Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Kidney Int ; 97(3): 609-614, 2020 03.
Article in English | MEDLINE | ID: mdl-31784048

ABSTRACT

Pathological deposition of collagen is a hallmark of kidney fibrosis. To illustrate this process we employed multimodal optical imaging to visualize and quantify collagen deposition in murine models of kidney fibrosis (ischemia-reperfusion or unilateral ureteral obstruction) using the collagen-binding adhesion protein CNA35. For in vivo imaging, we used hybrid computed tomography-fluorescence molecular tomography and CNA35 labeled with the near-infrared fluorophore Cy7. Upon intravenous injection, CNA35-Cy7 accumulation was significantly higher in fibrotic compared to non-fibrotic kidneys. This difference was not detected for a non-specific scrambled version of CNA35-Cy7. Ex vivo, on kidney sections of mice and patients with renal fibrosis, CNA35-FITC co-localized with fibrotic collagen type I and III, but not with the basement membrane collagen type IV. Following intravenous injection, CNA35-FITC bound to both interstitial and perivascular fibrotic areas. In line with this perivascular accumulation, we observed significant perivascular fibrosis in the mouse models and in biopsy sections from patients with chronic kidney disease using computer-based morphometry quantification. Thus, molecular imaging of collagen using CNA35 enabled specific non-invasive quantification of kidney fibrosis. Collagen imaging revealed significant perivascular fibrosis as a consistent component next to the more commonly assessed interstitial fibrosis. Our results lay the basis for further probe and protocol optimization towards the clinical translation of molecular imaging of kidney fibrosis.


Subject(s)
Carrier Proteins , Ureteral Obstruction , Animals , Collagen/metabolism , Fibrosis , Humans , Kidney/pathology , Mice , Molecular Imaging , Ureteral Obstruction/pathology
2.
Mol Cancer ; 16(1): 44, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28231808

ABSTRACT

BACKGROUND: Extracellular matrix (ECM) is known to maintain epithelial integrity. In carcinogenesis ECM degradation triggers metastasis by controlling migration and differentiation including cancer stem cell (CSC) characteristics. The ECM-modulator inter- α-trypsin inhibitor heavy chain family member five (ITIH5) was recently identified as tumor suppressor potentially involved in impairing breast cancer progression but molecular mechanisms underlying its function are still elusive. METHODS: ITIH5 expression was analyzed using the public TCGA portal. ITIH5-overexpressing single-cell clones were established based on T47D and MDA-MB-231 cell lines. Colony formation, growth, apoptosis, migration, matrix adhesion, traction force analyses and polarization of tumor cells were studied in vitro. Tumor-initiating characteristics were analyzed by generating a metastasis mouse model. To identify ITIH5-affected pathways we utilized genome wide gene expression and DNA methylation profiles. RNA-interference targeting the ITIH5-downstream regulated gene DAPK1 was used to confirm functional involvement. RESULTS: ITIH5 loss was pronounced in breast cancer subtypes with unfavorable prognosis like basal-type tumors. Functionally, cell and colony formation was impaired after ITIH5 re-expression in both cell lines. In a metastasis mouse model, ITIH5 expressing MDA-MB-231 cells almost completely failed to initiate lung metastases. In these metastatic cells ITIH5 modulated cell-matrix adhesion dynamics and altered biomechanical cues. The profile of integrin receptors was shifted towards ß1-integrin accompanied by decreased Rac1 and increased RhoA activity in ITIH5-expressing clones while cell polarization and single-cell migration was impaired. Instead ITIH5 expression triggered the formation of epithelial-like cell clusters that underwent an epigenetic reprogramming. 214 promoter regions potentially marked with either H3K4 and /or H3K27 methylation showed a hyper- or hypomethylated DNA configuration due to ITIH5 expression finally leading to re-expression of the tumor suppressor DAPK1. In turn, RNAi-mediated knockdown of DAPK1 in ITIH5-expressing MDA-MB-231 single-cell clones clearly restored cell motility. CONCLUSIONS: Our results provide evidence that ITIH5 triggers a reprogramming of breast cancer cells with known stem CSC properties towards an epithelial-like phenotype through global epigenetic changes effecting known tumor suppressor genes like DAPK1. Therewith, ITIH5 may represent an ECM modulator in epithelial breast tissue mediating suppression of tumor initiating cancer cell characteristics which are thought being responsible for the metastasis of breast cancer.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Death-Associated Protein Kinases/genetics , Lung Neoplasms/secondary , Proteinase Inhibitory Proteins, Secretory/genetics , Animals , Cell Line, Tumor , Epigenesis, Genetic , Extracellular Matrix , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Mice , Neoplasm Transplantation , Prognosis , Survival Analysis
3.
Hepatology ; 63(4): 1310-24, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26699087

