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










Publication year range
1.
Cells ; 13(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38727269

ABSTRACT

The histone deacetylase inhibitor (HDACi) valproic acid (VPA) has neuroprotective and anti-inflammatory effects in experimental traumatic brain injury (TBI), which have been partially attributed to the epigenetic disinhibition of the transcription repressor RE1-Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). Additionally, VPA changes post-traumatic brain injury (TBI) brain metabolism to create a neuroprotective environment. To address the interconnection of neuroprotection, metabolism, inflammation and REST/NRSF after TBI, we subjected C57BL/6N mice to experimental TBI and intraperitoneal VPA administration or vehicle solution at 15 min, 1, 2, and 3 days post-injury (dpi). At 7 dpi, TBI-induced an up-regulation of REST/NRSF gene expression and HDACi function of VPA on histone H3 acetylation were confirmed. Neurological deficits, brain lesion size, blood-brain barrier permeability, or astrogliosis were not affected, and REST/NRSF target genes were only marginally influenced by VPA. However, VPA attenuated structural damage in the hippocampus, microgliosis and expression of the pro-inflammatory marker genes. Analyses of plasma lipidomic and polar metabolomic patterns revealed that VPA treatment increased lysophosphatidylcholines (LPCs), which were inversely associated with interleukin 1 beta (Il1b) and tumor necrosis factor (Tnf) gene expression in the brain. The results show that VPA has mild neuroprotective and anti-inflammatory effects likely originating from favorable systemic metabolic changes resulting in increased plasma LPCs that are known to be actively taken up by the brain and function as carriers for neuroprotective polyunsaturated fatty acids.


Subject(s)
Brain Injuries, Traumatic , Inflammation , Lysophosphatidylcholines , Mice, Inbred C57BL , Neurons , Valproic Acid , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/complications , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Mice , Male , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Inflammation/pathology , Inflammation/drug therapy , Lysophosphatidylcholines/blood , Cell Death/drug effects , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Repressor Proteins/metabolism , Repressor Proteins/genetics
2.
BMC Neurosci ; 25(1): 10, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38424488

ABSTRACT

TBI is a leading cause of death and disability in young people and older adults worldwide. There is no gold standard treatment for TBI besides surgical interventions and symptomatic relief. Post-injury infections, such as lower respiratory tract and surgical site infections or meningitis are frequent complications following TBI. Whether the use of preventive and/or symptomatic antibiotic therapy improves patient mortality and outcome is an ongoing matter of debate. In contrast, results from animal models of TBI suggest translational perspectives and support the hypothesis that antibiotics, independent of their anti-microbial activity, alleviate secondary injury and improve neurological outcomes. These beneficial effects were largely attributed to the inhibition of neuroinflammation and neuronal cell death. In this review, we briefly outline current treatment options, including antibiotic therapy, for patients with TBI. We then summarize the therapeutic effects of the most commonly tested antibiotics in TBI animal models, highlight studies identifying molecular targets of antibiotics, and discuss similarities and differences in their mechanistic modes of action.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Neuroprotective Agents , Animals , Humans , Aged , Adolescent , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/complications , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
3.
Neuropharmacology ; 237: 109648, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37385435

ABSTRACT

The connection between dysbiosis of the gut microbiome and diseases and injuries of the brain has attracted considerable interest in recent years. Interestingly, antibiotic-induced microbial dysbiosis has been implicated in the pathogenesis of traumatic brain injury (TBI), while early administration of antibiotics associates with improved survival in TBI patients. In animal models of TBI, short- or long-term administration of antibiotics, both peri- or post-operatively, were shown to induce gut microbiome dysbiosis but also anti-inflammatory and neuroprotective effects. However, the acute consequences of microbial dysbiosis on TBI pathogenesis after discontinuation of antibiotic treatment are elusive. In this study, we tested whether pre-traumatic antibiotic-induced microbial depletion by vancomycin, amoxicillin, and clavulanic acid affects pathogenesis during the acute phase of TBI in adult male C57BL/6 mice. Pre-traumatic microbiome depletion did not affect neurological deficits over 72 h post injury (hpi) and brain histopathology, including numbers of activated astrocytes and microglia, at 72 hpi. However, astrocytes and microglia were smaller after pre-traumatic microbiome depletion compared to vehicle treatment at 72 hpi, indicating less inflammatory activation. Accordingly, TBI-induced gene expression of the inflammation markers Interleukin-1ß, complement component C3, translocator protein TSPO and the major histocompatibility complex MHC2 was attenuated in microbiome-depleted mice along with reduced Immunoglobulin G extravasation as a proxy of blood-brain barrier (BBB) impairment. These results suggest that the gut microbiome contributes to early neuroinflammatory responses to TBI but does not have a significant impact on brain histopathology and neurological deficits. This article is part of the Special Issue on "Microbiome & the Brain: Mechanisms & Maladies".


