Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Eur J Neurol ; 31(4): e16204, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38240416

ABSTRACT

BACKGROUND AND PURPOSE: In 2016, we concluded a randomized controlled trial testing 1 mg rasagiline per day add-on to standard therapy in 252 amyotrophic lateral sclerosis (ALS) patients. This article aims at better characterizing ALS patients who could possibly benefit from rasagiline by reporting new subgroup analysis and genetic data. METHODS: We performed further exploratory in-depth analyses of the study population and investigated the relevance of single nucleotide polymorphisms (SNPs) related to the dopaminergic system. RESULTS: Placebo-treated patients with very slow disease progression (loss of Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised [ALSFRS-R] per month before randomization of ≤0.328 points) showed a per se survival probability after 24 months of 0.85 (95% confidence interval = 0.65-0.94). The large group of intermediate to fast progressing ALS patients showed a prolonged survival in the rasagiline group compared to placebo after 6 and 12 months (p = 0.02, p = 0.04), and a reduced decline of ALSFRS-R after 18 months (p = 0.049). SNP genotypes in the MAOB gene and DRD2 gene did not show clear associations with rasagiline treatment effects. CONCLUSIONS: These results underline the need to consider individual disease progression at baseline in future ALS studies. Very slow disease progressors compromise the statistical power of studies with treatment durations of 12-18 months using clinical endpoints. Analysis of MAOB and DRD2 SNPs revealed no clear relationship to any outcome parameter. More insights are expected from future studies elucidating whether patients with DRD2CC genotype (Rs2283265) show a pronounced benefit from treatment with rasagiline, pointing to the opportunities precision medicine could open up for ALS patients in the future.


Subject(s)
Amyotrophic Lateral Sclerosis , Humans , Amyotrophic Lateral Sclerosis/complications , Indans/therapeutic use , Disease Progression
2.
J Oral Rehabil ; 48(9): 1044-1049, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34185922

ABSTRACT

BACKGROUND: Dysphagia as a sequel and possible early sign of amyotrophic lateral sclerosis (ALS) is caused by progressive impaired bulbar motor function. OBJECTIVE: To evaluate bulbar motor dysfunction in patients suffering from ALS compared to a healthy reference group. METHODS: A clinical study and prospective group comparison was designed. Patients and healthy volunteers were examined in the outpatient clinic of our university medical center. Ten patients with ALS and 20 healthy volunteers were included. All participants underwent a flexible endoscopic evaluation of swallowing (FEES) and a manometric measurement of the maximal sub-palatal atmospheric pressure generated by suction as well as of the prevalent pressure during swallowing. Additionally, the Sydney Swallow Questionnaire (SSQ) was completed by all participants to score the self-rated extent of dysphagia. RESULTS: Comparing maximal suction pressures, the group of patients showed significantly lower values (p < .001). There was a significant correlation between reduced pressures and the degree of dysphagia (SSQ score) (r = -0.73). CONCLUSIONS: As the oral cavity is an easily accessible compartment of the upper digestive tract, manometric measurements might serve as a simple instrument in order to detect or to monitor bulbar motor dysfunction. Oral manometry may facilitate early detection and monitoring of dysphagia in ALS. Larger studies are required to confirm our findings.


Subject(s)
Amyotrophic Lateral Sclerosis , Deglutition Disorders , Amyotrophic Lateral Sclerosis/complications , Deglutition , Deglutition Disorders/diagnosis , Deglutition Disorders/etiology , Humans , Prospective Studies
3.
Lancet Neurol ; 17(8): 681-688, 2018 08.
Article in English | MEDLINE | ID: mdl-29934198

