Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cancers (Basel) ; 14(17)2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36077614

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), due to its genomic heterogeneity and lack of effective treatment, despite decades of intensive research, will become the second leading cause of cancer-related deaths by 2030. Step-wise acquisition of mutations, due to genomic instability, is considered to drive the development of PDAC; the KRAS mutation occurs in 95 to 100% of human PDAC, and is already detectable in early premalignant lesions designated as pancreatic intraepithelial neoplasia (PanIN). This mutation is possibly the key event leading to genomic instability and PDAC development. Our study aimed to investigate the role of the error-prone DNA double-strand breaks (DSBs) repair pathway, alt-EJ, in the presence of the KRAS G12D mutation in pancreatic cancer development. Our findings show that oncogenic KRAS contributes to increasing the expression of Polθ, Lig3, and Mre11, key components of alt-EJ in both mouse and human PDAC models. We further confirm increased catalytic activity of alt-EJ in a mouse and human model of PDAC bearing the KRAS G12D mutation. Subsequently, we focused on estimating the impact of alt-EJ inactivation by polymerase theta (Polθ) deletion on pancreatic cancer development, and survival in genetically engineered mouse models (GEMMs) and cancer patients. Here, we show that even though Polθ deficiency does not fully prevent the development of pancreatic cancer, it significantly delays the onset of PanIN formation, prolongs the overall survival of experimental mice, and correlates with the overall survival of pancreatic cancer patients in the TCGA database. Our study clearly demonstrates the role of alt-EJ in the development of PDAC, and alt-EJ may be an attractive therapeutic target for pancreatic cancer patients.

2.
Int J Mol Sci ; 23(9)2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35563322

ABSTRACT

The B-cell CLL/lymphoma 11B gene (BCL11B) plays a crucial role in T-cell development, but its role in T-cell malignancies is still unclear. To study its role in the development of T-cell neoplasms, we generated an inducible BCL11B knockout in a murine T cell leukemia/lymphoma model. Mice, bearing human oncogenes TAL BHLH Transcription Factor 1 (TAL1; SCL) or LIM Domain Only 1 (LMO1), responsible for T-cell acute lymphoblastic leukemia (T-ALL) development, were crossed with BCL11B floxed and with CRE-ER/lox mice. The mice with a single oncogene BCL11Bflox/floxCREtg/tgTAL1tg or BCL11Bflox/floxCREtg/tgLMO1tg were healthy, bred normally, and were used to maintain the mice in culture. When crossed with each other, >90% of the double transgenic mice BCL11Bflox/floxCREtg/tgTAL1tgLMO1tg, within 3 to 6 months after birth, spontaneously developed T-cell leukemia/lymphoma. Upon administration of synthetic estrogen (tamoxifen), which binds to the estrogen receptor and activates the Cre recombinase, the BCL11B gene was knocked out by excision of its fourth exon from the genome. The mouse model of inducible BCL11B knockout we generated can be used to study the role of this gene in cancer development and the potential therapeutic effect of BCL11B inhibition in T-cell leukemia and lymphoma.


Subject(s)
Leukemia, T-Cell , Transcription Factors , Animals , Disease Models, Animal , LIM Domain Proteins/genetics , Leukemia, T-Cell/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , Repressor Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics
3.
PLoS One ; 16(12): e0260878, 2021.
Article in English | MEDLINE | ID: mdl-34879092

ABSTRACT

Proper and size selective blood filtration in the kidney depends on an intact morphology of podocyte foot processes. Effacement of interdigitating podocyte foot processes in the glomeruli causes a leaky filtration barrier resulting in proteinuria followed by the development of chronic kidney diseases. Since the function of the filtration barrier is depending on a proper actin cytoskeleton, we studied the role of the important actin-binding protein palladin for podocyte morphology. Podocyte-specific palladin knockout mice on a C57BL/6 genetic background (PodoPalldBL/6-/-) were back crossed to a 129 genetic background (PodoPalld129-/-) which is known to be more sensitive to kidney damage. Then we analyzed the morphological changes of glomeruli and podocytes as well as the expression of the palladin-binding partners Pdlim2, Lasp-1, Amotl1, ezrin and VASP in 6 and 12 months old mice. PodoPalld129-/- mice in 6 and 12 months showed a marked dilatation of the glomerular tuft and a reduced expression of the mesangial marker protein integrin α8 compared to controls of the same age. Furthermore, ultrastructural analysis showed significantly more podocytes with morphological deviations like an enlarged sub-podocyte space and regions with close contact to parietal epithelial cells. Moreover, PodoPalld129-/- of both age showed a severe effacement of podocyte foot processes, a significantly reduced expression of pLasp-1 and Pdlim2, and significantly reduced mRNA expression of Pdlim2 and VASP, three palladin-interacting proteins. Taken together, the results show that palladin is essential for proper podocyte morphology in mice with a 129 background.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/physiology , Genetic Background , Homeodomain Proteins/metabolism , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , Podocytes/metabolism , Actin Cytoskeleton , Adaptor Proteins, Signal Transducing/genetics , Animals , Cytoskeletal Proteins/genetics , Homeodomain Proteins/genetics , Kidney/metabolism , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Phosphorylation , Podocytes/cytology
4.
Ann Work Expo Health ; 64(6): 604-613, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32313934