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Animals , Mice , Male , Neuroinflammatory Diseases , Anti-Bacterial Agents/pharmacology , Dysbiosis , Mice, Inbred C57BL , Brain Injuries/metabolism , Brain Injuries, Traumatic/metabolism , Inflammation/metabolism , Disease Models, Animal , Microglia
4.
Sci Rep ; 13(1): 4348, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36928073

ABSTRACT

Traumatic brain injury (TBI) causes the release of danger-associated molecular patterns (DAMP) from damaged or dead cells, which contribute to secondary brain damage after TBI. Cell-free DNA (cfDNA) is a DAMP known to cause disruption of the blood-brain barrier (BBB), promote procoagulant processes, brain edema, and neuroinflammation. This study tested the hypothesis that administration of deoxyribonuclease-I (DNase-I) has a beneficial effect after TBI. Mice (n = 84) were subjected to controlled cortical impact (CCI) and posttraumatic intraperitoneal injections of low dose (LD) or high dose (HD) of DNase-I or vehicle solution at 30 min and 12 h after CCI. LD was most effective to reduce lesion volume (p = 0.003), brain water content (p < 0.0001) and to stabilize BBB integrity (p = 0.019) 1 day post-injury (dpi). At 6 h post injury LD-treated animals showed less cleavage of fibrin (p = 0.0014), and enhanced perfusion as assessed by micro-computer-tomography (p = 0.027). At 5 dpi the number of Iba1-positive cells (p = 0.037) were reduced, but the number of CD45-positive cells, motoric function and brain lesion volume was not different. Posttraumatic-treatment with DNase-I therefore stabilizes the BBB, reduces the formation of brain edema, immune response, and delays secondary brain damage. DNase-I might be a new approach to extend the treatment window after TBI.


Subject(s)
Brain Edema , Brain Injuries, Traumatic , Deoxyribonucleases , Animals , Mice , Blood-Brain Barrier , Brain/pathology , Brain Edema/drug therapy , Brain Edema/pathology , Brain Injuries/drug therapy , Brain Injuries/pathology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Deoxyribonucleases/pharmacology , Deoxyribonucleases/therapeutic use , Disease Models, Animal , Mice, Inbred C57BL , Cell-Free Nucleic Acids/adverse effects , Cell-Free Nucleic Acids/metabolism
5.
Intensive Care Med Exp ; 10(1): 37, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36058954

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a common disease in intensive care medicine. Despite intensive research, mortality rates are high, not even in COVID-19 ARDS. Thereby, pigs offer some advantages to study the characteristics of ARDS. Many different ARDS models exist. Most of the articles published focused on histopathological and microscopic lung alterations to identify the most suitable animal ARDS model. "Macroscopic" observations and descriptions are often missing. Therefore, we performed a post-hoc comparison of two common ARDS models for pigs: lipopolysaccharide (LPS) vs. a double-hit model (bronchoalveolar lavage + oleic acid infusion). We investigated hemodynamic, spirometric and laboratory changes as another main clinical part of ARDS. RESULTS: The groups were compared by two-way analysis of variance (ANOVA) with a post-hoc Student-Newman-Keuls test. A p value lower than 0.05 was accepted as significant. All animals (n = 8 double-hit ARDS; n = 8 LPS ARDS) survived the observation period of 8 h. ARDS induction with reduced oxygen indices was successful performed in both models (76 ± 35/225 ± 54/212 ± 79 vs. 367 ± 64; T0/T4/T8 vs. BLH for double-hit; 238 ± 57/144 ± 59 vs. 509 ± 41; T4/T8 vs. BLH for LPS; p < 0.05). ARDS induced with LPS leads to more hemodynamic (mean arterial pulmonary pressure 35 ± 3/30 ± 3 vs. 28 ± 4/23 ± 4; T4/T8 LPS vs. double-hit; p < 0.05; doses of norepinephrine 1.18 ± 1.05 vs. 0.11 ± 0.16; LPS vs. double-hit for T8; p < 0.05) and inflammatory (pulmonary IL-6 expression: 2.41e-04 ± 1.08e-04 vs. 1.45e-05 ± 7.26e-06; LPS vs. double-hit; p < 0.05) alterations. ARDS induced by double-hit requires a more invasive ventilator strategy to maintain a sufficient oxygenation (PEEP at T4: 8 ± 3 vs. 6 ± 2; double-hit vs. LPS; p < 0.05). CONCLUSIONS: Both animal ARDS models are feasible and are similar to human presentation of ARDS. If your respiratory research focus on hemodynamic/inflammation variables, the LPS-induced ARDS is a feasible model. Studying different ventilator strategies, the double-hit ARDS model offers a suitable approach.

