Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Sensors (Basel) ; 24(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39000864

ABSTRACT

Quartz-Enhanced Photoacoustic Spectroscopy (QEPAS) is a technique in which the sound wave is detected by a quartz tuning fork (QTF). It enables particularly high specificity with respect to the excitation frequency and is well known for an extraordinarily sensitive analysis of gaseous samples. We have developed the first photoacoustic (PA) cell for QEPAS on solid samples. Periodic heating of the sample is excited by modulated light from an interband cascade laser (ICL) in the infrared region. The cell represents a half-open cylinder that exhibits an acoustical resonance frequency equal to that of the QTF and, therefore, additionally amplifies the PA signal. The antinode of the sound pressure of the first longitudinal overtone can be accessed by the sound detector. A 3D finite element (FE) simulation confirms the optimal dimensions of the new cylindrical cell with the given QTF resonance frequency. An experimental verification is performed with an ultrasound micro-electromechanical system (MEMS) microphone. The presented frequency-dependent QEPAS measurement exhibits a low noise signal with a high-quality factor. The QEPAS-based investigation of three different solid synthetics resulted in a linearly dependent signal with respect to the absorption.

2.
Brain Behav Immun ; 108: 148-161, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36427809

ABSTRACT

Stress-associated somatic and psychiatric disorders are often linked to non-resolving low-grade inflammation, which is promoted at least in part by glucocorticoid (GC) resistance of distinct immune cell subpopulations. While the monocyte/macrophage compartment was in the focus of many clinical and preclinical studies, the role of myeloid-derived suppressor cells (MDSCs) in stress-associated pathologies and GC resistance is less understood. As GC resistance is a clear risk factor for posttraumatic complications in patients on intensive care, the exact interplay of physical and psychosocial traumatization in the development of GC resistance needs to be further clarified. In the current study we employ the chronic subordinate colony housing (CSC) paradigm, a well-characterized mouse model of chronic psychosocial stress, to study the role of myeloid cells, in particular of MDSCs, in innate immune activation and GC resistance following combined psychosocial and physical (e.g., bite wounds) trauma. Our findings support the hypothesis that stress-induced neutrophils, polymorphonuclear (PMN)-MDSCs and monocytes/monocyte-like (MO)-MDSCs get primed and activated locally in the bone marrow as determined by toll-like receptor (TLR)2 upregulation and increased basal and lipopolysaccharide (LPS)-induced in vitro cell viability. These primed and activated myeloid cells emigrate into the peripheral circulation and subsequently, if CSC is accompanied by significant bite wounding, accumulate in the spleen. Here, PMN-MDSCs and monocytes/MO-MDSCs upregulate TLR4 expression, which exclusively in PMN-MDSCs promotes NF-κB hyperactivation upon LPS-stimulation, thereby exceeding the anti-inflammatory capacities of GCs and resulting in GC resistance.


Subject(s)
Glucocorticoids , Myeloid-Derived Suppressor Cells , Stress, Psychological , Animals , Mice , Glucocorticoids/pharmacology , Lipopolysaccharides , Monocytes , Myeloid Cells , Myeloid-Derived Suppressor Cells/metabolism
3.
EMBO J ; 37(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29875132

ABSTRACT

Astrocytes are involved in non-cell-autonomous pathogenic cascades in amyotrophic lateral sclerosis (ALS); however, their role is still debated. We show that astrocytic NF-κB activation drives microglial proliferation and leukocyte infiltration in the SOD1 (G93A) ALS model. This response prolongs the presymptomatic phase, delaying muscle denervation and decreasing disease burden, but turns detrimental in the symptomatic phase, accelerating disease progression. The transition corresponds to a shift in the microglial phenotype showing two effects that can be dissociated by temporally controlling NF-κB activation. While NF-κB activation in astrocytes induced a Wnt-dependent microglial proliferation in the presymptomatic phase with neuroprotective effects on motoneurons, in later stage, astrocyte NF-κB-dependent microglial activation caused an accelerated disease progression. Notably, suppression of the early microglial response by CB2R agonists had acute detrimental effects. These data identify astrocytes as important regulators of microglia expansion and immune response. Therefore, stage-dependent microglia modulation may be an effective therapeutic strategy in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Astrocytes/immunology , NF-kappa B/immunology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/therapy , Animals , Astrocytes/pathology , Disease Models, Animal , Mice , Mice, Transgenic , Microglia/immunology , Microglia/pathology , Motor Neurons/immunology , Motor Neurons/pathology , NF-kappa B/genetics , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/immunology , Superoxide Dismutase/genetics , Superoxide Dismutase/immunology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/immunology
4.
Cell Mol Life Sci ; 78(6): 2987-3003, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33206203

