealed that orthotopic H tumors grew significantly slower compared to subcutaneous ones, whereas the growth of HI and AT1 tumors was comparable at both locations. Histologic analysis indicated that glandular differentiation and a close interaction of tumor cells and smooth muscle cells (SMC) were associated with slow tumor growth. Furthermore, there was a significantly lower SMC density in subcutaneous H tumors than in orthotopic H tumors. Perfusion was observed to be significantly lower in orthotopic H tumors than in subcutaneous H tumors. Regional blood volume and permeability-surface area product showed no significant differences between tumor models and their implantation sites. Differences in growth between subcutaneous and orthotopic H tumors can be attributed to tumor-stroma interaction and perfusion. Here, SMC, may stabilize glandular structures and contribute to the maintenance of differentiated phenotype.


Subject(s)
Prostatic Neoplasms/pathology , Stromal Cells/physiology , Animals , Cell Proliferation , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Myocytes, Smooth Muscle/pathology , Phenotype , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Rats
12.
J Invest Dermatol ; 127(3): 545-54, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17008882

ABSTRACT

Nidogens are considered as classical linkers joining laminin and collagen IV networks in basement membranes (BMs); however, recent genetic approaches have suggested that nidogens function in a tissue-specific and developmental context. Thus, in mice lacking both nidogen-1 and -2 heart and lung were severely affected, causing neonatal death. Furthermore, in various locations, extravasation of erythrocytes was observed implying microvascular defects. Mice expressing solely either isoform, had a functional BM, although nidogen-2 binds with lower affinity to the laminin gamma1 chain. Having previously blocked BM formation by interfering with nidogen-1 binding to laminin in skin-organotypic cocultures, here we investigated the roles of nidogen-1 and -2 in this model. For that purpose, human HaCaT cells were grown in three-dimensional cocultures on collagen matrices containing murine fibroblasts of varying nidogen deficiency. As with our experiments blocking laminin-nidogen interaction, lack of both nidogens completely prevented BM deposition and ultrastructural assembly of BM and hemidesmosomes, although other BM proteins remained detectable at comparable levels with no signs of degradation. Supplementation by recombinant nidogen-1 or -2 restored these structures, as shown by immunofluorescence and electron microscopy, confirming that in this system nidogen-2 is equivalent to nidogen-1, and both can promote the development of a functional BM zone.


Subject(s)
Basement Membrane/metabolism , Cell Adhesion Molecules/physiology , Membrane Glycoproteins/physiology , Skin/immunology , Animals , Calcium-Binding Proteins , Cell Adhesion Molecules/biosynthesis , Coculture Techniques , Collagen/chemistry , Dermis/metabolism , Epidermis/metabolism , Erythrocytes/metabolism , Fibroblasts/metabolism , Humans , Membrane Glycoproteins/biosynthesis , Mice , Organ Culture Techniques , Protein Isoforms
SELECTION OF CITATIONS
SEARCH DETAIL
...