6.
Front Psychol ; 13: 903879, 2022.
Article in English | MEDLINE | ID: mdl-36059723

ABSTRACT

This paper tested the ability of Mandarin learners of German, whose native language has lexical tone, to imitate pitch accent contrasts in German, an intonation language. In intonation languages, pitch accents do not convey lexical information; also, pitch accents are sparser than lexical tones as they only associate with prominent words in the utterance. We compared two kinds of German pitch-accent contrasts: (1) a "non-merger" contrast, which Mandarin listeners perceive as different and (2) a "merger" contrast, which sounds more similar to Mandarin listeners. Speakers of a tone language are generally very sensitive to pitch. Hypothesis 1 (H1) therefore stated that Mandarin learners produce the two kinds of contrasts similarly to native German speakers. However, the documented sensitivity to tonal contrasts, at the expense of processing phrase-level intonational contrasts, may generally hinder target-like production of intonational pitch accents in the L2 (Hypothesis 2, H2). Finally, cross-linguistic influence (CLI) predicts a difference in the realization of these two contrasts as well as improvement with higher proficiency (Hypothesis 3, H3). We used a delayed imitation paradigm, which is well-suited for assessing L2-phonetics and -phonology because it does not necessitate access to intonational meaning. We investigated the imitation of three kinds of accents, which were associated with the sentence-final noun in short wh-questions (e.g., Wer malt denn Mandalas, lit: "Who draws PRT mandalas?" "Who likes drawing mandalas?"). In Experiment 1, 28 native speakers of Mandarin participated (14 low- and 14 high-proficient). The learners' productions of the two kinds of contrasts were analyzed using General Additive Mixed Models to evaluate differences in pitch accent contrasts over time, in comparison to the productions of native German participants from an earlier study in our lab. Results showed a more pronounced realization of the non-merger contrast compared to German natives and a less distinct realization of the merger contrast, with beneficial effects of proficiency, lending support to H3. Experiment 2 tested low-proficient Italian learners of German (whose L1 is an intonation language) to contextualize the Mandarin data and further investigate CLI. Italian learners realized the non-merger contrast more target-like than Mandarin learners, lending additional support to CLI (H3).

7.
Brain Behav Immun ; 106: 49-66, 2022 11.
Article in English | MEDLINE | ID: mdl-35933030

ABSTRACT

BACKGROUND: There is a need for early therapeutic interventions after traumatic brain injury (TBI) to prevent neurodegeneration. Microglia/macrophage (M/M) depletion and repopulation after treatment with colony stimulating factor 1 receptor (CSF1R) inhibitors reduces neurodegeneration. The present study investigates short- and long-term consequences after CSF1R inhibition during the early phase after TBI. METHODS: Sex-matched mice were subjected to TBI and CSF1R inhibition by PLX3397 for 5 days and sacrificed at 5 or 30 days post injury (dpi). Neurological deficits were monitored and brain tissues were examined for histo- and molecular pathological markers. RNAseq was performed with 30 dpi TBI samples. RESULTS: At 5 dpi, CSF1R inhibition attenuated the TBI-induced perilesional M/M increase and associated gene expressions by up to 50%. M/M attenuation did not affect structural brain damage at this time-point, impaired hematoma clearance, and had no effect on IL-1ß expression. At 30 dpi, following drug discontinuation at 5 dpi and M/M repopulation, CSF1R inhibition attenuated brain tissue loss regardless of sex, as well as hippocampal atrophy and thalamic neuronal loss in male mice. Selected gene markers of brain inflammation and apoptosis were reduced in males but increased in females after early CSF1R inhibition as compared to corresponding TBI vehicle groups. Neurological outcome in behaving mice was almost not affected. RNAseq and gene set enrichment analysis (GSEA) of injured brains at 30 dpi revealed more genes associated with dendritic spines and synapse function after early CSF1R inhibition as compared to vehicle, suggesting improved neuronal maintenance and recovery. In TBI vehicle mice, GSEA showed high oxidative phosphorylation, oxidoreductase activity and ribosomal biogenesis suggesting oxidative stress and increased abundance of metabolically highly active cells. More genes associated with immune processes and phagocytosis in PLX3397 treated females vs males, suggesting sex-specific differences in response to early CSF1R inhibition after TBI. CONCLUSIONS: M/M attenuation after CSF1R inhibition via PLX3397 during the early phase of TBI reduces long-term brain tissue loss, improves neuronal maintenance and fosters synapse recovery. Overall effects were not sex-specific but there is evidence that male mice benefit more than female mice.