ABSTRACT

The pathogenesis of obesity-related metabolic diseases has been linked to the inflammation of white adipose tissue (WAT), but the molecular interconnections are still not fully understood. MiR-146a controls inflammatory processes by suppressing pro-inflammatory signaling pathways. The aim of this study was to characterize the role of miR-146a in obesity and insulin resistance. MiR-146a-/- mice were subjected to a high-fat diet followed by metabolic tests and WAT transcriptomics. Gain- and loss-of-function studies were performed using human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes. Compared to controls, miR-146a-/- mice gained significantly more body weight on a high-fat diet with increased fat mass and adipocyte hypertrophy. This was accompanied by exacerbated liver steatosis, insulin resistance, and glucose intolerance. Likewise, adipocytes transfected with an inhibitor of miR-146a displayed a decrease in insulin-stimulated glucose uptake, while transfecting miR-146a mimics caused the opposite effect. Natriuretic peptide receptor 3 (NPR3) was identified as a direct target gene of miR-146a in adipocytes and CRISPR/Cas9-mediated knockout of NPR3 increased insulin-stimulated glucose uptake and enhanced de novo lipogenesis. In summary, miR-146a regulates systemic and adipocyte insulin sensitivity via downregulation of NPR3.


Subject(s)
Insulin Resistance , MicroRNAs/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Adipocytes/cytology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Antagomirs/metabolism , Body Weight , Diet, High-Fat , Fatty Liver/pathology , Glucose Tolerance Test , Humans , Insulin Resistance/genetics , Lipogenesis , Liver/metabolism , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Triglycerides/metabolism
5.
Proc Natl Acad Sci U S A ; 116(3): 970-975, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30591564

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a critical regulator of cell death and inflammation, but its relevance for human disease pathogenesis remains elusive. Studies of monogenic disorders might provide critical insights into disease mechanisms and therapeutic targeting of RIPK1 for common diseases. Here, we report on eight patients from six unrelated pedigrees with biallelic loss-of-function mutations in RIPK1 presenting with primary immunodeficiency and/or intestinal inflammation. Mutations in RIPK1 were associated with reduced NF-κB activity, defective differentiation of T and B cells, increased inflammasome activity, and impaired response to TNFR1-mediated cell death in intestinal epithelial cells. The characterization of RIPK1-deficient patients highlights the essential role of RIPK1 in controlling human immune and intestinal homeostasis, and might have critical implications for therapies targeting RIPK1.


Subject(s)
Cell Differentiation , Immunity, Mucosal/genetics , Inflammatory Bowel Diseases , Intestinal Mucosa , Receptor-Interacting Protein Serine-Threonine Kinases , Severe Combined Immunodeficiency , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , HCT116 Cells , HEK293 Cells , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mutation , NF-kappa B/genetics , NF-kappa B/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
6.
FASEB J ; 32(4): 1916-1932, 2018 04.
Article in English | MEDLINE | ID: mdl-29187362

ABSTRACT

Traumatic brain injury (TBI) is the leading cause of death in young adults. After the initial injury, a poorly understood secondary phase, including a strong inflammatory response determines the final outcome of TBI. The inhibitor of NF-κB kinase (IKK)/NF-κB signaling system is the key regulator of inflammation and also critically involved in regulation of neuronal survival and synaptic plasticity. We addressed the neuron-specific function of IKK2/NF-κB signaling pathway in TBI using an experimental model of closed-head injury (CHI) in combination with mouse models allowing conditional regulation of IKK/NF-κB signaling in excitatory forebrain neurons. We found that repression of IKK2/NF-κB signaling in neurons increases the acute posttraumatic mortality rate, worsens the neurological outcome, and promotes neuronal cell death by apoptosis, thus resulting in enhanced proinflammatory gene expression. As a potential mechanism, we identified elevated levels of the proapoptotic mediators Bax and Bad and enhanced expression of stress response genes. This phenotype is also observed when neuronal IKK/NF-κB activity is inhibited just before CHI. In contrast, neuron-specific activation of IKK/NF-κB signaling does not alter the TBI outcome. Thus, this study demonstrates that physiological neuronal IKK/NF-κB signaling is necessary and sufficient to protect neurons from trauma consequences.-Mettang, M., Reichel, S. N., Lattke, M., Palmer, A., Abaei, A., Rasche, V., Huber-Lang, M., Baumann, B., Wirth, T. IKK2/NF-κB signaling protects neurons after traumatic brain injury.