ABSTRACT

BACKGROUND: Rasagiline, a monoamine oxidase B inhibitor with neuroprotective potential in Parkinson's disease, has shown a disease-modifying effect in the SOD1-Gly93Ala low-expressing mouse model of amyotrophic lateral sclerosis, both alone and in combination with riluzole. We sought to test whether or not rasagiline 1 mg/day can prolong survival in patients with amyotrophic lateral sclerosis also receiving riluzole. METHODS: Patients with possible, probable, or definite amyotrophic lateral sclerosis were enrolled to our randomised, placebo-controlled, parallel-group, double-blind, phase 2 trial from 15 German network for motor neuron diseases (MND-NET) centres (university hospitals or clinics). Eligible patients were aged at least 18 years, had onset of progressive weakness within the 36 months before the study, had disease duration of more than 6 months and less than 3 years, and had a best-sitting slow vital capacity of at least 50%. After a 4-week screening period, eligible patients were randomly assigned (1:1) to receive either rasagiline (1 mg/day) or placebo in addition to riluzole (100 mg/day), after stratification for site of onset (bulbar or spinal) and study centre. Patients and all personnel assessing outcome parameters were masked to treatment allocation. Patients were followed up 2, 6, 12, and 18 months after randomisation. The primary endpoint was survival time, defined as the time to death or time to study cutoff date (ie, the last patient's last visit plus 14 days). Analyses of primary outcome and safety measures were done in all patients who received at least one dose of trial treatment (intention-to-treat population). The trial is registered with ClinicalTrials.gov, number NCT01879241. FINDINGS: Between July 2, 2013, and Nov 11, 2014, 273 patients were screened for eligibility, and 252 patients were randomly assigned to receive rasagiline (n=127) or placebo (n=125). 126 patients taking rasagiline and 125 taking placebo were included in the intention-to-treat analysis. For the primary outcome, the survival probability at the end of the study was 0·43 (95% CI 0·25-0·59) in the rasagiline group (n=126) and 0·53 (0·43-0·62) in the placebo group (n=125). The estimated effect size (hazard ratio) was 0·91 (one-sided 97·5% CI -infinity to 1·34; p=0·31). Rasagiline was well tolerated, and most adverse events were due to amyotrophic lateral sclerosis disease progression rather than treatment; the most frequent of these were dysphagia (32 [25%] taking rasagiline vs 24 [19%] taking placebo) and respiratory failure (25 [20%] vs 31 [25%]). Frequency of adverse events were comparable between both groups. INTERPRETATION: Rasagiline was safe in patients with amyotrophic lateral sclerosis. There was no difference between groups in the primary outcome of survival, although post-hoc analysis suggested that rasagiline might modify disease progression in patients with an initial slope of Amyotrophic Lateral Sclerosis Functional Rating Scale Revised greater than 0·5 points per month at baseline. This should be confirmed in another clinical trial. FUNDING: Teva Pharmaceutical Industries.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Indans/therapeutic use , Neuroprotective Agents/therapeutic use , Riluzole/therapeutic use , Aged , Body Mass Index , Disease Progression , Double-Blind Method , Female , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Vital Capacity/drug effects
4.
Acta Neuropathol ; 130(5): 619-31, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26439832

ABSTRACT

Pathological tau aggregation leads to filamentous tau inclusions and characterizes neurodegenerative tauopathies such as Alzheimer's disease and frontotemporal dementia and parkinsonism linked to chromosome 17. Tau aggregation coincides with clinical symptoms and is thought to mediate neurodegeneration. Transgenic mice overexpressing mutant human P301S tau exhibit many neuropathological features of human tauopathies including behavioral deficits and increased mortality. Here, we show that the di-phenyl-pyrazole anle138b binds to aggregated tau and inhibits tau aggregation in vitro and in vivo. Furthermore, anle138b treatment effectively ameliorates disease symptoms, increases survival time and improves cognition of tau transgenic PS19 mice. In addition, we found decreased synapse and neuron loss accompanied by a decreased gliosis in the hippocampus. Our results suggest that reducing tau aggregates with anle138b may represent an effective and promising approach for the treatment of human tauopathies.


Subject(s)
Benzodioxoles/pharmacology , Neuroprotective Agents/pharmacology , Pyrazoles/pharmacology , Tauopathies/drug therapy , tau Proteins/metabolism , Animals , Cell Death/drug effects , Cell Death/physiology , Disease Models, Animal , Disease Progression , Female , Gliosis/drug therapy , Gliosis/pathology , Gliosis/physiopathology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Protein Aggregates/drug effects , Random Allocation , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Tauopathies/pathology , tau Proteins/genetics
5.
PLoS One ; 7(8): e43963, 2012.
Article in English | MEDLINE | ID: mdl-22952827