Subject(s)
Brain Injuries, Traumatic , Macrophage Colony-Stimulating Factor , Aminopyridines , Animals , Brain Injuries, Traumatic/metabolism , Disease Models, Animal , Female , Inflammation/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Oxidoreductases/metabolism , Oxidoreductases/pharmacology , Pyrroles , Receptors, Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
8.
Ann Hematol ; 101(8): 1755-1767, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35759026

ABSTRACT

CD19-directed chimeric antigen receptor T cells (CAR-T) have emerged as a highly efficacious treatment for patients with relapsed/refractory (r/r) B cell lymphoma (BCL). The value of CAR-T for these patients is indisputable, but one-off production costs are high, and little is known about the ancillary resource consumption associated with CAR-T treatment. Here, we compared the resource use and costs of CAR-T treatment with high-dose chemotherapy followed by autologous stem cell transplantation (ASCT) for patients with r/r BCL. Standard operating procedures were used to develop a process model in ClipMedPPM, which comprises all activities and processes to sustain or generate treatment components that together constitute a treatment path. The software allows a graphic representation and the use of standardized linguistic elements for comparison of different treatment paths. Detailed processes involved in CAR-T treatments (n = 1041 processes) and in ASCT (n = 1535) were analyzed for time consumption of treatment phases and personnel. Process costs were calculated using financial controlling data. CAR-T treatment required ~ 30% less staff time than ASCT (primarily nursing staff) due to fewer chemotherapy cycles, less outpatient visits, and shorter hospital stays. For CAR-T, production costs were ~ 8 × higher, but overall treatment time was shorter compared with ASCT (30 vs 48 days), and direct labor and overhead costs were 40% and 10% lower, respectively. Excluding high product costs, CAR-T uses fewer hospital resources than ASCT for r/r BCL. Fewer hospital days for CAR-T compared to ASCT treatment and the conservation of hospital resources are beneficial to patients and the healthcare system.


Subject(s)
Hematopoietic Stem Cell Transplantation , Lymphoma, B-Cell , Receptors, Chimeric Antigen , Antigens, CD19 , Humans , Lymphoma, B-Cell/drug therapy , Receptors, Antigen, T-Cell , Transplantation, Autologous/methods
9.
Sci Rep ; 12(1): 5731, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35388024

ABSTRACT

Traumatic brain injury (TBI) involves primary mechanical damage and delayed secondary damage caused by vascular dysfunction and neuroinflammation. Intracellular components released into the parenchyma and systemic circulation, termed danger-associated molecular patterns (DAMPs), are major drivers of vascular dysfunction and neuroinflammation. These DAMPs include cell-free RNAs (cfRNAs), which damage the blood-brain barrier (BBB), thereby promoting edema, procoagulatory processes, and infiltration of inflammatory cells. We tested the hypothesis that intraperitoneal injection of Ribonuclease-1 (RNase1, two doses of 20, 60, or 180 µg/kg) at 30 min and 12 h after controlled-cortical-impact (CCI) can reduce secondary lesion expansion compared to vehicle treatment 24 h and 120 h post-CCI. The lowest total dose (40 µg/kg) was most effective at reducing lesion volume (- 31% RNase 40 µg/kg vs. vehicle), brain water accumulation (- 5.5%), and loss of BBB integrity (- 21.6%) at 24 h post-CCI. RNase1 also reduced perilesional leukocyte recruitment (- 53.3%) and microglial activation (- 18.3%) at 120 h post-CCI, but there was no difference in lesion volume at this time and no functional benefit. Treatment with RNase1 in the early phase following TBI stabilizes the BBB and impedes leukocyte immigration, thereby suppressing neuroinflammation. RNase1-treatment may be a novel approach to delay brain injury to extend the window for treatment opportunities after TBI.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Animals , Blood-Brain Barrier , Brain/pathology , Brain Injuries/pathology , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Ribonucleases/pharmacology
10.
Front Psychol ; 12: 714363, 2021.
Article in English | MEDLINE | ID: mdl-34925127

ABSTRACT

Variability is pervasive in spoken language, in particular if one is exposed to two varieties of the same language (e.g., the standard variety and a dialect). Unlike in bilingual settings, standard and dialectal forms are often phonologically related, increasing the variability in word forms (e.g., German Fuß "foot" is produced as [fus] in Standard German and as [fs] in the Alemannic dialect). We investigate whether dialectal variability in children's input affects their ability to recognize words in Standard German, testing non-dialectal vs. dialectal children. Non-dialectal children, who typically grow up in urban areas, mostly hear Standard German forms, and hence encounter little segmental variability in their input. Dialectal children in turn, who typically grow up in rural areas, hear both Standard German and dialectal forms, and are hence exposed to a large amount of variability in their input. We employ the familiar word paradigm for German children aged 12-18 months. Since dialectal children from rural areas are hard to recruit for laboratory studies, we programmed an App that allows all parents to test their children at home. Looking times to familiar vs. non-familiar words were analyzed using a semi-automatic procedure based on neural networks. Our results replicate the familiarity preference for non-dialectal German 12-18-month-old children (longer looking times to familiar words than vs. non-familiar words). Non-dialectal children in the same age range, on the other hand, showed a novelty preference. One explanation for the novelty preference in dialectal children may be more mature linguistic processing, caused by more variability of word forms in the input. This linguistic maturation hypothesis is addressed in Experiment 2, in which we tested older children (18-24-month-olds). These children, who are not exposed to dialectal forms, also showed a novelty preference. Taken together, our findings show that both dialectal and non-dialectal German children recognized the familiar Standard German word forms, but their looking pattern differed as a function of the variability in the input. Frequent exposure to both dialectal and Standard German word forms may hence have affected the nature of (prelexical and/or) lexical representations, leading to more mature processing capacities.

11.
Mediators Inflamm ; 2021: 8835730, 2021.
Article in English | MEDLINE | ID: mdl-33531878

ABSTRACT

Traumatic brain injury (TBI) represents a major cause of death and disability in early adulthood. Concomitant extracranial injury such as long bone fracture was reported to exacerbate TBI pathology. However, early reciprocal effects and mechanisms have been barely investigated. To address this issue, C57BL/6N mice were subjected to either the controlled cortical impact (CCI) model of TBI, fracture of the left femur (FF), combined injury (CCI+FF), or sham procedure. Behavioral alterations were monitored until 5 days post injury (dpi), followed by (immuno-)histology, gene and protein expression analyses using quantitative PCR, western blot, and ELISA. We found that CCI+FF mice exhibited increased neurological impairments, reduced recovery, and altered anxiety-related behavior compared to single injury groups. At 5 dpi, cerebral lesion size was not affected by combined injury but exaggerated hippocampal substance loss and increased perilesional astrogliosis were observed in CCI+FF mice compared to isolated CCI. Bone gene expression of the osteogenic markers Runx2, osteocalcin, alkaline phosphatase, and bone sialoprotein was induced by fracture injury but attenuated by concomitant TBI. Plasma concentrations of the biomarkers osteopontin and progranulin were elevated in CCI+FF mice compared to other experimental groups. Taken together, using a murine model of TBI and femoral fracture, we report early reciprocal impairments of brain tissue maintenance, behavioral recovery, and bone repair gene expression. Increased circulating levels of the biomarkers osteopontin and progranulin indicate ongoing tissue inflammation and repair. Our results may have implications for future therapeutic approaches to interfere with the pathological crosstalk between TBI and concomitant bone fracture.


Subject(s)
Analgesics/pharmacology , Brain Injuries, Traumatic/physiopathology , Femoral Fractures/physiopathology , Osteopontin/metabolism , Progranulins/metabolism , Alkaline Phosphatase/metabolism , Animals , Behavior, Animal , Biomarkers/metabolism , Brain/pathology , Brain Injuries/metabolism , Disease Models, Animal , Female , Femur , Gliosis/metabolism , Hippocampus/metabolism , Inflammation , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...