Subject(s)
Brain Injuries, Traumatic/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Neurons/metabolism , Animals , Apoptosis , Mice , Signal Transduction
7.
Brain Behav Immun ; 81: 228-246, 2019 10.
Article in English | MEDLINE | ID: mdl-31207335

ABSTRACT

Traumatic brain injury (TBI) and ethanol intoxication (EI) frequently coincide, particularly in young subjects. However, the mechanisms of their interaction remain poorly understood. Among other pathogenic pathways, TBI induces glial activation and neuroinflammation in the hippocampus, resulting in acute and chronic hippocampal dysfunction. In this regard, we investigated the role of EI affecting these responses unfolding after TBI. We used a blunt, weight-drop approach to model TBI in mice. Male mice were pre-administered with ethanol or vehicle to simulate EI. The neuroinflammatory response in the hippocampus was assessed by monitoring the expression levels of >20 cytokines, the phosphorylation status of transcription factors and the phenotype of microglia and astrocytes. We used AS1517499, a brain-permeable STAT6 inhibitor, to elucidate the role of this pathway in the EI/TBI interaction. We showed that TBI causes the elevation of IL-33, IL-1ß, IL-38, TNF-α, IFN-α, IL-19 in the hippocampus at 3 h time point and concomitant EI results in the dose-dependent downregulation of IL-33, IL-1ß, IL-38, TNF-α and IL-19 (but not of IFN-α) and in the selective upregulation of IL-13 and IL-12. EI is associated with the phosphorylation of STAT6 and the transcription of STAT6-controlled genes. Moreover, ethanol-induced STAT6 phosphorylation and transcriptional activation can be recapitulated in vitro by concomitant exposure of neurons to ethanol, depolarization and inflammatory stimuli (simulating the acute trauma). Acute STAT6 inhibition prevents the effects of EI on IL-33 and TNF-α, but not on IL-13 and negates acute EI beneficial effects on TBI-associated neurological impairment. Additionally, EI is associated with reduced microglial activation and astrogliosis as well as preserved synaptic density and baseline neuronal activity 7 days after TBI and all these effects are prevented by acute administration of the STAT6 inhibitor concomitant to EI. EI concomitant to TBI exerts significant immunomodulatory effects on cytokine induction and microglial activation, largely through the activation of STAT6 pathway, ultimately with beneficial outcomes.


Subject(s)
Brain Injuries, Traumatic/metabolism , Ethanol/pharmacology , STAT6 Transcription Factor/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Cytokines/metabolism , Disease Models, Animal , Macrophage Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microglia/metabolism , Microglia/pathology , Neuroimmunomodulation/drug effects , Neurons/metabolism , Neurons/pathology , STAT6 Transcription Factor/immunology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
8.
Sensors (Basel) ; 19(13)2019 Jun 29.
Article in English | MEDLINE | ID: mdl-31261919

ABSTRACT

T-cell resonators have been used lately for non-invasive blood glucose measurements for photoacoustic spectroscopy on skin samples. A resonator has a significant role in determining the strength of the measured signal and the overall sensitivity of the sensor. Here we present results of the measurement of the photoacoustic signal of such a T-cell resonator. The signal is also modelled using the amplitude mode expansion method, which is based on eigenmode expansion and the introduction of losses in the form of loss factors. The measurement reproduced almost all the calculated resonances from the numerical models with fairly good agreement. The cause of the differences between the measured and the simulated resonances are explained. In addition, the amplitude mode expansion simulation model is established as a faster and computationally less demanding photoacoustic simulation alternative to the viscothermal model. The resonance frequencies from the two models differ by less than 1.8%. It is noted that the relative height of the amplitudes from the two models depends on the location of the antinodes within the different parts of the resonator. The amplitude mode expansion model provides a quick simulation tool for the optimization and design of macro resonators.