ABSTRACT

Mutations in SOD1 cause hereditary variants of the fatal motor neuron disease amyotrophic lateral sclerosis (ALS). Pathophysiology of the disease is non-cell-autonomous, with toxicity deriving also from glia. In particular, microglia contribute to disease progression. Methylene blue (MB) inhibits the effect of nitric oxide, which mediates microglial responses to injury. In vivo 2P-LSM imaging was performed in ALS-linked transgenic SOD1(G93A) mice to investigate the effect of MB on microglia-mediated inflammation in the spinal cord. Local superfusion of the lateral spinal cord with MB inhibited the microglial reaction directed at a laser-induced axon transection in control and SOD1(G93A) mice. In vitro, MB at high concentrations inhibited cytokine and chemokine release from microglia of control and advanced clinical SOD1(G93A) mice. Systemic MB-treatment of SOD1(G93A) mice at early preclinical stages significantly delayed disease onset and motor dysfunction. However, an increase of MB dose had no additional effect on disease progression; this was unexpected in view of the local anti-inflammatory effects. Furthermore, in vivo imaging of systemically MB-treated mice also showed no alterations of microglia activity in response to local lesions. Thus although systemic MB treatment had no effect on microgliosis, instead, its use revealed an important influence on motor neuron survival as indicated by an increased number of lumbar anterior horn neurons present at the time of disease onset. Thus, potentially beneficial effects of locally applied MB on inflammatory events contributing to disease progression could not be reproduced in SOD1(G93A) mice via systemic administration, whereas systemic MB application delayed disease onset via neuroprotection.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Methylene Blue/pharmacology , Microglia/drug effects , Microglia/pathology , Motor Neurons/drug effects , Motor Neurons/pathology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Animals, Genetically Modified , Cell Survival/drug effects , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Microglia/metabolism , Motor Activity/drug effects , Superoxide Dismutase-1 , Time Factors
6.
Brain ; 135(Pt 6): 1914-25, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22427327

ABSTRACT

Recent proof-of-principle data showed that the haematopoietic growth factor granulocyte colony-stimulating factor (filgrastim) mediates neuroprotection in rodent models of Parkinson's disease. In preparation for future clinical trials, we performed a preclinical characterization of a pegylated derivative of granulocyte colony-stimulating factor (pegfilgrastim) in the mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson's disease. We determined serum and cerebrospinal fluid drug levels after subcutaneous injection. A single injection of pegfilgrastim was shown to achieve stable levels of granulocyte colony-stimulating factor in both serum and cerebrospinal fluid with substantially higher levels compared to repetitive filgrastim injections. Leucocyte blood counts were only transiently increased after repeated injections. We demonstrated substantial dose-dependent long-term neuroprotection by pegfilgrastim in both young and aged mice, using bodyweight-adjusted doses that are applicable in clinical settings. Importantly, we found evidence for the functionally relevant preservation of nigrostriatal projections by pegfilgrastim in our model of Parkinson's disease, which resulted in improved motor performance. The more stable levels of pegylated neuroprotective proteins in serum and cerebrospinal fluid may represent a general advantage in the treatment of chronic neurodegenerative diseases and the resulting longer injection intervals are likely to improve patient compliance. In summary, we found that pegylation of a neuroprotective growth factor improved its pharmacokinetic profile over its non-modified counterpart in an in vivo model of Parkinson's disease. As the clinical safety profile of pegfilgrastim is already established, these data suggest that evaluation of pegfilgrastim in further Parkinson's disease models and ultimately clinical feasibility studies are warranted.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Polyethylene Glycols/therapeutic use , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Chromatography, High Pressure Liquid , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/metabolism , Homovanillic Acid/metabolism , Injections, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Parkinson Disease/blood , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/etiology , Polyethylene Glycols/metabolism , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Rotarod Performance Test , Time Factors , Tyrosine 3-Monooxygenase/metabolism
7.
BMC Neurosci ; 10: 49, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19442279

ABSTRACT

BACKGROUND: The hematopoietic Granulocyte-Colony Stimulating Factor (G-CSF) plays a crucial role in controlling the number of neutrophil progenitor cells. Its function is mediated via the G-CSF receptor, which was recently found to be expressed also in the central nervous system. In addition, G-CSF provided neuroprotection in models of neuronal cell death. Here we used the retinal ganglion cell (RGC) axotomy model to compare effects of local and systemic application of neuroprotective molecules. RESULTS: We found that the G-CSF receptor is robustly expressed by RGCs in vivo and in vitro. We thus evaluated G-CSF as a neuroprotectant for RGCs and found a dose-dependent neuroprotective effect of G-CSF on axotomized RGCs when given subcutaneously. As stem stell mobilization had previously been discussed as a possible contributor to the neuroprotective effects of G-CSF, we compared the local treatment of RGCs by injection of G-CSF into the vitreous body with systemic delivery by subcutaneous application. Both routes of application reduced retinal ganglion cell death to a comparable extent. Moreover, G-CSF enhanced the survival of immunopurified RGCs in vitro. CONCLUSION: We thus show that G-CSF neuroprotection is at least partially independent of potential systemic effects and provide further evidence that the clinically applicable G-CSF could become a treatment option for both neurodegenerative diseases and glaucoma.