ABSTRACT

Occupational noise exposure is a known risk factor for hearing loss and also adverse cardiovascular effects have been suggested. A job exposure matrix (JEM) would enable studies of noise and health on a large scale. The objective of this study was to create a quantitative JEM for occupational noise exposure assessment of the general working population. Between 2001-2003 and 2009-2010, we recruited workers from companies within the 10 industries with the highest reporting of noise-induced hearing loss according to the Danish Working Environment Authority and in addition workers of financial services and children day care to optimize the range in exposure levels. We obtained 1343 personal occupational noise dosimeter measurements among 1140 workers representing 100 different jobs according to the Danish version of the International Standard Classification of Occupations 1988 (DISCO 88). Four experts used 35 of these jobs as benchmarks and rated noise levels for the remaining 337 jobs within DISCO 88. To estimate noise levels for all 372 jobs, we included expert ratings together with sex, age, occupational class, and calendar year as fixed effects, while job and worker were included as random effects in a linear mixed regression model. The fixed effects explained 40% of the total variance: 72% of the between-jobs variance, -6% of the between-workers variance and 4% of the within-worker variance. Modelled noise levels showed a monotonic increase with increasing expert score and a 20 dB difference between the highest and lowest exposed jobs. Based on the JEM estimates, metal wheel-grinders were among the highest and finance and sales professionals among the lowest exposed. This JEM of occupational noise exposure can be used to prioritize preventive efforts of occupational noise exposure and to provide quantitative estimates of contemporary exposure levels in epidemiological studies of health effects potentially associated with noise exposure.


Subject(s)
Noise, Occupational , Occupational Exposure , Humans , Industry , Noise, Occupational/adverse effects , Occupations , Risk Factors
5.
Toxins (Basel) ; 12(2)2020 01 24.
Article in English | MEDLINE | ID: mdl-31991690

ABSTRACT

Rats are a reservoir of human- and livestock-associated methicillin-resistant Staphylococcus aureus (MRSA). However, the composition of the natural S. aureus population in wild and laboratory rats is largely unknown. Here, 144 nasal S. aureus isolates from free-living wild rats, captive wild rats and laboratory rats were genotyped and profiled for antibiotic resistances and human-specific virulence genes. The nasal S. aureus carriage rate was higher among wild rats (23.4%) than laboratory rats (12.3%). Free-living wild rats were primarily colonized with isolates of clonal complex (CC) 49 and CC130 and maintained these strains even in husbandry. Moreover, upon livestock contact, CC398 isolates were acquired. In contrast, laboratory rats were colonized with many different S.aureus lineages-many of which are commonly found in humans. Five captive wild rats were colonized with CC398-MRSA. Moreover, a single CC30-MRSA and two CC130-MRSA were detected in free-living or captive wild rats. Rat-derived S. aureus isolates rarely harbored the phage-carried immune evasion gene cluster or superantigen genes, suggesting long-term adaptation to their host. Taken together, our study revealed a natural S. aureus population in wild rats, as well as a colonization pressure on wild and laboratory rats by exposure to livestock- and human-associated S.aureus, respectively.