Subject(s)
Biosensing Techniques , Glucose/isolation & purification , Photoacoustic Techniques , T-Lymphocytes/metabolism , Computer Simulation , Glucose/metabolism , Humans , Insulin Infusion Systems , T-Lymphocytes/chemistry
9.
Int J Cancer ; 138(7): 1709-18, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26519239

ABSTRACT

Cancer stem cells (CSC) have been postulated to be responsible for the key features of a malignancy and its maintenances, as well as therapy resistance, while differentiated cells are believed to make up the rapidly growing tumour bulk. It is therefore important to understand the characteristics of those two distinct cell populations in order to devise treatment strategies which effectively target both cohorts, in particular with respect to cancers, such as glioblastoma. Glioblastoma is the most common primary brain tumour in adults, with a mean patient survival of 12-15 months. Importantly, therapeutic improvements have not been forthcoming in the last decade. In this study we compare key features of three pairs of glioblastoma cell populations, each pair consisting of stem cell-like and differentiated cells derived from an individual patient. Our data suggest that while growth rates and expression of key survival- and apoptosis-mediating proteins are more similar according to differentiation status than genetic similarity, we found no intrinsic differences in response to standard therapeutic interventions, namely exposure to radiation or the alkylating agent temozolomide. Interestingly, we could demonstrate that both stem cell-like and differentiated cells possess the ability to form stem cell-containing tumours in immunocompromised mice and that differentiated cells could potentially be dedifferentiated to potential stem cells. Taken together our data suggest that the differences between tumour stem cell and differentiated cell are particular fluent in glioblastoma.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Animals , Blotting, Western , Cell Differentiation , DNA Fragmentation , Heterografts , Humans , Mice , Tumor Cells, Cultured
10.
N Engl J Med ; 369(26): 2504-14, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24369075

ABSTRACT

BACKGROUND: Severe combined immunodeficiency (SCID) comprises a heterogeneous group of heritable deficiencies of humoral and cell-mediated immunity. Many patients with SCID have lymphocyte-activation defects that remain uncharacterized. METHODS: We performed genetic studies in four patients, from four families of Northern Cree ancestry, who had clinical characteristics of SCID, including early onset of severe viral, bacterial, and fungal infections despite normal B-cell and T-cell counts. Genomewide homozygosity mapping was used to identify a candidate region, which was found on chromosome 8; all genes within this interval were sequenced. Immune-cell populations, signal transduction on activation, and effector functions were studied. RESULTS: The patients had hypogammaglobulinemia or agammaglobulinemia, and their peripheral-blood B cells and T cells were almost exclusively of naive phenotype. Regulatory T cells and γδ T cells were absent. All patients carried a homozygous duplication--c.1292dupG in exon 13 of IKBKB, which encodes IκB kinase 2 (IKK2, also known as IKKß)--leading to loss of expression of IKK2, a component of the IKK-nuclear factor κB (NF-κB) pathway. Immune cells from the patients had impaired responses to stimulation through T-cell receptors, B-cell receptors, toll-like receptors, inflammatory cytokine receptors, and mitogens. CONCLUSIONS: A form of human SCID is characterized by normal lymphocyte development despite a loss of IKK2 function. IKK2 deficiency results in an impaired response to activation stimuli in a variety of immune cells, leading to clinically relevant impairment of adaptive and innate immunity. Although Ikk2 deficiency is lethal in mouse embryos, our observations suggest a more restricted, unique role of IKK2-NF-κB signaling in humans. (Funded by the German Federal Ministry of Education and Research and others.).


Subject(s)
Agammaglobulinemia/genetics , I-kappa B Kinase/genetics , Mutation , Severe Combined Immunodeficiency/genetics , Adaptive Immunity/genetics , B-Lymphocytes/physiology , Fatal Outcome , Female , Genes, Recessive , Humans , I-kappa B Kinase/deficiency , Immunity, Innate/genetics , Indians, North American , Infant , Infant, Newborn , Lymphocyte Activation , Lymphocyte Count , Male , Pedigree , Sequence Analysis, DNA , T-Lymphocytes/physiology
11.
FASEB J ; 29(7): 2843-58, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25829510

ABSTRACT

Redox imbalance is believed to contribute to the development and progression of several neurodegenerative disorders. Our aim was to develop an animal model that exhibits neuron-specific oxidative stress in the CNS to study the consequences and eventually find clues regarding the pathomechanisms of oxidative insults in neuronal homeostasis. We therefore generated a novel neuron-specific superoxide dismutase 2 (SOD2)-deficient mouse by deleting exon 3 of the SOD2 gene using CamKIIα promoter-driven Cre expression. These neuron-specific SOD2 knockout (SOD2(nko)) mice, although born at normal frequencies, died at the age of 4 weeks with critical growth retardation, severe energy failure, and several neurologic phenotypes. In addition, SOD2(nko) mice exhibited severe neuronal alterations such as reactive astrogliosis, neuronal cell cycle inhibition, and induction of apoptosis. JNK activation and stabilization of p53, as a result of reactive oxygen species accumulation, are most likely the inducers of neuronal apoptosis in SOD2(nko) mice. It is remarkable that hypothalamic regulation of glucose metabolism was affected, which in turn induced necrotic brain lesions in SOD2(nko) mice. Taken together, our findings suggest that exclusive deficiency of SOD2 in neurons results in an impaired central regulation of energy homeostasis that leads to persistent hypoglycemia, hypoglycemia-related neuropathology, and an early lethality of the mutant mice.


Subject(s)
Neurons/metabolism , Superoxide Dismutase/deficiency , Animals , Apoptosis , Brain/metabolism , Brain/pathology , Cells, Cultured , Disease Models, Animal , Energy Metabolism , Female , Homeostasis , Hypoglycemia/metabolism , Hypoglycemia/pathology , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/pathology , Oxidation-Reduction , Signal Transduction , Superoxide Dismutase/genetics
12.
EMBO J ; 30(1): 43-56, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21102556

ABSTRACT

The evolutionarily conserved Notch signal transduction pathway regulates fundamental cellular processes during embryonic development and in the adult. Ligand binding induces presenilin-dependent cleavage of the receptor and a subsequent nuclear translocation of the Notch intracellular domain (NICD). In the nucleus, NICD binds to the recombination signal sequence-binding protein J (RBP-J)/CBF-1 transcription factor to induce expression of Notch target genes. Here, we report the identification and functional characterization of RBP-J interacting and tubulin associated (RITA) (C12ORF52) as a novel RBP-J/CBF-1-interacting protein. RITA is a highly conserved 36 kDa protein that, most interestingly, binds to tubulin in the cytoplasm and shuttles rapidly between cytoplasm and nucleus. This shuttling RITA exports RBP-J/CBF-1 from the nucleus. Functionally, we show that RITA can reverse a Notch-induced loss of primary neurogenesis in Xenopus laevis. Furthermore, RITA is able to downregulate Notch-mediated transcription. Thus, we propose that RITA acts as a negative modulator of the Notch signalling pathway, controlling the level of nuclear RBP-J/CBF-1, where its amounts are limiting.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Microtubule-Associated Proteins/metabolism , Receptors, Notch/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/metabolism , Active Transport, Cell Nucleus , Animals , Centrosome/ultrastructure , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Gene Expression , HeLa Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/genetics , Neurogenesis , Protein Binding , Protein Transport , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Notch/genetics , Transcription, Genetic , Tubulin/metabolism , Xenopus Proteins/analysis , Xenopus Proteins/genetics , Xenopus laevis/genetics
13.
Nucleic Acids Res ; 40(1): 181-95, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21908405

ABSTRACT

NF-κB is involved in immune responses, inflammation, oncogenesis, cell proliferation and apoptosis. Even though NF-κB can be activated by DNA damage via Ataxia telangiectasia-mutated (ATM) signalling, little was known about an involvement in DNA repair. In this work, we dissected distinct DNA double-strand break (DSB) repair mechanisms revealing a stimulatory role of NF-κB in homologous recombination (HR). This effect was independent of chromatin context, cell cycle distribution or cross-talk with p53. It was not mediated by the transcriptional NF-κB targets Bcl2, BAX or Ku70, known for their dual roles in apoptosis and DSB repair. A contribution by Bcl-xL was abrogated when caspases were inhibited. Notably, HR induction by NF-κB required the targets ATM and BRCA2. Additionally, we provide evidence that NF-κB interacts with CtIP-BRCA1 complexes and promotes BRCA1 stabilization, and thereby contributes to HR induction. Immunofluorescence analysis revealed accelerated formation of replication protein A (RPA) and Rad51 foci upon NF-κB activation indicating HR stimulation through DSB resection by the interacting CtIP-BRCA1 complex and Rad51 filament formation. Taken together, these results define multiple NF-κB-dependent mechanisms regulating HR induction, and thereby providing a novel intriguing explanation for both NF-κB-mediated resistance to chemo- and radiotherapies as well as for the sensitization by pharmaceutical intervention of NF-κB activation.