ABSTRACT

UNLABELLED: Pathogen- and injury-related danger signals as well as cytokines released by immune cells influence the functional differentiation of macrophages in chronic inflammation. Recently, the liver-derived plasma protein, histidine-rich glycoprotein (HRG), was demonstrated, in mouse tumor models, to mediate the transition of alternatively activated (M2) to proinflammatory (M1) macrophages, which limit tumor growth and metastasis. We hypothesized that liver-derived HRG is a critical endogenous modulator of hepatic macrophage functionality and investigated its implications for liver inflammation and fibrosis by comparing C57BL/6N wild-type (WT) and Hrg(-/-) mice. In homeostatic conditions, hepatic macrophages were overall reduced and preferentially polarized toward the anti-inflammatory M2 subtype in Hrg(-/-) mice. Upon chronic liver damage induced by CCl4 or methionine-choline-deficient (MCD) diet, liver injury and fibrosis were attenuated in Hrg(-/-) , compared to WT, mice. Macrophage populations were reduced and skewed toward M2 polarization in injured livers of Hrg(-/-) mice. Moreover, HRG-deficient mice showed significantly enhanced hepatic vascularization by micro-computed tomography and histology, corroborating proangiogenic activities of M2-polarized liver macrophages. Purified HRG protein induced, but HRG-deficient serum prevented, M1 macrophage differentiation in vitro. Accordingly, Hrg(-/-) mice transplanted with Hrg(+/+) bone marrow, but not Hrg(-/-) -transplanted Hrg(+/+) mice, remained protected from experimental steatohepatitis. Consistent with these findings, patients with chronic hepatitis C and nonalcoholic steatohepatitis significantly up-regulated hepatocytic HRG expression, which was associated with M1 polarization of adjacent macrophages. CONCLUSIONS: Liver-derived HRG, similar to alarmins, appears to be an endogenous molecular factor promoting polarization of hepatic macrophages toward the M1 phenotype, thereby promoting chronic liver injury and fibrosis progression, but limiting angiogenesis. Therefore, controlling tissue levels of HRG or PGF might be a promising strategy in chronic inflammatory liver diseases.


Subject(s)
Fatty Liver/pathology , Hepatitis C/pathology , Liver Cirrhosis/pathology , Macrophage Activation , Proteins/metabolism , Animals , Biomarkers/metabolism , Biopsy, Needle , Cells, Cultured , Disease Models, Animal , Fatty Liver/physiopathology , Hepatitis C/physiopathology , Humans , Immunohistochemistry , Liver Cirrhosis/physiopathology , Mice , Mice, Inbred C57BL , Prognosis , Random Allocation , Risk Assessment
4.
J Am Soc Nephrol ; 27(2): 520-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26195818

ABSTRACT

Progressive kidney diseases and renal fibrosis are associated with endothelial injury and capillary rarefaction. However, our understanding of these processes has been hampered by the lack of tools enabling the quantitative and noninvasive monitoring of vessel functionality. Here, we used micro-computed tomography (µCT) for anatomical and functional imaging of vascular alterations in three murine models with distinct mechanisms of progressive kidney injury: ischemia-reperfusion (I/R, days 1-56), unilateral ureteral obstruction (UUO, days 1-10), and Alport mice (6-8 weeks old). Contrast-enhanced in vivo µCT enabled robust, noninvasive, and longitudinal monitoring of vessel functionality and revealed a progressive decline of the renal relative blood volume in all models. This reduction ranged from -20% in early disease stages to -61% in late disease stages and preceded fibrosis. Upon Microfil perfusion, high-resolution ex vivo µCT allowed quantitative analyses of three-dimensional vascular networks in all three models. These analyses revealed significant and previously unrecognized alterations of preglomerular arteries: a reduction in vessel diameter, a prominent reduction in vessel branching, and increased vessel tortuosity. In summary, using µCT methodology, we revealed insights into macro-to-microvascular alterations in progressive renal disease and provide a platform that may serve as the basis to evaluate vascular therapeutics in renal disease.