Subject(s)
Granulocyte Colony-Stimulating Factor/physiology , Nerve Degeneration/drug therapy , Neuroprotective Agents/administration & dosage , Optic Nerve Injuries/drug therapy , Retinal Ganglion Cells/drug effects , Analysis of Variance , Animals , Apoptosis/drug effects , Axotomy , Dose-Response Relationship, Drug , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Injections, Subcutaneous , Rats , Rats, Sprague-Dawley , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Retinal Ganglion Cells/metabolism , Statistics, Nonparametric , Vitreous Body/drug effects
8.
J Neurochem ; 97(3): 675-86, 2006 May.
Article in English | MEDLINE | ID: mdl-16573658

ABSTRACT

We have recently shown that the hematopoietic Granulocyte-Colony Stimulating Factor (G-CSF) is neuroprotective in rodent stroke models, and that this action appears to be mediated via a neuronal G-CSF receptor. Here, we report that the G-CSF receptor is expressed in rodent dopaminergic substantia nigra neurons, suggesting that G-CSF might be neuroprotective for dopaminergic neurons and a candidate molecule for the treatment of Parkinson's disease. Thus, we investigated protective effects of G-CSF in 1-methyl-4-phenylpyridinium (MPP+)-challenged PC12 cells and primary neuronal midbrain cultures, as well as in the mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson's disease. Substantial protection was found against MPP+-induced dopaminergic cell death in vitro. Moreover, subcutaneous application of G-CSF at a dose of 40 microg/Kg body weight daily over 13 days rescued dopaminergic substantia nigra neurons from MPTP-induced death in aged mice, as shown by quantification of tyrosine hydroxylase-positive substantia nigra cells. Using HPLC, a corresponding reduction in striatal dopamine depletion after MPTP application was observed in G-CSF-treated mice. Thus our data suggest that G-CSF is a novel therapeutic opportunity for the treatment of Parkinson's disease, because it is well-tolerated and already approved for the treatment of neutropenic conditions in humans.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/prevention & control , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Blotting, Northern/methods , Brain/cytology , Brain/drug effects , Brain/metabolism , Cell Count/methods , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Disease Models, Animal , Dopamine/metabolism , Embryo, Mammalian , Gene Expression/drug effects , Gene Expression/physiology , Green Fluorescent Proteins , Homovanillic Acid/metabolism , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Parkinsonian Disorders/chemically induced , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transfection/methods , Tyrosine 3-Monooxygenase/metabolism
9.
Am J Pathol ; 162(1): 37-45, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12507888

ABSTRACT

Therapies for liver diseases with stem and progenitor cells will require a detailed knowledge of the molecular mechanisms driving the in vivo differentiation process toward adult hepatic tissue. We applied quantitative gene expression methods to analyze the differentiation process of fetal liver progenitor cells after transplantation into an animal model of liver regeneration. Enhanced green fluorescent protein (EGFP)-transgenic liver progenitor cells were isolated from fetal mouse liver at stage embryonic day 13.5 and transplanted into uPA/RAG-2 mice. Two, 4, and 6 weeks after cell transplantation cryosections of liver tissue were analyzed for EGFP-positive regeneration nodules. RNA from laser-microdissected EGFP-positive tissue was isolated and used as template for quantitative real-time reverse transcriptase-polymerase chain reaction. Phenotypic differentiation was analyzed by staining of the canalicular marker enzyme dipeptidyl-peptidase IV. Proliferation in regenerative nodules and surrounding tissue was monitored with the BrdU incorporation assay. Alpha fetoprotein gene expression had already decreased 2 weeks after transplantation in EGFP-positive regeneration nodules compared to pretransplantation values and was not detectable after 4 and 6 weeks, whereas albumin slightly increased in transplanted cells indicating differentiation into a mature phenotype. The dipeptidyl-peptidase IV antigen was associated with some liver progenitor cells 2 weeks after transplantation and in virtually all cells after 4 and 6 weeks. Cell proliferation index in transplanted cells was maximally increased (4.8% BrdU-positive cells) after 2 weeks and decreased (0.4%) after 6 weeks to normal levels. Our results demonstrate that gene expression in liver progenitor cells changes from fetal to adult phenotype within 4 to 6 weeks after transplantation despite ongoing proliferation of the transplanted cells in a mouse model of liver regeneration. Quantitative gene expression profiles as shown here will have important implications in our understanding of the in vivo differentiation process of stem cells.


Subject(s)
Gene Expression Profiling , Hepatocytes/cytology , Liver/cytology , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Division , Crosses, Genetic , DNA-Binding Proteins/genetics , Flow Cytometry , Genes, Reporter , Liver/embryology , Liver/growth & development , Liver Regeneration/physiology , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Urokinase-Type Plasminogen Activator/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...