Subject(s)
Animals, Wild/microbiology , Staphylococcal Infections/epidemiology , Staphylococcus aureus/isolation & purification , Animals , Anti-Bacterial Agents/pharmacology , Blood Coagulation , Czech Republic , Ecosystem , Germany , Methicillin/pharmacology , Molecular Epidemiology , Nose/microbiology , Rats, Sprague-Dawley , Staphylococcal Infections/veterinary , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Virulence Factors/genetics
6.
J Exp Neurosci ; 13: 1179069519883580, 2019.
Article in English | MEDLINE | ID: mdl-31765441

ABSTRACT

Currently, it is controversially discussed whether a relationship between obesity and cognition exists. We here analyzed a mouse model of obesity (leptin-deficient mice) to study the effects of obesity on the morphology of the hippocampus (a brain structure involved in mechanisms related to learning and memory) and on behavior. Mice aged 4 to 6 months were analyzed. At this age, the obese mice have nearly double the body weight as controls, but display smaller brains (brain volume is about 10% smaller) as control animals of the same age. Adult hippocampal neurogenesis, a process that is linked to learning and memory, might be disturbed in the obese mice and contribute to the smaller brain volume. Adult hippocampal neurogenesis was examined using specific markers for cell proliferation (phosphohistone H3), neuronal differentiation (doublecortin), and apoptosis (caspase 3). The number of phosphohistone H3 and doublecortin-positive cells was markedly reduced in leptin-deficient mice, but not the number of apoptotic cells, indicating that adult hippocampal neurogenesis on the level of cell proliferation was affected. In addition, dendritic spine densities of pyramidal neurons in the hippocampal area CA1 were analyzed using Golgi impregnation. However, no significant change in dendritic spine densities was noted in the obese mice. Moreover, the performance of the mice was analyzed in the open field as well as in the Morris water maze. In the open field test, obese mice showed reduced locomotor activity, but in the Morris water maze they showed similar performance compared with control animals.

7.
J Am Soc Nephrol ; 29(6): 1662-1678, 2018 06.
Article in English | MEDLINE | ID: mdl-29720549

ABSTRACT

Background Podocyte loss and effacement of interdigitating podocyte foot processes are the major cause of a leaky filtration barrier and ESRD. Because the complex three-dimensional morphology of podocytes depends on the actin cytoskeleton, we studied the role in podocytes of the actin bundling protein palladin, which is highly expressed therein.Methods We knocked down palladin in cultured podocytes by siRNA transfection or in zebrafish embryos by morpholino injection and studied the effects by immunofluorescence and live imaging. We also investigated kidneys of mice with podocyte-specific knockout of palladin (PodoPalld-/- mice) by immunofluorescence and ultrastructural analysis and kidney biopsy specimens from patients by immunostaining for palladin.Results Compared with control-treated podocytes, palladin-knockdown podocytes had reduced actin filament staining, smaller focal adhesions, and downregulation of the podocyte-specific proteins synaptopodin and α-actinin-4. Furthermore, palladin-knockdown podocytes were more susceptible to disruption of the actin cytoskeleton with cytochalasin D, latrunculin A, or jasplakinolide and showed altered migration dynamics. In zebrafish embryos, palladin knockdown compromised the morphology and dynamics of epithelial cells at an early developmental stage. Compared with PodoPalld+/+ controls, PodoPalld-/- mice developed glomeruli with a disturbed morphology, an enlarged subpodocyte space, mild effacement, and significantly reduced expression of nephrin and vinculin. Furthermore, nephrotoxic serum injection led to significantly higher levels of proteinuria in PodoPalld-/- mice than in controls. Kidney biopsy specimens from patients with diabetic nephropathy and FSGS showed downregulation of palladin in podocytes as well.Conclusions Palladin has an important role in podocyte function in vitro and in vivo.


Subject(s)
Actins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Podocytes/metabolism , Animals , Cytoskeleton , Female , Focal Adhesions , Gene Expression , Gene Silencing , Humans , Kidney Glomerulus/pathology , Male , Mice, Knockout , Microfilament Proteins/metabolism , Morpholinos/pharmacology , Podocytes/pathology , RNA, Messenger/metabolism , Vinculin/genetics , Vinculin/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
8.
Int J Med Microbiol ; 308(6): 598-606, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29174495

ABSTRACT

We previously reported that laboratory mice from all global vendors are frequently colonized with Staphylococcus aureus (S. aureus). Genotyping of a snap sample of murine S. aureus isolates from Charles River, US, showed that mice were predominantly colonized with methicillin-sensitive CC88 strains. Here, we expanded our view and investigated whether laboratory mice from other global animal facilities are colonized with similar strains or novel S. aureus lineages, and whether the murine S. aureus isolates show features of host adaptation. In total, we genotyped 230 S. aureus isolates from various vendor facilities of laboratory mice around the globe (Charles River facilities in the USA, Canada, France, and Germany; another US facility) and university- or company-associated breeding facilities in Germany, China and New Zealand. Spa typing was performed to analyse the clonal relationship of the isolates. Moreover, multiplex PCRs were performed for human-specific virulence factors, the immune-evasion cluster (IEC) and superantigen genes (SAg). We found a total of 58 different spa types that clustered into 15 clonal complexes (CCs). Three of these S. aureus lineages had spread globally among laboratory mice and accounted for three quarters of the isolates: CC1 (13.5%), CC15 (14.3%), and CC88 (47.0%). Compared to human colonizing isolates of the same lineages, the murine isolates frequently lacked IEC genes and SAg genes on mobile genetic elements, implying long-term adaptation to the murine host. In conclusion, laboratory mice from various vendors are colonized with host-adapted S. aureus-strains of a few lineages, predominantly the CC88 lineage. S. aureus researchers must be cautioned that S. aureus colonization might be a relevant confounder in infection and vaccination studies and are therefore advised to screen their mice before experimentation.


Subject(s)
Animals, Laboratory/microbiology , Staphylococcal Infections/transmission , Staphylococcus aureus/classification , Adaptation, Physiological , Animals , Anti-Bacterial Agents/pharmacology , Breeding , Canada , China , Drug Resistance, Bacterial/genetics , France , Genotype , Germany , Immune Evasion , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Multilocus Sequence Typing , New Zealand , Staphylococcus aureus/drug effects , Staphylococcus aureus/isolation & purification , United States , Virulence Factors/genetics
9.
Article in English | MEDLINE | ID: mdl-28512627

ABSTRACT

Whether mice are an appropriate model for S. aureus infection and vaccination studies is a matter of debate, because they are not considered as natural hosts of S. aureus. We previously identified a mouse-adapted S. aureus strain, which caused infections in laboratory mice. This raised the question whether laboratory mice are commonly colonized with S. aureus and whether this might impact on infection experiments. Publicly available health reports from commercial vendors revealed that S. aureus colonization is rather frequent, with rates as high as 21% among specific-pathogen-free mice. In animal facilities, S. aureus was readily transmitted from parents to offspring, which became persistently colonized. Among 99 murine S. aureus isolates from Charles River Laboratories half belonged to the lineage CC88 (54.5%), followed by CC15, CC5, CC188, and CC8. A comparison of human and murine S. aureus isolates revealed features of host adaptation. In detail, murine strains lacked hlb-converting phages and superantigen-encoding mobile genetic elements, and were frequently ampicillin-sensitive. Moreover, murine CC88 isolates coagulated mouse plasma faster than human CC88 isolates. Importantly, S. aureus colonization clearly primed the murine immune system, inducing a systemic IgG response specific for numerous S. aureus proteins, including several vaccine candidates. Phospholipase C emerged as a promising test antigen for monitoring S. aureus colonization in laboratory mice. In conclusion, laboratory mice are natural hosts of S. aureus and therefore, could provide better infection models than previously assumed. Pre-exposure to the bacteria is a possible confounder in S. aureus infection and vaccination studies and should be monitored.


Subject(s)
Disease Models, Animal , Mice/immunology , Mice/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Ampicillin Resistance , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacteriophages/enzymology , Bacteriophages/genetics , Drug Resistance, Bacterial , Genotype , Humans , Immune Evasion/genetics , Immunoglobulin G/blood , Interspersed Repetitive Sequences/genetics , Interspersed Repetitive Sequences/immunology , Male , Mice, Inbred C57BL , Multigene Family , Staphylococcal Infections/transmission , Staphylococcal Protein A/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , Type C Phospholipases/immunology , Vaccination , Virulence/genetics , Virulence/immunology , Virulence Factors/genetics , Virulence Factors/immunology
10.
J Immunol ; 198(5): 1910-1920, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28115529

ABSTRACT

T cell activation is an energy-demanding process fueled by increased glucose consumption and accompanied by upregulation of the insulin receptor (INSR). In this article, we report that silencing the INSR in inducible knockdown rats impairs selective T cell functions but not thymocyte development. Glucose transport and glycolysis in activated CD4+ T cells were compromised in the absence of the INSR, which was associated with alterations in intracellular signaling pathways. The observed metabolic defects coincided with reduced cytokine production, proliferation, and migration, as well as increased apoptosis of CD4+ T cells. The cytotoxicity of CD8+ T cells in response to alloantigens was also diminished under these conditions, whereas the frequency and suppressive capacity of regulatory T cells were unaffected. The observed impairments proved to be decisive in vivo because silencing of the INSR attenuated clinical symptoms in animal models of acute graft-versus-host disease and multiple sclerosis. Taken together, our results suggest that upregulation of the INSR on T cells following activation is required for efficient adaptive immunity.


Subject(s)
Adaptive Immunity , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/physiology , Receptor, Insulin/deficiency , Receptor, Insulin/physiology , Thymocytes/physiology , Animals , Apoptosis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Gene Knockdown Techniques , Glucose/metabolism , Glycolysis/immunology , Graft vs Host Disease/immunology , Lymphocyte Activation/immunology , Rats , Receptor, Insulin/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymocytes/immunology
11.
Br J Pharmacol ; 174(2): 163-176, 2017 01.
Article in English | MEDLINE | ID: mdl-27858997

ABSTRACT

BACKGROUND AND PURPOSE: Therapeutic options for treating glomerulopathies, the main cause of chronic kidney disease, are limited. Podocyte dedifferentiation is a major event in the pathogenesis of glomerulopathies. The goal of the present study was, therefore, to develop an assay to monitor podocyte differentiation suitable for compound screening. EXPERIMENTAL APPROACH: We isolated and cultured glomeruli from transgenic mice, expressing cyan fluorescent protein (CFP) under the control of the promoter of nephrin, a marker of podocyte differentiation. Mean CFP fluorescence intensity per glomerulus (MFG) was determined by summation of all glomerular voxels from confocal z-stacks in the absence and presence of pharmaceutical compounds. KEY RESULTS: In untreated cultured glomeruli, MFG remained fairly stable during the first 5 days, when foot processes were already effaced, and the level of many podocyte-specific proteins was only mildly affected, as revealed by proteomics. Between day 6 and 9, MFG decreased to almost zero. The decrease in MFG was paralleled by a decrease in CFP and nephrin expression, as determined by RT-PCR, western blots and proteomics. Puromycin aminonucleoside (PAN), which damages podocytes, concentration-dependently induced a complete loss of MFG. Dexamethasone (25 µM) and pioglitazone (10 µM) markedly attenuated the effect of 0.6 µg·mL-1 PAN on MFG. CONCLUSION AND IMPLICATIONS: In summary, we established a novel assay to assess the effect of pharmaceutical compounds on the differentiation of podocytes in situ. Our assay is suitable for compound screening to identify drugs for the treatment of glomerulopathies.


Subject(s)
Cell Differentiation/drug effects , Dexamethasone/pharmacology , Microscopy, Electron, Scanning , Podocytes/drug effects , Puromycin Aminonucleoside/pharmacology , Thiazolidinediones/pharmacology , Animals , Dose-Response Relationship, Drug , Mice , Mice, Transgenic , Pioglitazone , Podocytes/cytology , Structure-Activity Relationship
12.
Endocrinology ; 157(12): 4677-4690, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27715254

ABSTRACT

The angiotensin-converting enzyme 2/angiotensin (Ang)-(1-7)/Mas axis of the renin-angiotensin system often opposes the detrimental effects of the angiotensin-converting enzyme/Ang II/Ang II type 1 receptor axis and has been associated with beneficial effects on glucose homeostasis, whereas underlying mechanisms are mostly unknown. Here we investigate the effects of Ang-(1-7) and its receptor Mas on ß-cell function. Isolated islets from Mas-deficient and wild-type mice were stimulated with Ang-(1-7) or its antagonists and effects on insulin secretion determined. Islets' cytoplasmic calcium and cAMP concentrations, mRNA amounts of Ins1, Ins2, Pdx1, and Mafa and effects of inhibitors of cAMP downstream signaling were determined. Ang-(1-7) was also applied to mice by osmotic pumps for 14 days and effects on glucose tolerance and insulin secretion were assessed. Ang-(1-7) increased insulin secretion from wild-type islets, whereas antagonists and genetic Mas deficiency led to reduced insulin secretion. The Mas-dependent effects of Ang-(1-7) on insulin secretion did not result from changes in insulin gene expression or changes in the excitation-secretion coupling but from increased intracellular cAMP involving exchange protein activated directly by cAMP. Administration of Ang-(1-7) in vivo had only marginal effects on glucose tolerance in wild-type mice but still resulted in improved insulin secretion from islets isolated of these mice. Interestingly, although less pronounced than in wild types, Ang-(1-7) still affected insulin secretion in Mas-deficient islets. The data indicate a significant function of Ang-(1-7) in the regulation of insulin secretion from mouse islets in vitro and in vivo, mainly, but not exclusively, by Mas-dependent signaling, modulating the accessory pathway of insulin secretion via increase in cAMP.


Subject(s)
Angiotensin I/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Animals , Cyclic AMP/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin Resistance/physiology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Mice, Knockout , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
13.
FASEB J ; 30(5): 1724-32, 2016 05.
Article in English | MEDLINE | ID: mdl-26740263

ABSTRACT

T-cell lymphopenia is a major risk factor for autoimmunity. Here we describe congenic Lewis (LEW) rats with a loss-of-function mutation in the Gimap5 gene, leading to a 92% reduction in peripheral T-cell numbers. Gimap5-deficient LEW rats developed eosinophilic autoimmune gastroenteritis accompanied by a 40-fold increase in IgE serum levels. This phenotype was ameliorated by antibiotic treatment, indicating a critical role of the microbial flora in the development of inflammatory bowel disease. Interestingly, Gimap5-deficient LEW rats showed strongly aggravated experimental autoimmune encephalomyelitis (EAE) after immunization with guinea pig myelin basic protein. This phenotype, however, persisted after antibiosis, confirming that the enhanced CNS autoimmune response in T-cell lymphopenic Gimap5-deficient LEW rats was unrelated to the composition of the microbial flora. Rather, it seems that it was caused by the 7-fold increase in the percentage of activated T cells producing IL-17 and IFN-γ, and the skewed T-cell receptor (TCR) repertoire, both of which were the result of T-cell lymphopenia and not affected by antibiosis. This notion was supported by the observation that adoptive T-cell transfer corrected the TCR repertoire and improved EAE. Collectively, our findings confirm a critical albeit differential role of T-cell lymphopenia in the susceptibility to organ-specific autoimmune responses.-Fischer, H. J., Witte, A.-K., Walter, L., Gröne, H.-J., van den Brandt, J., Reichardt, H. M. Distinct roles of T-cell lymphopenia and the microbial flora for gastrointestinal and CNS autoimmunity.


Subject(s)
Autoimmune Diseases/microbiology , Central Nervous System/microbiology , GTP-Binding Proteins/metabolism , Gastrointestinal Tract/immunology , Lymphopenia , T-Lymphocytes/physiology , Adoptive Transfer , Animals , Autoimmune Diseases/metabolism , Central Nervous System/immunology , Central Nervous System Diseases/genetics , Central Nervous System Diseases/metabolism , GTP-Binding Proteins/genetics , Gastroenteritis/genetics , Gastroenteritis/metabolism , Gastroenteritis/microbiology , Gastrointestinal Tract/microbiology , Gene Expression Regulation/immunology , Point Mutation , Rats
14.
J Neuroimmunol ; 290: 26-32, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26711565

ABSTRACT

MS is a highly prevalent neuroinflammatory disease of presumed autoimmune origin. Clinical observations and animal studies suggest that CD8(+) T cells play an important role in MS but their exact mechanisms are ill defined. When we actively induced EAE in CD8 knock-out DA rats, or adoptively transferred encephalitogenic CD4(+) T cells into CD8 knock-out DA rats, the disease course was indistinguishable from controls. Since our previous findings had revealed that the absence of CD8(+) T cells in Lewis rats ameliorated EAE, we compared antigen-induced T cell differentiation in both strains. Disease onset and the composition of the draining lymph nodes were similar but T cell activation in DA rats was much weaker. Moreover, oligoclonal expansion of CD8(+) T cells was exclusively observed in Lewis but not in DA rats. This suggests that myelin-specific CD8(+) T cells are involved in the differentiation of encephalitogenic CD4(+) T cells in Lewis rats, whilst they do not impact CD4(+) T cell priming in DA rats. Hence, clonal expansion of CD8(+) T cells in secondary lymphoid organs appears to be linked to their ability to modulate CNS autoimmune responses.


Subject(s)
Adoptive Transfer , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Adoptive Transfer/methods , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Gene Knockout Techniques , Rats , Rats, Inbred Lew
15.
PLoS One ; 10(9): e0135444, 2015.
Article in English | MEDLINE | ID: mdl-26327508

ABSTRACT

A20 is an ubiquitin-editing enzyme that ensures the transient nature of inflammatory signaling pathways induced by cytokines like TNF-α and IL-1 or pathogens via Toll-like receptor (TLR) pathways. It has been identified as a negative regulator of dendritic cell (DC) maturation and attenuator of their immunostimulatory properties. Ex vivo A20-depleted dendritic cells showed enhanced expression of pro-inflammatory cytokines and costimulatory molecules, which resulted in hyperactivation of tumor-infiltrating T lymphocytes and inhibition of regulatory T cells. In the present study, we demonstrate that a synthetic molecule consisting of a CpG oligonucleotide TLR9 agonist linked to A20-specific siRNAs silences its expression in TLR9+ mouse dendritic cells in vitro and in vivo. In the B16 mouse melanoma tumor model, silencing of A20 enhances the CpG-triggered induction of NFκB activity followed by elevated expression of IL-6, TNF-α and IL-12. This leads to potentiated antitumor immune responses manifested by increased numbers of tumor-specific cytotoxic T cells, high levels of tumor cell apoptosis and delayed tumor growth. Our findings confirm the central role of A20 in controlling the immunostimulatory potency of DCs and provide a strategy for simultaneous A20 silencing and TLR activation in vivo.


Subject(s)
Cysteine Endopeptidases/drug effects , Gene Silencing , Intracellular Signaling Peptides and Proteins/drug effects , Melanoma, Experimental/drug therapy , RNA, Small Interfering/pharmacology , Toll-Like Receptor 9/drug effects , Animals , Blotting, Western , Cell Line, Tumor , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Immunity, Humoral/drug effects , Immunity, Humoral/physiology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Molecular Targeted Therapy , NF-kappa B/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3
16.
Eur J Immunol ; 45(5): 1326-38, 2015 May.
Article in English | MEDLINE | ID: mdl-25627579

ABSTRACT

Brain-derived neurotrophic factor (BDNF) promotes neuronal survival, regeneration, and plasticity. Emerging evidence also indicates an essential role for BDNF outside the nervous system, for instance in immune cells. We therefore investigated the impact of BDNF on T cells using BDNF knockout (KO) mice and conditional KO mice lacking BDNF specifically in this lymphoid subset. In both settings, we observed diminished T-cell cellularity in peripheral lymphoid organs and an increase in CD4(+) CD44(+) memory T cells. Analysis of thymocyte development revealed diminished total thymocyte numbers, accompanied by a significant increase in CD4/CD8 double-negative (DN) thymocytes due to a partial block in the transition from the DN3 to the DN4 stage. This was neither due to increased thymocyte apoptosis nor defects in the expression of the TCR-ß chain or the pre-TCR. In contrast, pERK but not pAKT levels were diminished in DN3 BDNF-deficient thymocytes. BDNF deficiency in T cells did not result in gross deficits in peripheral acute immune responses nor in changes of the homeostatic proliferation of peripheral T cells. Taken together, our data reveal a critical autocrine and/or paracrine role of T-cell-derived BDNF in thymocyte maturation involving ERK-mediated TCR signaling pathways.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/genetics , Cell Differentiation , Female , Immunologic Memory , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphopoiesis , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology
17.
Endocrinology ; 155(10): 3899-908, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25057793

ABSTRACT

Glucocorticoids (GCs) constitute a highly pleiotropic class of drugs predominantly employed in the treatment of inflammatory diseases. In our search for new mechanisms of action, we identified a hitherto unknown effect of GCs in the gastrointestinal tract. We found that oral administration of dexamethasone (Dex) to mice caused an enlargement of the stomach due to the induction of gastroparesis and that this effect was abolished in GR(dim) mice carrying the A458T mutation in the GC receptor (GR). Gastroparesis was unrelated to the enhanced gastric acid secretion observed after Dex treatment, although both effects were mediated by the same molecular mechanism of the GR. Using conditional GR-knockout mice, we could further rule out that GC effects on enterocytes or myeloid cells were involved in the induction of gastroparesis. In contrast, we found that Dex upregulated arginase 2 (Arg2) in the stomach both at the mRNA and protein level. This suggests that GC treatment leads to a depletion of l-arginine thereby impeding the production of nitric oxide (NO), which is required for gastric motility. We tested this hypothesis by supplementing the drinking water of the mice with exogenous l-arginine to compensate for the presumed shortage of this major substrate of NO synthases. Importantly, this measure completely prevented both the enlargement of the stomach and the induction of gastroparesis after Dex treatment. Our findings raise considerations of combining orally applied GCs with l-arginine to improve tolerability of GC treatment and provide a possible explanation for the antiemetic effects of GCs widely exploited in chemotherapy.


Subject(s)
Arginine/deficiency , Dexamethasone/adverse effects , Gastroparesis/chemically induced , Glucocorticoids/adverse effects , Animals , Arginase/genetics , Arginase/metabolism , Dexamethasone/administration & dosage , Female , Gastroparesis/genetics , Gastroparesis/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Glucocorticoids/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Up-Regulation/drug effects
18.
Acta Neuropathol ; 127(5): 713-29, 2014 May.
Article in English | MEDLINE | ID: mdl-24488308

ABSTRACT

Glucocorticoids (GCs) are the standard therapy for treating multiple sclerosis (MS) patients suffering from an acute relapse. One of the main mechanisms of GC action is held to be the induction of T cell apoptosis leading to reduced lymphocyte infiltration into the CNS, yet our analysis of experimental autoimmune encephalomyelitis (EAE) in three different strains of genetically manipulated mice has revealed that the induction of T cell apoptosis is not essential for the therapeutic efficacy of GCs. Instead, we identified the redirection of T cell migration in response to chemokines as a new therapeutic principle of GC action. GCs inhibited the migration of T cells towards CCL19 while they enhanced their responsiveness towards CXCL12. Importantly, blocking CXCR4 signaling in vivo by applying Plerixafor(®) strongly impaired the capacity of GCs to interfere with EAE, as revealed by an aggravated disease course, more pronounced CNS infiltration and a more dispersed distribution of the infiltrating T cells throughout the parenchyma. Our observation that T cells lacking the GC receptor were refractory to CXCL12 further underscores the importance of this pathway for the treatment of EAE by GCs. Importantly, methylprednisolone pulse therapy strongly increased the capacity of peripheral blood T cells from MS patients of different subtypes to migrate towards CXCL12. This indicates that modulation of T cell migration is an important mechanistic principle responsible for the efficacy of high-dose GC therapy not only of EAE but also of MS.


Subject(s)
Chemokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Glucocorticoids/therapeutic use , Multiple Sclerosis/drug therapy , Neuroprotective Agents/therapeutic use , T-Lymphocytes/drug effects , Adult , Aged , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Movement/drug effects , Cell Movement/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Multiple Sclerosis/physiopathology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology , T-Lymphocytes/physiology
19.
J Neuroimmunol ; 260(1-2): 17-27, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23664330

ABSTRACT

The role of CD8⁺ T cells in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) is still unclear. We describe here significantly reduced disease activity of EAE both in Lewis rats depleted of CD8⁺ T cells by monoclonal antibodies and CD8 knockout rats, which was accompanied by reduced leukocyte infiltration into the spinal cord. We detected myelin basic protein (MBP)-specific CD8⁺ T cells in peripheral lymphoid organs of CD8-depleted animals which, however, failed to differentiate into interferon-γ-producing effector cells. Our results indicate that CD8⁺ T cells interact with myelin-specific CD8⁺ T cells early in EAE enabling them to differentiate into pathogenic effector cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Leukocyte Reduction Procedures , Animals , CD8-Positive T-Lymphocytes/cytology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Flow Cytometry , Immunization , Interferon-gamma/immunology , Male , Multiple Sclerosis/immunology , Multiple Sclerosis/prevention & control , Multiple Sclerosis/therapy , Myelin Basic Protein/immunology , Rats , Rats, Inbred Lew , Rats, Mutant Strains , Spinal Cord/cytology , Spinal Cord/immunology
20.
PLoS One ; 8(4): e61034, 2013.
Article in English | MEDLINE | ID: mdl-23577186

ABSTRACT

Hedgehog (Hh) signaling modulates T cell development and function but its exact role remains a matter of debate. To further address this issue we made use of conditional knock-out mice in which the Hh receptor Patched1 (Ptch) is inactivated in the T cell lineage. Thymocyte development was moderately compromised by the deletion of Ptch as characterized by reduced numbers of CD4 and CD8 single-positive cells. In contrast, peripheral T cells were not affected. Proliferation and IFNγ secretion by Ptch-deficient T cells were indistinguishable from controls irrespectively of whether we used strong or suboptimal conditions for stimulation. Analysis of CTL and Treg cell functions did not reveal any differences between both genotypes, and T cell apoptosis induced by glucocorticoids or γ-irradiation was also similar. Surprisingly, absence of Ptch did not lead to an activation of canonic Hh signaling in peripheral T cells as indicated by unaltered expression levels of Gli1 and Gli2. To test whether we could uncover any role of Ptch in T cells in vivo we subjected the mutant mice to three different disease models, namely allogeneic bone marrow transplantation mimicking graft-versus-host disease, allergic airway inflammation as a model of asthma and growth of adoptively transferred melanoma cells as a means to test tumor surveillance by the immune system. Nonetheless, we were neither able to demonstrate any difference in the disease courses nor in any pathogenic parameter in these three models of adaptive immunity. We therefore conclude that the Hh receptor Ptch is dispensable for T cell function in vitro as well as in vivo.


Subject(s)
Adaptive Immunity , Hedgehog Proteins/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Lineage , Female , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...