Subject(s)
Blood Vessels/physiopathology , Kidney Diseases/diagnostic imaging , Kidney Diseases/physiopathology , Kidney/blood supply , X-Ray Microtomography , Animals , Disease Progression , Mice
5.
Angiogenesis ; 19(4): 525-35, 2016 10.
Article in English | MEDLINE | ID: mdl-27582011

ABSTRACT

Renal microvascular rarefaction characterizes chronic kidney disease (CKD). In murine models of CKD, micro-CT imaging reflected capillary rarefaction using quantification of renal relative blood volume (rBV). In addition, micro-CT imaging revealed morphological alterations of the intrarenal vasculature including reduced vascular branching and lumen diameter. Here, we retrospectively quantified rBV in contrast-enhanced CT angiography in patients and found that, compared to non-CKD patients, those with CKD and renal fibrosis had significantly reduced rBV in the renal cortex. rBV values closely mirrored capillary rarefaction in the corresponding nephrectomy specimens. In patients with follow-up CT angiography, reduction of renal function was paralleled by a decline in rBV. Using virtual autopsy, i.e., postmortem CT angiography, morphometry of intrarenal arteries in 3D-rendered CT images revealed significantly reduced arterial diameter and branching in CKD compared to non-CKD cases. In conclusion, in CKD patients, contrast-enhanced CT imaging with quantification of rBV correlates with functional renal vasculature, whereas virtual autopsy allows morphometric analyses of macrovascular changes. Importantly, the observed vascular alterations in CKD patients mirror those in animals with progressive CKD, suggesting a high relevance of animal models for studying vascular alterations in CKD and renal fibrosis.


Subject(s)
Computed Tomography Angiography/methods , Renal Insufficiency, Chronic/diagnostic imaging , Aged , Animals , Blood Volume , Capillaries/diagnostic imaging , Capillaries/pathology , Cohort Studies , Contrast Media , Disease Progression , Fibrosis , Humans , Imaging, Three-Dimensional , Kidney/blood supply , Kidney/diagnostic imaging , Kidney/pathology , Male , Microvessels/diagnostic imaging , Microvessels/pathology , Middle Aged , Renal Circulation , Renal Insufficiency, Chronic/pathology , Retrospective Studies
6.
Angiogenesis ; 19(2): 245-254, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26902100

ABSTRACT

Vascular endothelial growth factor receptor 2 (VEGFR-2) and α v ß 3 integrin are the most frequently addressed targets in molecular imaging of tumor angiogenesis. In preclinical studies, molecular imaging of angiogenesis has shown potential to detect and differentiate benign and malignant lesions of the breast. Thus, in this retrospective clinical study employing patient tissues, the diagnostic value of VEGFR-2, α v ß 3 integrin and vascular area fraction for the diagnosis and differentiation of breast neoplasia was evaluated. To this end, tissue sections of breast cancer (n = 40), pre-invasive ductal carcinoma in situ (DCIS; n = 8), fibroadenoma (n = 40), radial scar (n = 6) and normal breast tissue (n = 40) were used to quantify (1) endothelial VEGFR-2, (2) endothelial α v ß 3 integrin and (3) total α v ß 3 integrin expression, as well as (4) the vascular area fraction. Sensitivity and specificity to differentiate benign from malignant lesions were calculated for each marker by receiver operating characteristics (ROC) analyses. Whereas vessel density, as commonly used, did not significantly differ between benign and malignant lesions (AUROC: 0.54), VEGFR-2 and α v ß 3 integrin levels were gradually up-regulated in carcinoma versus fibroadenoma versus healthy tissue. The highest diagnostic accuracy for differentiating carcinoma from fibroadenoma was found for total α v ß 3 integrin expression (AUROC: 0.76), followed by VEGFR-2 (AUROC: 0.71) and endothelial α v ß 3 integrin expression (AUROC: 0.68). In conclusion, total α v ß 3 integrin expression is the best discriminator between breast cancer, fibroadenoma and normal breast tissue. With respect to vascular targeting and molecular imaging of angiogenesis, endothelial VEGFR-2 appeared to be slightly superior to endothelial α v ß 3 for differentiating benign from cancerous lesions.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Integrin alphaVbeta3/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Endothelial Cells/metabolism , Female , Humans , Middle Aged , Reproducibility of Results
7.
Am J Pathol ; 185(8): 2132-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26216283

ABSTRACT

We have identified platelet-derived growth factor (PDGF)-CC as a potent profibrotic mediator in kidney fibrosis and pro-angiogenic mediator in glomeruli. Because renal fibrosis is associated with progressive capillary rarefaction, we asked whether PDGF-CC neutralization in fibrosis might have detrimental anti-angiogenic effects leading to aggravated peritubular capillary loss. We analyzed capillary rarefaction in mice with and without PDGF-CC neutralization (using genetically deficient mice and neutralizing antibodies), in three different models of renal interstitial fibrosis, unilateral ureteral obstruction, unilateral ischemia-reperfusion, Col4a3-deficient (Alport) mice, and healthy animals. Independent of the effect of PDGF-CC neutralization on renal fibrosis, we found no difference in capillary rarefaction between PDGF-CC-neutralized mice and mice with intact PDGF-CC. We also found no differences in microvascular leakage (determined by extravasation of Evans Blue Dye) and in renal relative blood volume quantified using in vivo microcomputed tomography. PDGF-CC neutralization had no effects on renal microvasculature in healthy animals. Capillary endothelium did not express PDGF receptor-α, suggesting that potential PDGF-CC effects would have to be indirect. PDGF-CC neutralization or deficiency was not associated with preservation or accelerated loss of peritubular capillaries, suggesting no significant pro-angiogenic effects of PDGF-CC during renal fibrosis. From a clinical perspective, the profibrotic effects of PDGF-CC outweigh the pro-angiogenic effects and, thus, do not limit a potential therapeutic use of PDGF-CC inhibition in renal fibrosis.


Subject(s)
Capillaries/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Capillaries/pathology , Disease Models, Animal , Fibrosis/metabolism , Fibrosis/pathology , Kidney/pathology , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lymphokines/genetics , Mice , Mice, Knockout , Platelet-Derived Growth Factor/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
8.
Adv Funct Mater ; 25(1): 36-43, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25729344

ABSTRACT

Efficient and safe drug delivery across the blood-brain barrier (BBB) remains to be one of the major challenges of biomedical and (nano-) pharmaceutical research. Here, we show that poly(butyl cyanoacrylate)-based microbubbles (MB), carrying ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles within their shell, can be used to mediate and monitor BBB permeation. Upon exposure to transcranial ultrasound pulses, USPIO-MB are destroyed, resulting in acoustic forces inducing vessel permeability. At the same time, USPIO are released from the MB shell, they extravasate across the permeabilized BBB and they accumulate in extravascular brain tissue, thereby providing non-invasive R2*-based magnetic resonance imaging information on the extent of BBB opening. Quantitative changes in R2* relaxometry were in good agreement with 2D and 3D microscopy results on the extravascular deposition of the macromolecular model drug FITC-dextran into the brain. Such theranostic materials and methods are considered to be useful for mediating and monitoring drug delivery across the BBB, and for enabling safe and efficient treatment of CNS disorders.

9.
Am J Pathol ; 184(2): 431-41, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24262753

ABSTRACT

Angiogenesis is a hallmark of cancer, and its noninvasive visualization and quantification are key factors for facilitating translational anticancer research. Using four tumor models characterized by different degrees of aggressiveness and angiogenesis, we show that the combination of functional in vivo and anatomical ex vivo X-ray micro-computed tomography (µCT) allows highly accurate quantification of relative blood volume (rBV) and highly detailed three-dimensional analysis of the vascular network in tumors. Depending on the tumor model, rBV values determined using in vivo µCT ranged from 2.6% to 6.0%, and corresponds well with the values assessed using IHC. Using ultra-high-resolution ex vivo µCT, blood vessels as small as 3.4 µm and vessel branches up to the seventh order could be visualized, enabling a highly detailed and quantitative analysis of the three-dimensional micromorphology of tumor vessels. Microvascular parameters such as vessel size and vessel branching correlated very well with tumor aggressiveness and angiogenesis. In rapidly growing and highly angiogenic A431 tumors, the majority of vessels were small and branched only once or twice, whereas in slowly growing A549 tumors, the vessels were much larger and branched four to seven times. Thus, we consider that combining highly accurate functional with highly detailed anatomical µCT is a useful tool for facilitating high-throughput, quantitative, and translational (anti-) angiogenesis and antiangiogenesis research.


Subject(s)
Neoplasms/blood supply , Neoplasms/diagnostic imaging , Neovascularization, Pathologic/diagnostic imaging , X-Ray Microtomography , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Immunohistochemistry , Mice , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Xenograft Model Antitumor Assays
10.
J Theor Biol ; 365: 289-300, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25451523

ABSTRACT

Physiologically realistic geometric models of the vasculature in the liver are indispensable for modelling hepatic blood flow, the main connection between the liver and the organism. Current in vivo imaging techniques do not provide sufficiently detailed vascular trees for many simulation applications, so it is necessary to use algorithmic refinement methods. The method of Constrained Constructive Optimization (CCO) (Schreiner et al., 2006) is well suited for this purpose. Its results after calibration have been previously compared to experimentally acquired human vascular trees (Schwen and Preusser, 2012). The goal of this paper is to extend this calibration to the case of rodents (mice and rats), the most commonly used animal models in liver research. Based on in vivo and ex vivo micro-CT scans of rodent livers and their vasculature, we performed an analysis of various geometric features of the vascular trees. Starting from pruned versions of the original vascular trees, we applied the CCO procedure and compared these algorithmic results to the original vascular trees using a suitable similarity measure. The calibration of the postprocessing improved the algorithmic results compared to those obtained using standard CCO. In terms of angular features, the average similarity increased from 0.27 to 0.61, improving the total similarity from 0.28 to 0.40. Finally, we applied the calibrated algorithm to refine measured vascular trees to the (higher) level of detail desired for specific applications. Having successfully adapted the CCO algorithm to the rodent model organism, the resulting individual-specific refined hepatic vascular trees can now be used for advanced modeling involving, e.g., detailed blood flow simulations.


Subject(s)
Algorithms , Liver/blood supply , Animals , Calibration , Humans , Imaging, Three-Dimensional , Mice , Models, Biological , Rats
11.
Nano Lett ; 14(2): 972-81, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24422585

ABSTRACT

Enhanced permeability and retention (EPR) and the (over-) expression of angiogenesis-related surface receptors are key features of tumor blood vessels. As a consequence, EPR-mediated passive and Arg-Gly-Asp (RGD) and Asn-Gly-Arg (NGR) based active tumor targeting have received considerable attention in the last couple of years. Using several different in vivo and ex vivo optical imaging techniques, we here visualized and quantified the benefit of RGD- and NGR-based vascular vs EPR-mediated passive tumor targeting. This was done using ∼ 10 nm sized polymeric nanocarriers, which were either labeled with DY-676 (peptide-modified polymers) or with DY-750 (peptide-free polymers). Upon coinjection into mice bearing both highly leaky CT26 and poorly leaky BxPC3 tumors, it was found that vascular targeting did work, resulting in rapid and efficient early binding to tumor blood vessels, but that over time, passive targeting was significantly more efficient, leading to higher overall levels and to more efficient retention within tumors. Although this situation might be different for larger carrier materials, these insights indicate that caution should be taken not to overestimate the potential of active over passive tumor targeting.


Subject(s)
Antineoplastic Agents/administration & dosage , Nanocapsules/chemistry , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Oligopeptides/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Diffusion , Humans , Mice , Nanocapsules/administration & dosage , Nanocapsules/ultrastructure , Nanocomposites/chemistry , Nanocomposites/ultrastructure , Neoplasms, Experimental/pathology , Oligopeptides/chemistry , Particle Size
12.
Gut ; 63(12): 1960-1971, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24561613

ABSTRACT

OBJECTIVES: In chronic liver injury, angiogenesis, the formation of new blood vessels from pre-existing ones, may contribute to progressive hepatic fibrosis and to development of hepatocellular carcinoma. Although hypoxia-induced expression of vascular endothelial growth factor (VEGF) occurs in advanced fibrosis, we hypothesised that inflammation may endorse hepatic angiogenesis already at early stages of fibrosis. DESIGN: Angiogenesis in livers of c57BL/6 mice upon carbon tetrachloride- or bile duct ligation-induced chronic hepatic injury was non-invasively monitored using in vivo contrast-enhanced micro computed tomography (µCT) and ex vivo anatomical µCT after hepatic Microfil perfusion. Functional contributions of monocyte-derived macrophage subsets for angiogenesis were explored by pharmacological inhibition of CCL2 using the Spiegelmer mNOX-E36. RESULTS: Contrast-enhanced in vivo µCT imaging allowed non-invasive monitoring of the close correlation of angiogenesis, reflected by functional hepatic blood vessel expansion, with experimental fibrosis progression. On a cellular level, inflammatory monocyte-derived macrophages massively accumulated in injured livers, colocalised with newly formed vessels in portal tracts and exhibited pro-angiogenic gene profiles including upregulated VEGF and MMP9. Functional in vivo and anatomical ex vivo µCT analyses demonstrated that inhibition of monocyte infiltration by targeting the chemokine CCL2 prevented fibrosis-associated angiogenesis, but not fibrosis progression. Monocyte-derived macrophages primarily fostered sprouting angiogenesis within the portal vein tract. Portal vein diameter as a measure of portal hypertension depended on fibrosis, but not on angiogenesis. CONCLUSIONS: Inflammation-associated angiogenesis is promoted by CCL2-dependent monocytes during fibrosis progression. Innovative in vivo µCT methodology can accurately monitor angiogenesis and antiangiogenic therapy effects in experimental liver fibrosis.


Subject(s)
Aptamers, Nucleotide/pharmacology , Chemokine CCL2 , Liver Cirrhosis , Macrophages , Neovascularization, Pathologic , Animals , Carbon Tetrachloride/pharmacology , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/metabolism , Disease Models, Animal , Disease Progression , Inflammation/metabolism , Liver/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , X-Ray Microtomography/methods
13.
Adv Funct Mater ; 24(6): 754-762, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24569840

ABSTRACT

Non-invasive imaging holds significant potential for implementation in tissue engineering. It can e.g. be used to monitor the localization and function of tissue-engineered implants, as well as their resorption and remodelling. Thus far, however, the vast majority of efforts in this area of research have focused on the use of ultrasmall super-paramagnetic iron oxide (USPIO) nanoparticle-labeled cells, colonizing the scaffolds, to indirectly image the implant material. Reasoning that directly labeling scaffold materials might be more beneficial (enabling imaging also in case of non-cellularized implants), more informative (enabling the non-invasive visualization and quantification of scaffold degradation) and more easy to translate into the clinic (since cell-free materials are less complex from a regulatory point-of-view), we here prepared three different types of USPIO nanoparticles, and incorporated them both passively and actively (via chemical conjugation; during collagen crosslinking) into collagen-based scaffold materials. We furthermore optimized the amount of USPIO incorporated into the scaffolds, correlated the amount of entrapped USPIO with MR signal intensity, showed that the labeled scaffolds are highly biocompatible, demonstrated that scaffold degradation can be visualized using MRI and provided initial proof-of-principle for the in vivo visualization of the scaffolds. Consequently, USPIO-labeled scaffold materials seem to be highly suitable for image-guided tissue engineering applications.

14.
Rofo ; 2024 Apr 17.
Article in English, German | MEDLINE | ID: mdl-38631371

ABSTRACT

The aim of our work was to demonstrate the importance of artificial intelligence-based analysis of fractional flow reserves of computed tomographically detected coronary artery stenosis with regard to their hemodynamic relevance in patients with unclear chest pain and suspected stable coronary heart disease with a low to medium pre-test probability.The collective of our retrospective analysis includes 63 patients in whom coronary artery stenosis was detected by volume computed tomographic examination in "one beat, whole heart" mode in the period from March to October 2022. In these patients, the fractional flow reserve was also determined by computed tomography, which was modulated by the use of artificial intelligence.The calculated values of the fractional flow reserve and the degrees of stenosis determined by computed tomography showed a moderate and significant negative correlation for all three coronary vascular territories (LAD/CX/RCA) (correlation coefficient rho = 0.54/0.54/0.6; p < 0.01 respectively). In just over a third (37.6 %) of all stenoses classified as high-grade by computed tomography, the assessment of hemodynamic relevance by calculating the fractional flow reserve deviated from the severity of the stenosis diagnosed by computed tomography, while the results in the peripheral areas "no stenosis/vascular occlusion" were 100 % consistent in each case.The present results of this work illustrate that the calculation of the fractional flow reserve based on artificial intelligence as a supplement to volume computed tomography of the heart can make a decisive contribution to further therapy planning by increasing the specificity of the purely morphological method by the physiological aspect. · Calculation of fractional flow reserve is a useful addition to computed tomography of the heart.. · It provides possibility to dispense with unnecessary further diagnostics by increasing specificity.. · The combination of both procedures leads to therapy optimization for patients.. · Noblé H, Mühlbauer N, Ehling J et al. The value of AI-based analysis of fractional flow reserve of volume computed tomographically detected coronary artery stenosis with regard to their hemodynamic relevance. Fortschr Röntgenstr 2024; DOI: 10.1055/a-2271-0887.

15.
J Control Release ; 365: 358-368, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38016488

ABSTRACT

Blood vessel functionality is crucial for efficient tumor-targeted drug delivery. Heterogeneous distribution and perfusion of angiogenic blood vessels contribute to suboptimal accumulation of (nano-) therapeutics in tumors and metastases. To attenuate pathological angiogenesis, an L-RNA aptamer inhibiting the CC motif chemokine ligand 2 (CCL2) was administered to mice bearing orthotopic 4T1 triple-negative breast cancer tumors. The effect of CCL2 inhibition on tumor blood vessel functionality and tumor-targeted drug delivery was evaluated via multimodal and multiscale optical imaging, employing fluorophore-labeled polymeric (10 nm) and liposomal (100 nm) nanocarriers. Anti-CCL2 treatment induced a dose-dependent anti-angiogenic effect, reflected by a decreased relative blood volume, increased blood vessel maturity and functionality, and reduced macrophage infiltration, accompanied by a shift in the polarization of tumor-associated macrophages (TAM) towards a less M2-like and more M1-like phenotype. In line with this, CCL2 inhibitor treatment improved the delivery of polymers and liposomes to tumors, and enhanced the antitumor efficacy of free and liposomal doxorubicin. Together, these findings demonstrate that blocking the CCL2-CCR2 axis modulates TAM infiltration and polarization, resulting in vascular normalization and improved tumor-targeted drug delivery.


Subject(s)
Chemokine CCL2 , Neoplasms , Mice , Animals , Chemokine CCL2/pharmacology , Ligands , Nanomedicine , Neoplasms/pathology , Macrophages , Cell Line, Tumor
16.
Eur Radiol ; 23(2): 468-75, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22878592

ABSTRACT

OBJECTIVES: Tumour xenografts of well-discernible sizes can be examined well by molecular ultrasound. Here, we investigated whether very early breast carcinomas express sufficient levels of VEGFR2 for reliable molecular ultrasound imaging with targeted microbubbles. METHODS: MCF-7 breast cancer xenografts were orthotopically implanted in nude mice (n = 26). Tumours measuring from 4 mm(3) (2 mm diameter) up to 65 mm(3) (5 mm diameter) were examined with automated 3D molecular ultrasound using clinically translatable VEGFR2-targeted microbubbles (BR55). Additionally, the relative tumour blood volume was assessed with non-targeted microbubbles (BR38). In vivo ultrasound data were validated by quantitative immunohistochemistry. RESULTS: Very small lesions 2 mm in diameter showed the highest binding of VEGFR2-specific microbubbles. In larger tumours significantly less BR55 accumulated (p = 0.023). Nonetheless, binding of VEGFR2-targeted microbubbles was still high enough for imaging. The relative blood volume was comparable at all tumour sizes. Both findings were confirmed by immunohistochemistry. Additionally, a significantly enhanced number of large and mature vessels were detected with increasing tumour size (p < 0.01), explaining the decrease in VEGFR2 expression during tumour growth. CONCLUSIONS: 3D molecular ultrasound using BR55 is very well suited to depicting the angiogenic activity in very small breast lesions, suggesting its potential for detecting and characterising these lesions.


Subject(s)
Early Detection of Cancer/methods , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/diagnostic imaging , Microbubbles , Neovascularization, Pathologic/diagnostic imaging , Ultrasonography, Doppler, Color/methods , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Contrast Media , Disease Models, Animal , Female , Mammary Glands, Animal/blood supply , Mammary Neoplasms, Experimental/pathology , Mice , Molecular Biology , Neovascularization, Pathologic/metabolism , Predictive Value of Tests , Random Allocation , Sensitivity and Specificity , Transplantation, Heterologous
18.
Cancers (Basel) ; 14(3)2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35158755

ABSTRACT

Oncogenic drivers such as mutated EGFR are the preferred targets in modern drug development. However, restoring the lost function of tumor suppressor proteins could also be a valid approach to combatting cancer. ITIH5 has been revealed as a potent metastasis suppressor in both breast and pancreatic cancer. Here, we show that ITIH5 overexpression in MDA-MB-231 breast cancer cells can also locally suppress tumor growth by 85%, when transplanted into the mammary fat pad of nude mice. For a potential drug development approach, we further aimed to define downsized ITIH5 polypeptides that still are capable of mediating growth inhibitory effects. By cloning truncated and His-tagged ITIH5 fragments, we synthesized two recombinant N-terminal polypeptides (ITIH5681aa and ITIH5161aa), both covering the ITI heavy chain specific "vault protein inter-alpha-trypsin" (VIT) domain. Truncated ITIH5 variants caused dose-dependent cell growth inhibition by up to 50% when applied to various cancer cell lines (e.g., MDA-MB-231, SCaBER, A549) reflecting breast, bladder and lung cancer in vitro. Thus, our data suggest the substantial role of the ITIH5-specific VIT domain in ITIH5-mediated suppression of tumor cell proliferation. As extracellularly administered ITIH5 peptides mimic the growth-inhibitory effects of the full-length ITIH5 tumor suppressor protein, they may constitute the basis for developing anticancer drugs in the future.

19.
Biochim Biophys Acta ; 1801(9): 1025-35, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20470896

ABSTRACT

In the pathogenesis of nonalcoholic fatty liver disease, accumulation of lipids in hepatocytes and hepatocyte apoptosis are strongly implicated in disease progression from the potentially reversible condition of steatosis to severe acute and chronic liver injury. Acyl-CoA synthetase 5, a member of the ACSL gene family that catalyzes the activation of long-chain fatty acids for lipid biosynthesis, is the only ACSL isoform that is both, located on mitochondria and functionally involved in enterocyte apoptosis. In this study, the regulation of human ACSL5 in hepatocellular fatty acid degeneration and its involvement in hepatocyte apoptosis was investigated using models of in vitro and in vivo steatosis as well as plasmid-mediated stable gene transfer and RNAi-mediated gene silencing. ACSL5 mRNA and protein were strongly increased by uptake of dietary derived fatty acids in primary human hepatocytes, HepG2 cells and human steatotic liver. Over-expression of ACSL5 decreased HepG2 cell viability and increased susceptibility to TRAIL- and TNFalpha-, but not FAS- induced apoptosis, whereas knock down of ACSL5 reduced apoptosis susceptibility. High ACSL5 activity resulted in enhanced caspase-3/7 activity, but was not accompanied by up-regulation of death receptors, DR4, DR5 or TNF-R1. This study gives evidence that hepatocyte steatosis is associated with ACSL5 up-regulation resulting in increased susceptibility to hepatic cell death. We propose that ACSL5 could play a role in promoting fatty acid-induced lipoapoptosis in hepatocytes as important mechanism in fatty liver-related disorders.


Subject(s)
Apoptosis , Coenzyme A Ligases/metabolism , Fatty Liver/pathology , Hepatocytes/pathology , Lipids/pharmacology , Liver/pathology , Blotting, Western , Cell Proliferation , Coenzyme A Ligases/antagonists & inhibitors , Coenzyme A Ligases/genetics , Enzyme-Linked Immunosorbent Assay , Fatty Liver/enzymology , Fatty Liver/genetics , Fluorescent Antibody Technique , Hep G2 Cells , Hepatocytes/enzymology , Humans , Immunoenzyme Techniques , Liver/enzymology , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL