Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cancers (Basel) ; 12(11)2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33153037

ABSTRACT

Inter-case variations in immune cell and fibroblast composition are associated with prognosis in solid tumors, including colon cancer. A series of experimental studies suggest immune-modulatory roles of marker-defined fibroblast populations, including FAP-positive fibroblasts. These studies imply that the fibroblast status of tumors might affect the prognostic significance of immune-related features. Analyses of a population-based colon cancer cohort demonstrated good prognosis associations of FAP intensity and CD8a density. Notably, a significant prognostic interaction was detected between these markers (p = 0.013 in nonadjusted analyses and p = 0.003 in analyses adjusted for cofounding factors) in a manner where the good prognosis association of CD8 density was restricted to the FAP intensity-high group. This prognostic interaction was also detected in an independent randomized trial-derived colon cancer cohort (p = 0.048 in nonadjusted analyses). In the CD8-high group, FAP intensity was significantly associated with a higher total tumor density of FoxP3-positive immune cells and a higher ratio of epithelial-to-stromal density of CD8a T cells. The study presents findings relevant for the ongoing efforts to improve the prognostic performance of CD8-related markers and should be followed by additional validation studies. Furthermore, findings support, in general, earlier model-derived studies implying fibroblast subsets as clinically relevant modulators of immune surveillance. Finally, the associations between FAP intensity and specific immune features suggest mechanisms of fibroblast-immune crosstalk with therapeutic potential.

2.
Pancreas ; 45(8): 1167-74, 2016 09.
Article in English | MEDLINE | ID: mdl-26825865

ABSTRACT

OBJECTIVES: Comprehensive assessment of cMET and HER family receptor tyrosine kinases expression, changes of expression during metastatic progression, amplification status of the MET gene, and correlations with patient characteristics in pancreatic ductal adenocarcinoma (PDAC) was conducted. METHODS: We investigated 56 PDACs and corresponding lymph node metastases for HER1 to HER4 and cMET expression by immunohistochemistry, as well as cMET gene copy numbers by chromogenic in situ hybridization. RESULTS: Of all receptor tyrosine kinases evaluated, cMET expression was highest with 46.5% of tumors showing moderate or strong expression and a weak correlation with gene copy number status (P = 0.04; Spearman ρ = 0.28). cMET expression was increased in metastases. In contrast, expression levels of HER family receptors were generally low both in primaries and metastases. A weak yet significant correlation of HER1 and cMET expression levels was observed (P < 0.001; Spearman ρ = 0.44) and HER1 was often present in poorly differentiated tumors (G3, P = 0.049). CONCLUSIONS: Our data suggest that cMET might constitute an interesting molecule for combining targeted and chemotherapeutic approaches in PDAC, because expression is frequent and increased during metastatic progression. In PDAC, cMET protein expression might be a more useful stratification biomarker than cMET gene amplification, which does not seem to be its primary regulator.


Subject(s)
Carcinoma, Pancreatic Ductal , ErbB Receptors , Female , Humans , Lymph Nodes , Lymphatic Metastasis , Proto-Oncogene Proteins c-met
3.
Cancer Chemother Pharmacol ; 75(4): 837-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25702049

ABSTRACT

PURPOSE: RG7116 is a novel anti-HER3 therapeutic antibody that inhibits HER3 signalling and induces antibody-dependent cellular cytotoxicity of tumor cells due to a glycoengineered antibody Fc moiety. We investigated the efficacy and pharmacokinetic/pharmacodynamic properties of HER3 signal inhibition by RG7116 in a murine xenograft model of human head and neck cancer. METHODS: SCID-beige mice bearing FaDu cells were treated with RG7116 at a weekly dose of 0.3-10 mg/kg, and tumor growth control and modulation of selected proteins (HER3 and AKT) were examined. RESULTS: Complete tumor stasis up to Day 46 was observed at a dose >3 mg/kg, and this dose down-modulated membrane HER3 expression and inhibited HER3 and AKT phosphorylation. Systemic RG7116 exposure was greater than dose-proportional and total clearance declined with increasing dose, indicating that RG7116 elimination is target-mediated. This is consistent with the better efficacy, and the HER3 and pAKT inhibition, that was observed at doses >1 mg/kg. Tumor regrowth occurred from Day 46 onwards and was associated with HER1 and HER2 upregulation, indicating the activation of alternative HER escape pathways. Modulation of HER3 and phospho-HER3 was also demonstrated in the skin and mucosa of an RG7116-treated cynomolgus monkey, suggesting that these may be useful surrogate tissues for monitoring RG7116 activity. CONCLUSIONS: These data confirm the promising efficacy of RG7116 and highlight the value of assessing the PK behavior of the antibody and measuring target protein modulation as a marker of biological activity. Clinical development of RG7116 has now begun, and phase I trials are ongoing.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents , Glycoproteins , Hypopharyngeal Neoplasms/drug therapy , Receptor, ErbB-3/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Dose-Response Relationship, Drug , Glycoproteins/pharmacokinetics , Glycoproteins/pharmacology , Glycoproteins/therapeutic use , Humans , Hypopharyngeal Neoplasms/metabolism , Hypopharyngeal Neoplasms/pathology , Macaca fascicularis , Mice, SCID , Mouth Mucosa/drug effects , Mouth Mucosa/metabolism , Skin/drug effects , Skin/metabolism , Xenograft Model Antitumor Assays
4.
MAbs ; 6(4): 1051-8, 2014.
Article in English | MEDLINE | ID: mdl-24870719

ABSTRACT

The humanized monoclonal antibody with high affinity for the human epidermal growth factor receptor (HER) 3, RG7116, is a glycoengineered, IgG1 class antibody. By labeling RG7116 with zirconium-89 ((89)Zr) we aimed to visualize in vivo HER3 expression and study the biodistribution of this antibody in human tumor-bearing mice. Biodistribution of (89)Zr-RG7116 was studied in subcutaneously xenografted FaDu tumor cells (HER3-positive). Dose-dependency of (89)Zr-RG7116 organ distribution and specific tumor uptake was assessed by administering doses ranging from 0.05 to 10 mg/kg RG7116 to SCID/Beige mice. Biodistribution was analyzed at 24 and 144 h after injection. MicroPET imaging was performed at 1, 3, and 6 days after injection of 1.0 mg/kg (89)Zr-RG7116 in the FaDu, H441, QG-56 and Calu-1 xenografts with varying HER3 expression. The excised tumors were analyzed for HER3 expression. Biodistribution analyses showed a dose- and time-dependent (89)Zr-RG7116 tumor uptake in FaDu tumors. The highest tumor uptake of (89)Zr-RG7116 was observed in the 0.05 mg/kg dose group with 27.5%ID/g at 144 h after tracer injection. MicroPET imaging revealed specific tumor uptake of (89)Zr-RG7116 in FaDu and H441 models with an increase in tumor uptake over time. Biodistribution data was consistent with the microPET findings in FaDu, H441, QG56 and Calu-1 xenografts, which correlated with HER3 expression levels. In conclusion, (89)Zr-RG7116 specifically accumulates in HER3 expressing tumors. PET imaging with this tracer provides real-time non-invasive information about RG7116 distribution, tumor targeting and tumor HER3 expression levels.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Immunoconjugates/pharmacokinetics , Isotopes/pharmacology , Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacology , Receptor, ErbB-3/immunology , Zirconium/pharmacology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Heterografts , Humans , Immunoconjugates/immunology , Isotopes/immunology , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms/immunology , Radiography , Radiopharmaceuticals/immunology , Zirconium/immunology
5.
Leuk Lymphoma ; 55(9): 2151-5160, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24304419

ABSTRACT

Obinutuzumab (GA101) is a novel glycoengineered type II CD20 antibody in development for non-Hodgkin lymphoma. We compared the anti-tumor activity of obinutuzumab and rituximab in preclinical studies using subcutaneous Z138 and WSU-DLCL2 xenograft mouse models. Obinutuzumab and rituximab were assessed alone and in combination with bendamustine, fludarabine, chlorambucil, doxorubicin and cyclophosphamide/vincristine. Owing to strong single-agent efficacy in these models, suboptimal doses of obinutuzumab were applied to demonstrate a combination effect. Obinutuzumab plus bendamustine achieved superior tumor growth inhibition versus rituximab plus bendamustine and showed a statistically significant effect versus the respective single treatments. Combinations of obinutuzumab with fludarabine, chlorambucil or cyclophosphamide/vincristine demonstrated significantly superior activity to rituximab-based treatment. Obinutuzumab monotherapy was at least as effective as rituximab plus chemotherapy in vivo, and obinutuzumab plus chemotherapy was superior to the respective monotherapies. These data support further clinical investigation of obinutuzumab plus chemotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Lymphoma/metabolism , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antigens, CD20/metabolism , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Disease Models, Animal , Female , Humans , Lymphoma/drug therapy , Lymphoma/mortality , Lymphoma/pathology , Mice , Tumor Burden , Vincristine/administration & dosage , Vincristine/pharmacology , Xenograft Model Antitumor Assays
6.
J Radiat Res ; 53(3): 343-52, 2012.
Article in English | MEDLINE | ID: mdl-22739003

ABSTRACT

Genetic factors can modify susceptibility to the carcinogenic effect of ionising radiation. To establish if radioiodine-induced thyroid cancer is similarly genetically influenced, we studied F1 hybrid crosses between inbred mouse strains. Mice were perinatally exposed to iodine-131 and thyroid tissues examined after 18 months. Differences in the incidence and distribution of histological subtypes were quantified in relation to genetic background. As expected, the occurrence of thyroid lesions was significantly higher in irradiated mouse hybrids than in unirradiated controls. The most frequent alterations were the simple and the complex hyperplasias, followed by follicular adenoma and, less frequently, follicular carcinoma. Both the incidence and distribution of the histiotype were different between the hybrid mouse crosses. Crosses using JF1 mice (M. m. molossinus) produced F1 offspring that were more resistant to radiation-induced thyroid lesions. Sequence analysis of Braf, Ret, Hras, Kras, Kit and Trp53, all genes that are commonly mutated in human thyroid cancers, did not show any evidence of mutation in the tumours. However, microsatellite analysis of genomic DNA revealed frequent allelic imbalances in complex hyperplasia and follicular adenoma. We conclude that genetic background, in particular the JF1 genotype, confer differences in susceptibility to the carcinogenic effects of radioiodine on the thyroid.


Subject(s)
Iodine Radioisotopes/adverse effects , Neoplasms, Radiation-Induced/etiology , Thyroid Neoplasms/etiology , Adenocarcinoma, Follicular/etiology , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Adenoma/etiology , Adenoma/genetics , Adenoma/pathology , Allelic Imbalance , Animals , Female , Genetic Predisposition to Disease , Humans , Hybridization, Genetic , Hyperplasia , Male , Mice , Mice, Inbred Strains , Mutation , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Proto-Oncogenes , Radiation Tolerance/genetics , Species Specificity , Thyroid Gland/pathology , Thyroid Gland/radiation effects , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
7.
Mol Cell Biol ; 32(13): 2416-27, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22508990

ABSTRACT

Cks1 is an activator of the SCF(Skp2) ubiquitin ligase complex that targets the cell cycle inhibitor p27(Kip1) for degradation. The loss of Cks1 results in p27(Kip1) accumulation and decreased proliferation and inhibits tumorigenesis. We identify here a function of Cks1 in mammalian cell cycle regulation that is independent of p27(Kip1). Specifically, Cks1(-/-); p27(Kip1-/-) mouse embryonic fibroblasts retain defects in the G(1)-S phase transition that are coupled with decreased Cdk2-associated kinase activity and defects in proliferation that are associated with Cks1 loss. Furthermore, concomitant loss of Cks1 does not rescue the tumor suppressor function of p27(Kip1) that is manifest in various organs of p27(Kip1-/-) mice. In contrast, defects in mitotic entry and premature senescence manifest in Cks1(-/-) cells are p27(Kip1) dependent. Collectively, these findings establish p27(Kip1)-independent functions of Cks1 in regulating the G(1)-S transition.


Subject(s)
CDC2-CDC28 Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , S Phase Cell Cycle Checkpoints/physiology , Animals , CDC2-CDC28 Kinases/deficiency , CDC2-CDC28 Kinases/genetics , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinases/metabolism , Female , Fibroblasts/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , G2 Phase Cell Cycle Checkpoints/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , S-Phase Kinase-Associated Proteins/metabolism
8.
J Nucl Cardiol ; 19(4): 775-84, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22527796

ABSTRACT

BACKGROUND: Expression of α(v)ß(3) integrin has been proposed as a marker for atherosclerotic lesion inflammation. We studied whether diet intervention reduces uptake of α(v)ß(3) integrin-targeted positron emission tomography tracer (18)F-galacto-RGD in mouse atherosclerotic plaques. METHODS AND RESULTS: Hypercholesterolemic LDLR(-/-) ApoB(100/100) mice on high-fat diet for 4 months were randomized to further 3 months on high-fat diet (high-fat group, n = 8) or regular mouse chow (intervention group, n = 7). Intima-media ratio describing plaque burden was comparable between intervention and high-fat groups (2.0 ± 0.5 vs 2.3 ± 0.8, P = .5). Uptake of (18)F-galacto-RGD in the aorta was lower in the intervention than high-fat group (%ID/g 0.16 vs 0.23, P < .01). Autoradiography showed 35% lower uptake of (18)F-galacto-RGD in the atherosclerotic plaques in the intervention than high-fat group (P = .007). Uptake of (18)F-galacto-RGD in plaques correlated with uptake of (3)H-deoxyglucose and nuclear density, which was lower in the intervention than high-fat group (P = .01). Flow cytometry demonstrated macrophages expressing α(v) and ß(3) integrins in the aorta. CONCLUSIONS: Uptake of (18)F-galacto-RGD in mouse atherosclerotic lesions was reduced by lipid-lowering diet intervention. Expression of α(v)ß(3) integrin is a potential target for evaluation of therapy response in atherosclerosis.


Subject(s)
Animal Feed , Galactose/analogs & derivatives , Integrin alphaVbeta3/metabolism , Peptides, Cyclic/pharmacology , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/diet therapy , Plaque, Atherosclerotic/diagnosis , Positron-Emission Tomography/methods , Animals , Apolipoproteins B/metabolism , Autoradiography/methods , Cholesterol/metabolism , Diet, High-Fat , Flow Cytometry/methods , Galactose/pharmacology , Humans , Hypercholesterolemia/genetics , Inflammation , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Radiopharmaceuticals/pharmacology
9.
J Clin Gastroenterol ; 44 Suppl 1: S10-5, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20562631

ABSTRACT

BACKGROUND AND AIM: Inflammatory bowel diseases (IBD) are immune-mediated chronic diseases that are characterized by an overreaction of the intestinal immune system to the intestinal microbiota. VSL#3, a mixture of 8 different lactic acid bacteria, is a clinically relevant probiotic compound in the context of IBD, but the bacterial structures and molecular mechanisms underlying the observed protective effects are largely unknown. The intestinal epithelium plays a very important role in the maintenance of the intestinal homeostasis, as the intestinal epithelial cells (IEC) are capable of sensing, processing, and reacting upon signals from the luminal microbiota and the intestinal immune system. This immune regulatory function of the IEC is lost in IBD owing to dysregulated activation of the IEC. Thus, the aim of this study was to reveal protective mechanisms of VSL#3 on IEC function. RESULTS: In vitro, VSL#3 was found to selectively inhibit activation-induced secretion of the T-cell chemokine interferon-inducible protein (IP)-10 in IEC. Cell wall-associated proteins of VSL#3-derived Lactobacillus casei (L. casei) were identified to be the active anti-inflammatory component of VSL#3. Mechanistically, L. casei did not impair initial IP-10 protein production, but induced posttranslational degradation of IP-10 in IEC. Feeding studies in tumor necrosis factor (TNF)Delta ARE/+ mice, a mouse model for experimental ileitis, revealed that neither VSL#3 nor L. casei is capable of reducing ileal inflammation. Even preweaning feeding of VSL#3 did not prevent the development of severe ileitis in TNF Delta ARE/+ mice. In contrast, VSL#3 feeding studies in IL-10-/- mice, a model for experimental colitis, revealed that VSL#3 has local, intestinal compartment-specific protective effects on the development of inflammation. Reduced histopathologic inflammation in the cecum of IL-10-/- mice after VSL#3 treatment was found to correlate with reduced levels of IP-10 protein in primary cecal epithelial cells. CONCLUSION AND OUTLOOK: These results suggest that the inhibitory effect of VSL#3-derived L. casei on IP-10 secretion in IEC is an important probiotic mechanism that contributes to the anti-inflammatory effects of VSL#3 in specific subsets of patients with IBD. An important future aim is the identification of the active probiotic protein, which could serve as a basis for the development of new efficient therapies in the context of IBD.


Subject(s)
Bacterial Proteins/metabolism , Chemokines/metabolism , Epithelial Cells/microbiology , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/microbiology , Lacticaseibacillus casei/metabolism , Probiotics , Protein Processing, Post-Translational , Animals , Chemokine CXCL10/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Interleukin-10/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Time Factors , Tumor Necrosis Factors/metabolism
10.
Int J Cancer ; 126(10): 2362-72, 2010 May 15.
Article in English | MEDLINE | ID: mdl-19876893

ABSTRACT

Pheochromocytomas are neoplasias of neural crest origin that arise from the chromaffin cells of the adrenal medulla. Pheochromocytomas arise with complete penetrance in rats homozygous for a germ-line frameshift mutation of Cdkn1b, encoding the cell cycle inhibitor p27KIP1 (MENX syndrome). We performed a genome-wide scan for allelic imbalance comparing 20 rat pheochromocytoma DNAs with normal rat DNA to better understand the pathobiology of the tumors and to correlate the findings with human pheochromocytoma. We identified allelic imbalance (AI) at candidate regions on rat chromosomes 8 and 19. Interestingly, the regions often lost in rat tumors are syntenic to regions involved in human pheochromocytomas. Fluorescence in situ hybridization analysis further validated the AI data. Sdhd and Rassf1a were analyzed in detail as they map to regions of AI on chromosome 8 and their homologues are implicated in human pheochromocytoma: we found no genetic mutations nor decreased expression. We also analyzed additional candidate genes, that is, rat homologues of genes predisposing to human pheochromocytoma and known tumor-suppressor genes, but we found no AI. In contrast, we observed frequent overexpression of Cdkn2a and Cdkn2c, encoding the cell cycle inhibitors p16INK4a and p18INK4c, respectively. The relative small number of allelic changes we found in rat pheochromocytoma might be related to their nonmalignant status and losses at chromosomes 8 and 19 are events that precede malignancy. Because of the high concordance of affected loci between rat and human tumors, studies of the MENX-associated pheochromocytomas should facilitate the identification of novel candidate genes implicated in their human counterpart.


Subject(s)
Adrenal Gland Neoplasms/genetics , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 8 , Cyclin-Dependent Kinase Inhibitor p27/genetics , Frameshift Mutation , Loss of Heterozygosity , Pheochromocytoma/genetics , Adrenal Gland Neoplasms/pathology , Animals , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Microsatellite Repeats , Pheochromocytoma/pathology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Up-Regulation
11.
Circ Cardiovasc Imaging ; 2(4): 331-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19808614

ABSTRACT

BACKGROUND: (18)F-Galacto-RGD is a positron emission tomography (PET) tracer binding to alpha(v)beta(3) integrin that is expressed by macrophages and endothelial cells in atherosclerotic lesions. Therefore, we evaluated (18)F-galacto-RGD for imaging vascular inflammation by studying its uptake into atherosclerotic lesions of hypercholesterolemic mice in comparison to deoxyglucose. METHODS AND RESULTS: Hypercholesterolemic LDLR(-/-)ApoB(100/100) mice on a Western diet and normally fed adult C57BL/6 control mice were injected with (18)F-galacto-RGD and (3)H-deoxyglucose followed by imaging with a small animal PET/CT scanner. The aorta was dissected 2 hours after tracer injection for biodistribution studies, autoradiography, and histology. Biodistribution of (18)F-galacto-RGD was higher in the atherosclerotic than in the normal aorta. Autoradiography demonstrated focal (18)F-galacto-RGD uptake in the atherosclerotic plaques when compared with the adjacent normal vessel wall or adventitia. Plaque-to-normal vessel wall ratios were comparable to those of deoxyglucose. Although angiogenesis was not detected, (18)F-galacto-RGD uptake was associated with macrophage density and deoxyglucose accumulation in the plaques. Binding to atherosclerotic lesions was efficiently blocked in competition experiments. In vivo imaging visualized (18)F-galacto-RGD uptake colocalizing with calcified lesions of the aortic arch as seen in CT angiography. CONCLUSIONS: (18)F-Galacto-RGD demonstrates specific uptake in atherosclerotic lesions of mouse aorta. In this model, its uptake was associated with macrophage density. (18)F-Galacto-RGD is a potential tracer for noninvasive imaging of inflammation in atherosclerotic lesions.


Subject(s)
Aorta, Thoracic/diagnostic imaging , Aortitis/diagnostic imaging , Atherosclerosis/diagnostic imaging , Galactose/analogs & derivatives , Hypercholesterolemia/complications , Integrin alphaVbeta3/metabolism , Peptides, Cyclic , Positron-Emission Tomography , Radiopharmaceuticals , Animals , Aorta, Thoracic/metabolism , Aortitis/etiology , Aortitis/metabolism , Aortography/methods , Apolipoprotein B-100/deficiency , Apolipoprotein B-100/genetics , Atherosclerosis/etiology , Atherosclerosis/metabolism , Autoradiography , Calcinosis/diagnostic imaging , Calcinosis/metabolism , Deoxyglucose/pharmacokinetics , Disease Models, Animal , Galactose/pharmacokinetics , Hypercholesterolemia/diagnostic imaging , Hypercholesterolemia/metabolism , Macrophages/diagnostic imaging , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptides, Cyclic/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tissue Distribution , Tomography, X-Ray Computed , Tritium
12.
Cell ; 137(5): 961-71, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19490899

ABSTRACT

It has been proposed that two amino acid substitutions in the transcription factor FOXP2 have been positively selected during human evolution due to effects on aspects of speech and language. Here, we introduce these substitutions into the endogenous Foxp2 gene of mice. Although these mice are generally healthy, they have qualitatively different ultrasonic vocalizations, decreased exploratory behavior and decreased dopamine concentrations in the brain suggesting that the humanized Foxp2 allele affects basal ganglia. In the striatum, a part of the basal ganglia affected in humans with a speech deficit due to a nonfunctional FOXP2 allele, we find that medium spiny neurons have increased dendrite lengths and increased synaptic plasticity. Since mice carrying one nonfunctional Foxp2 allele show opposite effects, this suggests that alterations in cortico-basal ganglia circuits might have been important for the evolution of speech and language in humans.


Subject(s)
Amino Acid Substitution , Basal Ganglia/metabolism , Biological Evolution , Forkhead Transcription Factors/metabolism , Vocalization, Animal , Animals , Dendrites/metabolism , Dopamine/metabolism , Gene Expression , Heterozygote , Humans , Language , Long-Term Synaptic Depression , Mice , Neural Pathways , Neuronal Plasticity , Speech
13.
Methods Mol Biol ; 530: 463-509, 2009.
Article in English | MEDLINE | ID: mdl-19266331

ABSTRACT

With the completion of the mouse genome sequence an essential task for biomedical sciences in the twenty-first century will be the generation and functional analysis of mouse models for every gene in the mammalian genome. More than 30,000 mutations in ES cells will be engineered and thousands of mouse disease models will become available over the coming years by the collaborative effort of the International Mouse Knockout Consortium. In order to realize the full value of the mouse models proper characterization, archiving and dissemination of mouse disease models to the research community have to be performed. Phenotyping centers (mouse clinics) provide the necessary capacity, broad expertise, equipment, and infrastructure to carry out large-scale systemic first-line phenotyping. Using the example of the German Mouse Clinic (GMC) we will introduce the reader to the different aspects of the organization of a mouse clinic and present selected methods used in first-line phenotyping.


Subject(s)
Information Storage and Retrieval/methods , Phenotype , Animals , Database Management Systems , Mice
14.
J Proteome Res ; 8(5): 2376-87, 2009 May.
Article in English | MEDLINE | ID: mdl-19323467

ABSTRACT

Evidence has linked genetic predisposition and environmental exposures to the worldwide pandemic of inflammatory bowel diseases (IBD), but underlying biochemical events remain largely undefined. Here, we studied the gradual development of colitis in Interleukin 10 deficient mice using a combination of (i) histopathological analysis of intestinal sections, (ii) metabolic profiling of blood plasma, and (iii) measurement of plasma inflammatory biomarkers. Data integration using chemometric tools, including Independent Component Analysis, provided a new strategy for measuring and mapping the metabolic effects associated with the development of intestinal inflammation at the age of 1, 8, 16, and 24 weeks. Chronic inflammation appeared at 8 weeks and onward, and was associated with altered cecum and colon morphologies and increased inflammatory cell infiltration into the mucosa and the submucosa. Blood plasma profiles provided additional evidence of loss of energy homeostasis, impaired metabolism of lipoproteins and glycosylated proteins. In particular, IL-10-/-mice were characterized by decreased levels of VLDL and increased concentrations of LDL and polyunsaturated fatty acids, which are related to the etiology of IBD. Moreover, higher levels of lactate, pyruvate, citrate and lowered glucose suggested increased fatty acid oxidation and glycolysis, while higher levels of free amino acids reflected muscle atrophy, breakdown of proteins and interconversions of amino acids to produce energy. These integrated system investigations demonstrate the potential of metabonomics for investigating the mechanistic basis of IBD, and it will provide novel avenues for management of IBD.


Subject(s)
Colitis/blood , Interleukin-10/deficiency , Metabolome , Metabolomics/methods , Amyloid/blood , Animals , Blood Glucose/metabolism , Cecum/metabolism , Cecum/pathology , Citrates/blood , Colitis/genetics , Colitis/metabolism , Colon/metabolism , Colon/pathology , Disease Progression , Fatty Acids, Unsaturated/blood , Interleukin-10/genetics , Interleukin-10/physiology , Lactates/blood , Lipoproteins, LDL/blood , Lipoproteins, VLDL/blood , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred Strains , Mice, Knockout , Pyruvates/blood , Receptors, Tumor Necrosis Factor, Type II/blood , Time Factors
15.
PLoS One ; 4(2): e4365, 2009.
Article in English | MEDLINE | ID: mdl-19197385

ABSTRACT

BACKGROUND: Clinical and experimental studies suggest that the probiotic mixture VSL#3 has protective activities in the context of inflammatory bowel disease (IBD). The aim of the study was to reveal bacterial strain-specific molecular mechanisms underlying the anti-inflammatory potential of VSL#3 in intestinal epithelial cells (IEC). METHODOLOGY/PRINCIPAL FINDINGS: VSL#3 inhibited TNF-induced secretion of the T-cell chemokine interferon-inducible protein (IP-10) in Mode-K cells. Lactobacillus casei (L. casei) cell surface proteins were identified as active anti-inflammatory components of VSL#3. Interestingly, L. casei failed to block TNF-induced IP-10 promoter activity or IP-10 gene transcription at the mRNA expression level but completely inhibited IP-10 protein secretion as well as IP-10-mediated T-cell transmigration. Kinetic studies, pulse-chase experiments and the use of a pharmacological inhibitor for the export machinery (brefeldin A) showed that L. casei did not impair initial IP-10 production but decreased intracellular IP-10 protein stability as a result of blocked IP-10 secretion. Although L. casei induced IP-10 ubiquitination, the inhibition of proteasomal or lysosomal degradation did not prevent the loss of intracellular IP-10. Most important for the mechanistic understanding, the inhibition of vesicular trafficking by 3-methyladenine (3-MA) inhibited IP-10 but not IL-6 expression, mimicking the inhibitory effects of L. casei. These findings suggest that L. casei impairs vesicular pathways important for the secretion of IP-10, followed by subsequent degradation of the proinflammatory chemokine. Feeding studies in TNF(DeltaARE) and IL-10(-/-) mice revealed a compartimentalized protection of VSL#3 on the development of cecal but not on ileal or colonic inflammation. Consistent with reduced tissue pathology in IL-10(-/-) mice, IP-10 protein expression was reduced in primary epithelial cells. CONCLUSIONS/SIGNIFICANCE: We demonstrate segment specific effects of probiotic intervention that correlate with reduced IP-10 protein expression in the native epithelium. Furthermore, we revealed post-translational degradation of IP-10 protein in IEC to be the molecular mechanism underlying the anti-inflammatory effect.


Subject(s)
Chemokine CXCL10/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Inflammation/microbiology , Intestines/cytology , Lacticaseibacillus casei/metabolism , Protein Processing, Post-Translational , Animals , Autophagy/drug effects , Bacterial Proteins/metabolism , Chemotaxis/drug effects , Chronic Disease , Colitis/microbiology , Colitis/pathology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Lacticaseibacillus casei/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Organ Specificity/drug effects , Probiotics/pharmacology , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology
16.
Transgenic Res ; 18(1): 71-87, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18819014

ABSTRACT

Murine embryonic stem cells (mESCs) inoculated at passage P13 with the mycoplasma species M. hominis, M. fermentans and M. orale and cultured over 20 passages showed reduced growth rate and viability (P < 0.0001) compared to control mESCs. Spectral karyotypic analysis of mycoplasma-infected mESCs showed a number of non-clonal chromosomal aberrations which increased with the duration of infection. The differentiation status of the infected mESCs was most affected at passage P13+6 where the infection was strongest and 46.3% of the mESCs expressed both POU5F1 and SSEA-1 markers whereas 84.8% of control mESCs expressed both markers. The percentage of germline chimeras from mycoplasma-infected mESCs was examined after blastocyst injection and embryo transfer to suitable recipients at different passages and, compared to the respective control group, was most affected at passage P13+5 (50% vs. 90%; P < 0.07). Further reductions were obtained at the same passage in the percentage of litters born (50% vs. 100%; P < 0.07) and in the percentage of pups born (22% vs. 45%; P < 0.001). Thirty three chimeras (39.8%) obtained from blastocyst injection with mycoplasma-infected mESCs showed reduced body weight (P < 0.0001), nasal discharge, osteoarthropathia, and cachexia. Flow cytometric analysis of plasma from chimeras produced with mycoplasma-infected mESCs revealed statistically significant differences in the proportions of T-cells and increased levels of IgG1 (P < 0.001), IgG2a (P < 0.05) and IgM (P < 0.05), anti-DNA antibodies (P < 0.05) and rheumatoid factor (P < 0.01). The present data indicate that mycoplasma contamination of mESCs affects various cell parameters, germline transmission, and postnatal development of the resulting chimeras.


Subject(s)
Chimera/physiology , Embryo, Mammalian/cytology , Embryonic Stem Cells/microbiology , Germ Cells/physiology , Mycoplasma/physiology , Animals , Biomarkers/analysis , Blastocyst/microbiology , Blastocyst/physiology , Cell Differentiation , Cell Survival , Chimera/microbiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fibroblasts/cytology , Immunoglobulin G/metabolism , Karyotyping , Male , Mice , Mycoplasma/isolation & purification , Pregnancy
17.
J Nutr ; 138(9): 1684-91, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18716170

ABSTRACT

Monoassociations of germ-free animals with colitogenic and probiotic bacterial strains trigger intestinal epithelial cell (IEC) activation and host-derived feedback mechanisms. To characterize the impact of a single nonpathogenic bacterial strain on the intestinal epithelium in the presence of an established microbiota, we inoculated reconstituted Lacotobacillus-free (RLF) mice at 8 wk of age with Lactobacillus reuteri 100-23. Primary IEC from the small intestine of L. reuteri-inoculated and control RLF mice were isolated 2, 6, and 21 d after inoculation followed by gene expression analysis (real-time PCR; Affymetrix microarrays) as well as 2-dimensional-gel electrophoreses (2D SDS-PAGE) and peptide mass fingerprinting via matrix-assisted laser desorption/ionization time of flight MS. At d 6, gene expression of proinflammatory cytokines and chemokines including interleukin (IL)-1alpha, IL-6, interferon-gamma-inducible protein 10, and macrophage inflammatory protein 2 was transiently induced, whereas gene expression levels of regulatory proteins A20 and Toll-interacting protein decreased. In addition, 8 target proteins with changes in the steady-state protein expression levels were identified at d 2 and 6 of L. reuteri colonization. Consistent with the absence of histopathology, L. reuteri-induced activation of primary IEC returned to control levels by d 21 after inoculation of RLF mice. The capability of L. reuteri 100-23 to directly trigger epithelial cell activation was confirmed in small IEC cultures using the murine cell line Mode-K. These results clearly indicate that the intestinal epithelium is reactive toward environmental changes induced by the commensal bacterial strain L. reuteri even in the presence of an already-established microbiota. The induction of transient IEC activation may help to maintain mucosal homeostasis.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/microbiology , Intestinal Mucosa/cytology , Limosilactobacillus reuteri/physiology , Animals , Cell Line , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Intestinal Mucosa/microbiology , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Specific Pathogen-Free Organisms
18.
Carcinogenesis ; 28(9): 2002-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17638921

ABSTRACT

Genetic predisposition and environmental factors act in concert in the pathogenesis of multi-factorial diseases. Selenoproteins represent fundamental antioxidative systems for the maintenance of cellular redox homeostasis, which is altered in various disease processes. Optimal function of selenoproteins requires availability of sufficient amounts of the essential trace element selenium, but in many countries the nutritive selenium supply is regarded insufficient. Supplemental selenium has been shown to have cancer-protective effects in a variety of experimental settings and clinical studies. Pancreatic carcinoma has so far not been tested as an end-point in such studies. We thus investigated the influence of supplemental nutritive selenium on pancreatic carcinogenesis in selenium-deficient animals by use of a genetically defined disease model. Over a period of 800 days, all animals (n = 131) in the study developed tumours. Within this time, the mean total tumour latency was not influenced by the selenium status (471 versus 472 days). Also, the mean latency of pancreatic carcinomas (n = 83) was not influenced (464 versus 466 days). In contrast, the percentage of pancreatic tumors within all tumours was lower in the selenium-deficient group (55 versus 70%). A highly significant difference in the differentiation grade of the pancreatic tumours was evident between the two groups: selenium-deficient mice (n = 33) developed predominantly undifferentiated anaplastic carcinomas (26 anaplastic versus 7 differentiated), whereas in the selenium-supplemented group (n = 50) mainly well-differentiated carcinomas were detected (20 anaplastic versus 30 differentiated). These data point at a new role of the trace element selenium in carcinogenesis.


Subject(s)
Adenocarcinoma/epidemiology , Adenocarcinoma/pathology , Dietary Supplements , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/pathology , Selenium/pharmacology , Transcription Factors/genetics , Transforming Growth Factor alpha/genetics , Tumor Suppressor Protein p53/genetics , Animals , Elongin , Incidence , Mice , Mice, Inbred BALB C , Mice, Knockout , Selenium/administration & dosage , Transcription Factors/deficiency , Tumor Suppressor Protein p53/deficiency
19.
J Nutr ; 137(5): 1208-15, 2007 May.
Article in English | MEDLINE | ID: mdl-17449583

ABSTRACT

Flavonoids may play an important role for adjunct nutritional therapy of chronic intestinal inflammation. In this study, we characterized the molecular mechanisms by which quercetin and its enteric bacterial metabolites, taxifolin, alphitonin, and 3, 4-dihydroxy-phenylacetic acid, inhibit tumor necrosis factor alpha (TNF)-induced proinflammatory gene expression in the murine small intestinal epithelial cell (IEC) line Mode-K as well as in heterozygous TNFDeltaARE/WT mice, a murine model of experimental ileitis. Quercetin inhibited TNF-induced interferon-gamma-inducible protein 10 (IP-10) and macrophage inflammatory protein 2 (MIP-2) gene expression in Mode-K cells with effective inhibitory concentration of 40 and 44 micromol/L, respectively. Interestingly, taxifolin, alphitonin, and 3,4-dihydroxy-phenylacetic acid did not inhibit TNF responses in IEC, suggesting that microbial transformation of quercetin completely abolished its anti-inflammatory effect. At the molecular level, quercetin inhibited Akt phosphorylation but did not inhibit TNF-induced RelA/I-kappaB phosphorylation and IkappaB degradation or TNF-alpha-induced nuclear factor-kappaB transcriptional activity. Most important for understanding the mechanism involved, chromatin immunoprecipitation analysis revealed inhibitory effects of quercetin on phospho-RelA recruitment to the IP-10 and MIP-2 gene promoters. In addition, and consistent with the lack of cAMP response element binding protein (CBP)/p300 recruitment and phosphorylation/acetylation of histone 3 at the promoter binding site, quercetin inhibited histone acetyl transferase activity. The oral application of quercetin to heterozygous TNFDeltaARE/WT mice [10 mg/(d x kg body wt)] significantly inhibited IP-10 and MIP-2 gene expression in primary ileal epithelial cells but did not affect tissue pathology. These studies support an anti-inflammatory effect of quercetin in epithelial cells through mechanisms that inhibit cofactor recruitment at the chromatin of proinflammatory genes.


Subject(s)
Ileitis/genetics , Intestinal Mucosa/metabolism , NF-kappa B/genetics , Promoter Regions, Genetic , Quercetin/pharmacology , Transcription Factors/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Line , Chemokine CXCL10 , Chemokine CXCL2 , Chemokines/genetics , Chemokines/metabolism , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Epithelial Cells/metabolism , Gene Expression/drug effects , Genes, Reporter/drug effects , Ileitis/metabolism , Intestinal Mucosa/drug effects , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelA/physiology , Transcription Factors/metabolism
20.
Toxicol Pathol ; 35(3): 366-75, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17455084

ABSTRACT

Immunohistochemistry is an indispensable tool in human pathology enabling immunophenotypic characterization of tumor cells. Immunohistochemical analyses of mouse models of human hematopoietic neoplasias have become an important aspect for comparison of murine entities with their human counterparts. The aim of this study was to establish a diagnostic antibody panel for analysis of murine lymphomas/leukemias, useful in formalin-fixed/paraffin-embedded tissue. Overall, 48 antibodies (4 rabbit monoclonal, 12 rabbit polyclonal, 2 goat polyclonal, 11 rat, and 19 mouse monoclonal), which were either mouse-specific (14) or cross-reactive with murine tissue (34) were tested for staining quality and diagnostic value in 468 murine hematopoietic neoplasms. Specific staining was achieved with 29 antibodies, of which 18 were human antibodies cross-reactive with murine tissue. Only 23 (B220, BCL-2, BCL-6, CD117, CD138 (2x), CD3 (2x), CD43, CD45, CD5, CD79 alpha cy, cyclin D1, Ki-67 (2x), Mac-3, Mac-2, lysozyme, mast cell tryptase, MPO, Pax-5, TdT, and TER-119) were regarded as valuable for diagnostic evaluation. Immunohistochemistry was also established in an automated immunostainer for high throughput analysis. The antibody panel developed is useful for the classification of murine lymphomas and leukemias analyzed, and a valuable tool for human and veterinary pathologists involved in the diagnostic interpretation of murine models of hematopoietic neoplasias.


Subject(s)
Aging/immunology , Antibodies/immunology , Cross Reactions , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Immunohistochemistry/methods , Animals , Antibodies/genetics , Female , Fixatives/chemistry , Formaldehyde/chemistry , Hematologic Neoplasms/classification , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Paraffin Embedding
SELECTION OF CITATIONS
SEARCH DETAIL
...