Subject(s)
BRCA1 Protein/metabolism , Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Antineoplastic Agents/toxicity , Apoptosis , Cell Line, Tumor , DNA Damage , Endodeoxyribonucleases , Homologous Recombination , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Replication Protein A/analysis , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
14.
Sci Rep ; 14(1): 2441, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38286816

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of mortality and disability worldwide. Acute neuroinflammation is a prominent reaction after TBI and is mostly initiated by brain-resident glial cells such as microglia, NG2-glia and astrocytes. The magnitude of this reaction paves the way for long-lasting consequences such as chronic neurological pathologies, for which therapeutic options remain limited. The neuroinflammatory response to TBI is mostly studied with craniotomy-based animal models that are very robust but also rather artificial. Here, we aimed to analyze the reaction of glial cells in a highly translational but variable closed head injury (CHI) model and were able to correlate the severity of the trauma to the degree of glial response. Furthermore, we could show that the different glial cell types react in a temporally and spatially orchestrated manner in terms of morphological changes, proliferation, and cell numbers in the first 15 days after the lesion. Interestingly, NG2-glia, the only proliferating cells in the healthy brain parenchyma, divided at a rate that was correlated with the size of the injury. Our findings describe the previously uncharacterized posttraumatic response of the major brain glial cell types in CHI in order to gain a detailed understanding of the course of neuroinflammatory events; such knowledge may open novel avenues for future therapeutic approaches in TBI.


Subject(s)
Brain Injuries, Traumatic , Head Injuries, Closed , Animals , Neuroglia/metabolism , Brain/metabolism , Brain Injuries, Traumatic/pathology , Astrocytes/metabolism , Microglia/metabolism , Head Injuries, Closed/pathology , Disease Models, Animal
15.
J Neurosci ; 32(34): 11511-23, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915098

ABSTRACT

Hydrocephalus formation is a frequent complication of neuropathological insults associated with neuroinflammation. However, the mechanistic role of neuroinflammation in hydrocephalus development is unclear. We have investigated the function of the proinflammatory acting inhibitor of κB kinase (IKK)/nuclear factor κB (NF-κB) signaling system in neuroinflammatory processes and generated a novel mouse model that allows conditional activation of the IKK/NF-κB system in astrocytes. Remarkably, NF-κB activation in astrocytes during early postnatal life results in hydrocephalus formation and additional defects in brain development. NF-κB activation causes global neuroinflammation characterized by a strong, astrocyte-specific expression of proinflammatory NF-κB target genes as well as a massive infiltration and activation of macrophages. In this animal model, hydrocephalus formation is specifically induced during a critical time period of early postnatal development, in which IKK/NF-κB-induced neuroinflammation interferes with ependymal ciliogenesis. Our findings demonstrate for the first time that IKK/NF-κB activation is sufficient to induce hydrocephalus formation and provides a potential mechanistic explanation for the frequent association of neuroinflammation and hydrocephalus formation during brain development, namely impairment of ependymal cilia formation. Therefore, our study might open up new perspectives for the treatment of certain types of neonatal and childhood hydrocephalus associated with hemorrhages and infections.


Subject(s)
Encephalitis/etiology , Gene Expression Regulation, Developmental/physiology , Glioma, Subependymal/etiology , Hydrocephalus , I-kappa B Kinase/metabolism , Age Factors , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/enzymology , Brain/enzymology , Brain/growth & development , Brain/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Chemokines/genetics , Chemokines/metabolism , Complement System Proteins/genetics , Complement System Proteins/metabolism , Disease Models, Animal , Doxycycline/administration & dosage , Enzyme Activation/drug effects , Enzyme Activation/genetics , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/genetics , Glioma, Subependymal/pathology , Humans , Hydrocephalus/complications , Hydrocephalus/enzymology , Hydrocephalus/pathology , I-kappa B Kinase/genetics , I-kappa B Proteins/metabolism , Lateral Ventricles/growth & development , Lateral Ventricles/pathology , Lateral Ventricles/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Microscopy, Electron, Scanning , NF-KappaB Inhibitor alpha , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Statistics, Nonparametric , Transcription Factor RelA/metabolism , NF-kappaB-Inducing Kinase
16.
J Neurosci ; 32(16): 5688-703, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22514330

ABSTRACT

Alterations of learning and memory in mice with deregulated neuron-specific nuclear factor κB (NF-κB) activity support the idea that plastic changes of synaptic contacts may depend at least in part on IκB kinase (IKK)/NF-κB-related synapse-to-nucleus signaling. There is, however, little information on the molecular requirements and mechanisms regulating this IKK/NF-κB-dependent synapse development and remodeling. Here, we report that the NF-κB inducing IKK kinase complex is localized at the postsynaptic density (PSD) and activated under basal conditions in the adult mouse brain. Using different models of conditional genetic inactivation of IKK2 function in mouse principal neurons, we show that IKK/NF-κB signaling is critically involved in synapse formation and spine maturation in the adult brain. IKK/NF-κB blockade in the forebrain of mutant animals is associated with reduced levels of mature spines and postsynaptic proteins PSD95, SAP97, GluA1, AMPAR-mediated basal synaptic transmission and a spatial learning impairment. Synaptic deficits can be restored in adult animals within 1 week by IKK/NF-κB reactivation, indicating a highly dynamic IKK/NF-κB-dependent regulation process. We further identified the insulin-like growth factor 2 gene (Igf2) as a novel IKK/NF-κB target. Exogenous Igf2 was able to restore synapse density and promoted spine maturation in IKK/NF-κB signaling-deficient neurons within 24 h. This process depends on Igf2/Igf2R-mediated MEK/ERK activation. Our findings illustrate a fundamental role of IKK/NF-κB-Igf2-Igf2R signaling in synapse formation and maturation in adult mice, thus providing an intriguing link between the molecular actions of IKK/NF-κB in neurons and the memory enhancement factor Igf2.


Subject(s)
I-kappa B Kinase/metabolism , Insulin-Like Growth Factor II/metabolism , NF-kappa B/metabolism , Neurons/cytology , Signal Transduction/physiology , Synapses/physiology , Adaptor Proteins, Signal Transducing/metabolism , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Dendritic Spines , Discs Large Homolog 1 Protein , Disks Large Homolog 4 Protein , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Electrophoretic Mobility Shift Assay , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation, Developmental/genetics , Guanylate Kinases/metabolism , Hippocampus/cytology , I-kappa B Kinase/genetics , In Vitro Techniques , Insulin-Like Growth Factor II/genetics , Learning Disabilities/genetics , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Maze Learning/drug effects , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Patch-Clamp Techniques , Receptors, AMPA/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Silver Staining , Synapses/drug effects , Synapses/ultrastructure
17.
Hepatology ; 56(3): 1117-28, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22407857

ABSTRACT

UNLABELLED: Liver damage in humans is induced by various insults including alcohol abuse, hepatitis B/C virus infection, autoimmune or metabolic disorders and, when persistent, leads to development of liver fibrosis. Because the nuclear factor-κB (NF-κB) system is activated in response to several of these stresses, we hypothesized that NF-κB activation in hepatocytes may contribute to fibrosis development. To activate the NF-κB signaling pathway in a time- and cell-type-specific manner in the liver, we crossed transgenic mice carrying the tetracycline-responsive transactivator under the control of the liver activator protein promotor with transgenic mice carrying a constitutively active form of the Ikbkb gene (IKK2 protein [CAIKK2]). Double-transgenic mice displayed doxycycline-regulated CAIKK2 expression in hepatocytes. Removal of doxycycline at birth led to activation of NF-κB signaling, moderate liver damage, recruitment of inflammatory cells, hepatocyte proliferation, and ultimately to spontaneous liver fibrosis development. Microarray analysis revealed prominent up-regulation of chemokines and chemokine receptors and this induction was rapidly reversed after switching off the CAIKK2 expression. Turning off the transgene expression for 3 weeks reversed stellate cell activation but did not diminish liver fibrosis. The elimination of macrophages by clodronate-liposomes attenuated NF-κB-induced liver fibrosis in a liver-injury-independent manner. CONCLUSION: Our results revealed that hepatic activation of IKK/NF-κB is sufficient to induce liver fibrosis by way of macrophage-mediated chronic inflammation. Therefore, agents controlling the hepatic NF-κB system represent attractive therapeutic tools to prevent fibrosis development in multiple chronic liver diseases.


Subject(s)
I-kappa B Kinase/physiology , Inflammation/immunology , Liver Cirrhosis/immunology , Macrophages/immunology , NF-kappa B/physiology , Animals , Chronic Disease , Male , Mice , Mice, Inbred C57BL , Signal Transduction
18.
J Neural Transm (Vienna) ; 120(5): 785-98, 2013 May.
Article in English | MEDLINE | ID: mdl-23143281

ABSTRACT

The dynactin p150glued subunit, encoded by the gene DCTN1 is part of the dynein-dynactin motor protein complex responsible for retrograde axonal transport. This subunit is a candidate modifier for neurodegenerative diseases, in particular motoneuron and extrapyramidal diseases. Based on an extensive screening effort of all 32 exons in more than 2,500 ALS/MND patients, patients suffering from Parkinsonian Syndromes and controls, we investigated 24 sequence variants of p150 in cell-based studies. We used both non-neuronal cell lines and primary rodent spinal motoneurons and report on cell biological abnormalities in five of these sequence alterations and also briefly report on the clinical features. Our results suggest the presence of biological changes caused by some p150 mutants pointing to a potential pathogenetic significance as modifier of the phenotype of the human disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Microtubule-Associated Proteins/genetics , Motor Neurons/metabolism , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Adaptor Proteins, Signal Transducing , Amyotrophic Lateral Sclerosis/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagy-Related Proteins , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , Chlorocebus aethiops , Dynactin Complex , Embryo, Mammalian , Female , Green Fluorescent Proteins/genetics , Humans , Male , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Motor Neurons/pathology , Motor Neurons/ultrastructure , Mutation/genetics , Pregnancy , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Retrospective Studies , Spinal Cord/cytology , Time Factors
19.
Cell Mol Gastroenterol Hepatol ; 16(5): 783-807, 2023.
Article in English | MEDLINE | ID: mdl-37543088

ABSTRACT

BACKGROUND AND AIMS: Development of pancreatic ductal adenocarcinoma (PDAC) is a multistep process intensively studied; however, precocious diagnosis and effective therapy still remain unsatisfactory. The role for Notch signaling in PDAC has been discussed controversially, as both cancer-promoting and cancer-antagonizing functions have been described. Thus, an improved understanding of the underlying molecular mechanisms is necessary. Here, we focused on RBPJ, the receiving transcription factor in the Notch pathway, examined its expression pattern in PDAC, and characterized its function in mouse models of pancreatic cancer development and in the regeneration process after acute pancreatitis. METHODS: Conditional transgenic mouse models were used for functional analysis of RBPJ in the adult pancreas, initiation of PDAC precursor lesions, and pancreatic regeneration. Pancreata and primary acinar cells were tested for acinar-to-ductal metaplasia together with immunohistology and comprehensive transcriptional profiling by RNA sequencing. RESULTS: We identified reduced RBPJ expression in a subset of human PDAC specimens. Ptf1α-CreERT-driven depletion of RBPJ in transgenic mice revealed that its function is dispensable for the homeostasis and maintenance of adult acinar cells. However, primary RBPJ-deficient acinar cells underwent acinar-to-ductal differentiation in ex vivo. Importantly, oncogenic KRAS expression in the context of RBPJ deficiency facilitated the development of pancreatic intraepithelial neoplasia lesions with massive fibrotic stroma formation. Interestingly, RNA-sequencing data revealed a transcriptional profile associated with the cytokine/chemokine and extracellular matrix changes. In addition, lack of RBPJ delays the course of acute pancreatitis and critically impairs it in the context of KRASG12D expression. CONCLUSIONS: Our findings imply that downregulation of RBPJ in PDAC patients derepresses Notch targets and promotes KRAS-mediated pancreatic acinar cells transformation and desmoplasia development.


Subject(s)
Carcinoma in Situ , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Pancreatitis , Animals , Humans , Mice , Acinar Cells/metabolism , Acute Disease , Carcinoma in Situ/metabolism , Carcinoma, Pancreatic Ductal/pathology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice, Transgenic , Pancreatic Neoplasms/pathology , Pancreatitis/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms
20.
Aging Cell ; 22(9): e13911, 2023 09.
Article in English | MEDLINE | ID: mdl-37609868

ABSTRACT

Aging of the central nervous system (CNS) leads to motoric and cognitive decline and increases the probability for neurodegenerative disease development. Astrocytes fulfill central homeostatic functions in the CNS including regulation of immune responses and metabolic support of neurons and oligodendrocytes. In this study, we investigated the effect of redox imbalance in astrocytes by using a conditional astrocyte-specific SOD2-deficient mouse model (SOD2ako ) and analyzed these animals at different stages of their life. SOD2ako mice did not exhibit any overt phenotype within the first postnatal weeks. However, already as young adults, they displayed progressive motoric impairments. Moreover, as these mice grew older, they exhibited signs of a progeroid phenotype and early death. Histological analysis in moribund SOD2ako mice revealed the presence of age-related brain alterations, neuroinflammation, neuronal damage and myelin impairment in brain and spinal cord. Additionally, transcriptome analysis of primary astrocytes revealed that SOD2 deletion triggered a hypometabolic state and promoted polarization toward A1-neurotoxic status, possibly underlying the neuronal and myelin deficits. Conclusively, our study identifies maintenance of ROS homeostasis in astrocytes as a critical prerequisite for physiological CNS aging.


Subject(s)
Aging , Astrocytes , Neurodegenerative Diseases , Animals , Mice , Central Nervous System , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL