Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Med Cannabis Cannabinoids ; 7(1): 59-67, 2024.
Article in English | MEDLINE | ID: mdl-38655402

ABSTRACT

Introduction: Tetrahydrocannabinol (THC) is often prescribed for ambulatory palliative patients to improve sleep quality and appetite and to reduce anxiety, stress, and pain. However, it is not known if THC has also an effect on the mortality of these patients. Method: The objective was the impact of THC on mortality of ambulatory palliative patients. For this purpose, data from the palliative treatment documentation from 5 ambulatory palliative care teams in Brandenburg, Germany were used for this analysis. Survival time was calculated for 3 groups of patients: (1) without THC; (2) with THC in a low dosage (≤4.7 mg per day); and (3) THC in higher doses (≥4.7 mg per day). The analysis was done for 2 cohorts of patients. Cohort 1: all patients with a survival time of at least 7 days after inclusion in specialized ambulatory palliative care (SAPC) and cohort 2: a subgroup of patients with a survival time between 7 and 100 days. Kaplan-Meier curves were created, and multivariate analysis was done to investigate the impact of THC on mortality. Results: A total of 9,419 patients with a survival time of at least 7 days after inclusion in SAPC were included in the analysis (cohort 1). 7,085 among them had a survival time between 7 and 100 days (cohort 2). In both cohorts, survival time was significantly prolonged by THC, but only when the daily THC dose was above the median of 4.7 mg. Survival time was 15 days longer in cohort 2 (40 vs. 25 days), when more than 4.7 mg THC were prescribed per day. Conclusion: Use of THC is associated with a significant increase in survival time in ambulatory palliative patients which survive longer than 7 days the initiation of THC prescription and which use of THC >4.7 mg/day.

2.
Dtsch Arztebl Int ; 120(49): 833-840, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-37874128

ABSTRACT

BACKGROUND: Cannabinoid drugs containing tetrahydrocannabinol (THC), or its structural analogues, as monotherapeutic agents or as extracts or botanical preparations with or without cannabidiol (CBD) are often prescribed to multimorbid patients who are taking multiple drugs. This raises the question of the risk of drug interactions. METHODS: This review of the pharmacokinetics and pharmacodynamics of interactions with cannabinoid drugs and their potential effects is based on pertinent publications retrieved by a selective literature search. RESULTS: As THC and CBD are largely metabolized in the liver, their bioavailability after oral or oral-mucosal administration is low (6-8% and 11-13%, respectively). The plasma concentrations of THC and its active metabolite 11-OH-THC can be increased by strong CYP3A4 inhibitors (verapamil, clarithromycin) and decreased by strong CYP3A4 inductors (rifampicin, carbamazepine). The clinical significance of these effects is unclear because of the variable plasma level and therapeutic spectrum of THC. The metabolism of CBD is less dependent on cytochrome P450 enzymes than that of THC. THC and CBD inhibit CYP2C and CYP3A4; the corresponding clinically relevant drug interactions probably are likely to arise only with THC doses above 30 mg/day and CBD doses above 300 mg/day. CONCLUSION: Potential drug interactions with THC and CBD are probably of little importance at low or moderate doses. Strong CYP inhibitors or inductors can intensify or weaken their effect. Slowly ramping up the dose of oral cannabinoid drugs can lessen their pharmacodynamic interactions, which can generally be well controlled. Administration by inhalation can worsen the interactions.


Subject(s)
Cannabidiol , Cannabinoids , Humans , Cannabidiol/pharmacokinetics , Dronabinol/pharmacology , Pharmaceutical Preparations , Cytochrome P-450 CYP3A , Drug Interactions
3.
J Mol Med (Berl) ; 99(8): 1073-1083, 2021 08.
Article in English | MEDLINE | ID: mdl-33864097

ABSTRACT

The present study elucidates the neuroprotective mechanisms of the PPARγ (peroxisome proliferator-activated receptor γ) agonist pioglitazone in survival of ischemic neurons following middle cerebral artery occlusion with reperfusion (MCAO). Intracerebroventricular infusion of pioglitazone over 5 days before and 24 or 48 h after MCAO alleviated neurological impairments, inhibited apoptosis 24 h, and activated the PI3K/Akt pathway along with increased phosphorylation of Akt (ser473) and GSK-3ß (ser9) in the peri-infarct cortical areas 48 h after MCAO. In primary cortical neurons, pioglitazone suppressed the glutamate-induced release of lactate dehydrogenase by a PPARγ-dependent mechanism. This protective effect was reversed after co-treatment with PI3K and Akt inhibitors, LY294002 and SH-6, respectively. Pioglitazone enhanced the expression of the antioxidative transcription factor Nrf2 and its target gene protein, heme oxidase-1, in the peri-infarct area. Pioglitazone also increased activation of the antioxidant response element (ARE) in neuronal PC12 cells transfected with the pNQO1-rARE plasmid. We demonstrate in primary cortical neurons from Nrf2 knockout mice that the lack of Nrf2 completely abolished the neuroprotective effects of pioglitazone against oxidative and excitotoxic damage. Our results strongly suggest that the neuroprotective effects of PPARγ in peri-infarct brain tissues comprise the concomitant activation of the PI3K/Akt and Nrf2/ARE pathways. KEY MESSAGES: Pioglitazone inhibits apoptosis in ischemic brain tissue.  Pioglitazone acting on PPARγ activates PI3K/Akt pathway in ischemic brain tissue. Pioglitazone activates via Nrf2 the antioxidant defense pathway in injured neurons. Pioglitazone activates the antioxidant response element in neuronal PC12 cells. Pioglitazone fails to protect primary neurons lacking Nrf2 against oxidative damage. Activation of PPARγ supports the survival of viable neurons in peri-infarct regions.


Subject(s)
Antioxidants/pharmacology , Brain Ischemia/metabolism , Neuroprotective Agents/pharmacology , Pioglitazone/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Biomarkers , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Brain Ischemia/pathology , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Disease Susceptibility , Gene Expression , Male , NF-E2-Related Factor 2/metabolism , Neuroprotection/drug effects , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats
5.
Eur J Pain ; 25(8): 1739-1750, 2021 09.
Article in English | MEDLINE | ID: mdl-33909330

ABSTRACT

BACKGROUND: Spironolactone (SPL) is a reversible mineralocorticoid receptor (MR) and androgen receptor (AR) antagonist which attracts pharmacotherapeutic interest not only because of its beneficial effects in heart failure but also because of the pathogenetic roles of MR and AR activities in neuropsychiatric diseases. Recently, beneficial and rapid-onset effects of SPL have been documented in a case series of women with fibromyalgia syndrome (FMS). To reaffirm this observation, we performed a double-blind placebo-controlled randomized clinical trial (RCT). METHODS: A total of 69 patients were screened, 56 patients were eligible and randomized to SPL or placebo (each n = 28). Forty-three patients completed the clinical trial to the last visit (n = 21 and n = 22). After a run-in phase of 50 and 100 mg/day, 200 mg/day SPL or placebo were applied between days 7 and 28. Primary outcome was the change in the FIQ-G score (Fibromyalgia Impact Questionnaire, German version). Secondary outcome parameters were the changes in pain (numeric rating scale, NRS), mood (ADS), quality of life (SF-36) and change in FIQ scores 14 days after the end of the medication. RESULTS: SPL of 200 mg/day did not change significantly either the primary or the secondary end points. SPL evoked a transient rise in serum potassium and a transient fall in GFR maximal after 2 weeks, but without clinical relevance. CONCLUSIONS: SPL at 200 mg/day does not improve symptoms in women with FMS, but was considered not to cause harm. SIGNIFICANCE: The mineralocorticoid receptor and androgen receptor antagonist spironolactone is repeatedly tested for its therapeutic effectivity against neuropsychiatric disorders. The present RCT demonstrated that 200 mg spironolactone does not change the symptoms of the fibromyalgia syndrome (FMS) in adult women. Between 2 and 4 weeks, spironolactone evokes a transient decrease in GFR and increase in serum potassium. Spironolactone cannot be recommended for the treatment of FMS.


Subject(s)
Fibromyalgia , Spironolactone , Adult , Double-Blind Method , Female , Fibromyalgia/drug therapy , Humans , Pain , Spironolactone/therapeutic use , Treatment Outcome
6.
Sci Rep ; 11(1): 7264, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33790350

ABSTRACT

During Central Nervous System ontogenesis, myelinating oligodendrocytes (OLs) arise from highly ramified and proliferative precursors called oligodendrocyte progenitor cells (OPCs). OPC architecture, proliferation and oligodendro-/myelino-genesis are finely regulated by the interplay of cell-intrinsic and extrinsic factors. A variety of extrinsic cues converge on the extracellular signal-regulated kinase/mitogen activated protein kinase (ERK/MAPK) pathway. Here we found that the germinal ablation of the MAPK c-Jun N-Terminal Kinase isoform 1 (JNK1) results in a significant reduction of myelin in the cerebral cortex and corpus callosum at both postnatal and adult stages. Myelin alterations are accompanied by higher OPC density and proliferation during the first weeks of life, consistent with a transient alteration of mechanisms regulating OPC self-renewal and differentiation. JNK1 KO OPCs also show smaller occupancy territories and a less complex branching architecture in vivo. Notably, these latter phenotypes are recapitulated in pure cultures of JNK1 KO OPCs and of WT OPCs treated with the JNK inhibitor D-JNKI-1. Moreover, JNK1 KO and WT D-JNKI-1 treated OLs, while not showing overt alterations of differentiation in vitro, display a reduced surface compared to controls. Our results unveil a novel player in the complex regulation of OPC biology, on the one hand showing that JNK1 ablation cell-autonomously determines alterations of OPC proliferation and branching architecture and, on the other hand, suggesting that JNK1 signaling in OLs participates in myelination in vivo.


Subject(s)
Cell Proliferation , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 8/metabolism , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/enzymology , Oligodendroglia/enzymology , Animals , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Myelin Sheath/genetics
7.
Sci Rep ; 10(1): 8152, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32424229

ABSTRACT

IL-33, an IL-1 cytokine superfamily member, induces the activation of the canonical NF-κB signaling, and of Mitogen Activated Protein Kinases (MAPKs). In dendritic cells (DCs) IL-33 induces the production of IL-6, IL-13 and TNFα. Thereby, the production of IL-6 depends on RelA whereas the production of IL-13 depends on the p38-MK2/3 signaling module. Here, we show that in addition to p65 and the p38-MK2/3 signaling module, JNK1/2 are essential for the IL-33-induced TNFα production. The central roles of JNK1/2 and p38 in DCs are underpinned by the fact that these two MAPK pathways are controlled by activated ß-adrenergic receptors resulting in a selective regulation of the IL-33-induced TNFα response in DCs.


Subject(s)
Dendritic Cells/metabolism , Interleukin-33/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Receptors, Adrenergic, beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Interleukin-33/genetics , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Receptors, Adrenergic, beta/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
8.
Eur J Pharmacol ; 880: 173161, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32416188

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) are associated with the development of painful neuropathies and may further aggravate sensory neuropathy produced by HIV-1 infection, leading to discontinuation of NRTI therapy by HIV patients. Following antiretroviral-induced peripheral neuropathy, c-Jun N-terminal kinase (JNK) is activated in the dorsal root ganglia (DRG) and spinal cord. However, the contribution of individual JNK genes remains unknown. Here, we have tested the behavioural mechanical sensitivity of JNK1, JNK2 and JNK3 knockout (KO) mice in the von Frey test after treatment with 2',3'-dideoxycytidine (ddC). Protein expression was investigated in the spinal cord of wild type (wt) and KO mice by western blotting. The onset of neuropathic pain was prevented by the deletion of JNK3, leading us to hypothesize that JNK3 protein plays a major role in the regulation of pain threshold in antiretroviral neuropathy. The growth-associated protein 43 (GAP-43) and the transcription factor c-Jun are involved in regeneration processes. This study revealed an up-regulation of GAP-43 and c-Jun protein, 14 days after ddC administration. JNK1 deletion induced a significant reduction in c-Jun phosphorylation and GAP-43 protein contents. In contrast, there was no difference in ddC-induced reduction of hind paw intraepidermal nerve fibre density in all JNK KO mice. Overall, these findings indicate that JNK3 plays a critical role in regulating ddC neurotoxicity-induced mechanical pain hypersensitivity, while JNK1 is important for activation of c-Jun and GAP-43 as a critical pathway of a regeneration program. These data highlight the impact of individual JNK isoforms on antiretroviral neurotoxicity and neuro-regeneration processes.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Pain/enzymology , Peripheral Nervous System Diseases/enzymology , Animals , Anti-Retroviral Agents , Disease Models, Animal , Isoenzymes/genetics , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , Mice, Knockout , Pain/chemically induced , Peripheral Nervous System Diseases/chemically induced , Zalcitabine
9.
J Cell Biochem ; 120(4): 5974-5986, 2019 04.
Article in English | MEDLINE | ID: mdl-30320919

ABSTRACT

Neuroblastoma is a malignant childhood cancer arising from the embryonic sympathoadrenal lineage of the neural crest. Retinoic acid (RA) is included in the multimodal therapy of patients with high-risk neuroblastoma to eliminate minimal residual disease. However, the formation of RA-resistant cells substantially lowers 5-year overall survival rates. To examine mechanisms that lead to treatment failure, we chose human SH-SY5Y cells, which are known to tolerate incubation with RA by activating the survival kinases Akt and extracellular signal-regulated kinase 1/2. Characterization of downstream pathways showed that both kinases increased the phosphorylation of the ubiquitin ligase mouse double minute homolog 2 (Mdm2) and thereby enhanced p53 degradation. When p53 signaling was sustained by blocking complex formation with Mdm2 or enhancing c-Jun N-terminal kinase (JNK) activation, cell viability was significantly reduced. In addition, Akt-mediated phosphorylation of the cell-cycle regulator p21 stimulated complex formation with caspase-3, which also contributed to cell protection. Thus, treatment with RA augmented survival signaling and attenuated basal apoptotic pathways in SH-SY5Y cells, which increased cell viability.


Subject(s)
Cell Survival/drug effects , Neuroblastoma/metabolism , Tretinoin/pharmacology , Blotting, Western , Cell Line, Tumor , Humans , Immunoprecipitation , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Plasmids/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism
10.
Complement Med Res ; 26(1): 13-21, 2019.
Article in German | MEDLINE | ID: mdl-30476918

ABSTRACT

Hintergrund: Procain besitzt wie das strukturverwandte Cocain zentralnervöse Wirkungen unabhängig von seinem lokalanästhetischen Effekt. Procain-Infusionen werden Patienten mit psychovegetativen Störungen seit Jahrzehnten verabreicht, zumeist im Rahmen einer Neuraltherapie. Untersuchungen zur Wirksamkeit einer alleinigen Gabe von Procain im Praxisalltag stehen aus. Patienten und Methoden: Im Rahmen einer Anwendungsbeobachtung (AWB) wurde bei 56 Patienten aus 3 Arztpraxen die Wirkung von intravenösen Procain-Infusionen (1-3 Ampullen à 5 ml einer 2%igen Procain-Lösung in 250 ml Natriumchlorid pro Arztbesuch) auf das somatische und psychovegetative Befinden dokumentiert. Hierzu wurde ein validierter, 21 Fragen umfassender Befindlichkeitsskalierungsfragebogen verwendet, den die Patienten vor Beginn und nach 1, 2, 4 und 6 Monaten ausfüllten. Neben der laufenden Medikation waren keine weiteren therapeutischen Maßnahmen erlaubt. Ergebnisse: Nach 4 oder 6 Monaten zeigten 42 Patienten (75%) eine Verbesserung bei den 9 positiven Items (d.h. erfragte Eigenschaften) wie «Freude¼, «Genussfähigkeit¼ oder «Schlaf¼; 35 Patienten (62,5%) verbesserten sich bei den 12 negativen Items wie «Stressbelastung¼, «Energielosigkeit¼ oder «Ängstlichkeit¼. Diese Veränderungen waren nach 2, 4 und 6 Monaten signifikant gegenüber dem Ausgangswert. Schlussfolgerungen: Procain ruft im zentralen Nervensystem nachweislich Veränderungen in der Aktivität spezifischer Kerngebiete wie des limbischen und kortikalen Systems hervor. Die Ergebnisse dieser AWB deuten auf eine langanhaltende Verbesserung von somatischen und psychovegetativen Störungen unter der Infusion von Procain.


Subject(s)
Mental Disorders/drug therapy , Procaine/therapeutic use , Adult , Anesthetics, Local/therapeutic use , Case-Control Studies , Humans , Infusions, Intravenous , Surveys and Questionnaires , Treatment Outcome
11.
Ann Transl Med ; 6(Suppl 1): S36, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30613611
12.
Dtsch Arztebl Int ; 114(33-34): 559, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28855049
13.
Eur J Pharmacol ; 809: 1-12, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28479141

ABSTRACT

Azithromycin is a widely used macrolide antibiotic with sustained and high tissue penetration and intracellular accumulation. While short-term exposure to low-dose azithromycin is usually well tolerated, prolonged treatment can lead to unwanted neurological effects like paresthesia and hearing loss. However, the mechanism causing neurodegeneration is still unknown. Here, we show that even low therapeutically relevant azithromycin concentrations like 1µg/ml decreased cell viability by 15% and induced neurite loss of 47% after 96h in differentiated PC12 cells, which are a well-established model system for neuronal cells. When higher concentrations were used, the drug-induced effects occurred earlier and were more pronounced. Thereby, azithromycin altered tropomyosin-related kinase A (TrkA) signaling and attenuated protein kinase B (Akt) activity, which subsequently induced autophagy. Simultaneously, the antibiotic impaired lysosomal functions by blocking the autophagic flux, and this concurrence reduced cell viability. In good agreement with reversible effects observed in patients, PC12 cells could completely recover if azithromycin was removed after 24h. In addition, the detrimental effects of azithromycin were limited to differentiated cells, as confirmed in the human neuronal model cell line SH-SY5Y. Thus, azithromycin alters cell surface receptor signaling and autophagy in neuronal cells, but does not automatically induce irreversible damage when used in low concentrations and for a short time.


Subject(s)
Azithromycin/adverse effects , Cell Differentiation , Neurons/cytology , Neurons/drug effects , Animals , Autophagy/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factor/metabolism , Neurites/drug effects , Neurites/metabolism , Neurons/metabolism , PC12 Cells , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, Nerve Growth Factor/metabolism , Signal Transduction/drug effects , Time Factors
14.
Mol Cell Neurosci ; 82: 12-22, 2017 07.
Article in English | MEDLINE | ID: mdl-28414095

ABSTRACT

The c-Jun N-terminal kinases (JNKs) are important mediators of cell viability and structural integrity in postmitotic neurons, which is required for maintaining synaptic connections and neural plasticity. In the present study, we chose differentiated PC12 cells as a well-characterised neuronal model system to selectively examine the regulation of basal JNK activity by extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. We detected a complex interaction between the kinases to prevent cell death and neurite loss. Especially the appropriate level of JNK activation determined cellular survival. Basal activity of ERK1/2 attenuated the potentiation of JNK phosphorylation and thereby the induction of apoptosis. Importantly, when JNK activity was too low, cell viability and the number of neurite-bearing cells also decreased, even though the activation of ERK1/2 was enhanced. In this case, the JNK-mediated survival signals via activating transcription factor-3 (ATF3) were inhibited. Furthermore, the phosphorylation of ERK1/2 induced by the JNK inhibitor SP600125 inhibited the basal activity of Akt, which normally supported cell viability. Thus, controlling JNK activity is crucial to promote survival and neurite stability of differentiated neuronal cells.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Neurites/metabolism , Activating Transcription Factor 3/metabolism , Animals , Anthracenes/pharmacology , Cell Death/drug effects , Cell Death/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Neurites/drug effects , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats
15.
Med Monatsschr Pharm ; 40(4): 143-6, 2017 Apr.
Article in English, German | MEDLINE | ID: mdl-29952161

ABSTRACT

The antiretroviral therapy (ART) enables an almost normal life with HIV infection. Does this also hold true for sexual intercourse, i.e. safe sex without condoms? The European PARTNER-study provides evidence for a very low risk of transmission for condomless sex under optimized conditions such as early, adherent and lasting ART (<200 RNA copies/ml) as well as stable partnerships. The risk of transmission is enhanced with non-compliance or shortlasting (< 6 months) ART, increased HIV-1 RNA load and frequent partnerships.


Subject(s)
Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , HIV Infections/virology , Safe Sex , Condoms , Female , Humans , Male , Medication Adherence , Sexual Partners
16.
Expert Rev Clin Pharmacol ; 9(11): 1479-1492, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27575590

ABSTRACT

INTRODUCTION: Therapeutic efficiency of NSAID is handicapped by ongoing discussion of cardiovascular (CV) safety. Areas covered: We update meta-analyses on NSAIDs in patients with and without cardiovascular (CV) diseases and analyse the association between NSAIDs and cardiovascular events in patients with inflammation. We demonstrate the substantial influence of an indication bias and confounding, which falsely increase the CV risk. We demonstrate protective cardiovascular effects of NSAIDs due to their anti-inflammatory activity, in particular in patients with rheumatoid arthritis, osteoarthritis or inflammatory pain. Expert commentary: t-NSAIDs and Coxibes drugs resemble in their observed CV risk which, in contrast, reflects the intrinsic risk of patients with pain and inflammation. The anti-inflammatory NSAIDs reduce the risk of first myocardial infarction in patients with inflammation and elevated CRP. The extended use of NSAIDs is not associated with an increased CV risk in patients with pain and inflammation but with reduction in all-cause mortality.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cardiovascular Diseases/prevention & control , Inflammation/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Rheumatoid/drug therapy , Bias , Cardiovascular Diseases/etiology , Cardiovascular Diseases/mortality , Confounding Factors, Epidemiologic , Humans , Osteoarthritis/drug therapy , Pain/drug therapy
17.
PLoS One ; 11(6): e0157753, 2016.
Article in English | MEDLINE | ID: mdl-27326858

ABSTRACT

Pharmacovigilance contributes to health care. However, direct access to the underlying data for academic institutions and individual physicians or pharmacists is intricate, and easily employable analysis modes for everyday clinical situations are missing. This underlines the need for a tool to bring pharmacovigilance to the clinics. To address these issues, we have developed OpenVigil FDA, a novel web-based pharmacovigilance analysis tool which uses the openFDA online interface of the Food and Drug Administration (FDA) to access U.S. American and international pharmacovigilance data from the Adverse Event Reporting System (AERS). OpenVigil FDA provides disproportionality analyses to (i) identify the drug most likely evoking a new adverse event, (ii) compare two drugs concerning their safety profile, (iii) check arbitrary combinations of two drugs for unknown drug-drug interactions and (iv) enhance the relevance of results by identifying confounding factors and eliminating them using background correction. We present examples for these applications and discuss the promises and limits of pharmacovigilance, openFDA and OpenVigil FDA. OpenVigil FDA is the first public available tool to apply pharmacovigilance findings directly to real-life clinical problems. OpenVigil FDA does not require special licenses or statistical programs.


Subject(s)
Adverse Drug Reaction Reporting Systems , Data Mining , Pharmacovigilance , United States Food and Drug Administration , Access to Information , Affect/drug effects , Antiemetics/adverse effects , Drug Interactions , Drug Therapy, Combination , Epilepsy/drug therapy , Humans , Neuralgia/drug therapy , Prescription Drugs/adverse effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Risk Factors , United States
18.
Oncotarget ; 6(7): 5354-68, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25749030

ABSTRACT

Mast cell differentiation and proliferation depends on IL-3. IL-3 induces the activation of MAP-kinases and STATs and consequently induces proliferation and survival. Dysregulation of IL-3 signaling pathways also contribute to inflammation and tumorigenesis. We show here that IL-3 induces a SFK- and Ca²âº-dependent activation of the inhibitor of κB kinases 2 (IKK2) which results in mast cell proliferation and survival but does not induce IκBα-degradation and NFκB activation. Therefore we propose the term "subthreshold IKK activation".This subthreshold IKK activation also primes mast cells for enhanced responsiveness to IL-33R signaling. Consequently, co-stimulation with IL-3 and IL-33 increases IKK activation and massively enhances cytokine production induced by IL-33.We further reveal that in neoplastic mast cells expressing constitutively active Ras, subthreshold IKK activation is associated with uncontrolled proliferation. Consequently, pharmacological IKK inhibition reduces tumor growth selectively by inducing apoptosis in vivo.Together, subthreshold IKK activation is crucial to mediate the full IL-33-induced effector functions in primary mast cells and to mediate uncontrolled proliferation of neoplastic mast cells. Thus, IKK2 is a new molecularly defined target structure.


Subject(s)
Cell Transformation, Neoplastic/pathology , I-kappa B Kinase/metabolism , Interleukin-3/pharmacology , Mast Cells/pathology , NF-kappa B/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Homeodomain Proteins/physiology , Humans , I-kappa B Kinase/genetics , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred DBA , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , NF-kappa B/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Glia ; 63(6): 1083-99, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25731696

ABSTRACT

The putative protein tyrosine kinase (PTK) inhibitor tyrphostin AG126 has proven beneficial in various models of inflammatory disease. Yet molecular targets and cellular mechanisms remained enigmatic. We demonstrate here that AG126 treatment has beneficial effects in experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis. AG126 alleviates the clinical symptoms, diminishes encephalitogenic Th17 differentiation, reduces inflammatory CNS infiltration as well as microglia activation and attenuates myelin damage. We show that AG126 directly inhibits Bruton's tyrosine kinase (BTK), a PTK associated with B cell receptor and Toll-like receptor (TLR) signaling. However, BTK inhibition cannot account for the entire activity spectrum. Effects on TLR-induced proinflammatory cytokine expression in microglia involve AG126 hydrolysis and conversion of its dinitrile side chain to malononitrile (MN). Notably, while liberated MN can subsequently mediate critical AG126 features, full protection in EAE still requires delivery of intact AG126. Its anti-inflammatory potential and especially interference with TLR signaling thus rely on a dual mechanism encompassing BTK and a novel MN-sensitive target. Both principles bear great potential for the therapeutic management of disturbed innate and adaptive immune functions.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Neuroprotective Agents/pharmacology , Tyrphostins/pharmacology , Agammaglobulinaemia Tyrosine Kinase , Animals , Cells, Cultured , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Hydrolysis , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/physiology , Myeloid Differentiation Factor 88/metabolism , Neuroprotective Agents/chemistry , Nitriles/chemistry , Nitriles/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Spleen/cytology , Spleen/drug effects , Spleen/physiopathology , Th17 Cells/drug effects , Th17 Cells/pathology , Th17 Cells/physiology , Tyrphostins/chemistry
20.
Curr Eye Res ; 40(8): 853-7, 2015.
Article in English | MEDLINE | ID: mdl-25251900

ABSTRACT

PURPOSE: Oxidative stress is considered a major factor in the deterioration of retinal pigment epithelium (RPE) cells in dry age-related macular degeneration (AMD). The MAPK ERK1/2 can be activated by oxidative stress, may exert both pro- and anti-apoptotic functions, and has recently been proposed as a major factor in RPE degeneration in atrophic changes. Nrf2 is a master regulator of oxidative stress defense and ERK1/2 is an upstream activator of Nrf2. In this study, we investigate the participation of ERK1/2 in oxidative stress pathways in connection with Nrf2. METHODS: Nrf2 knock-out and wild-type primary RPE cells were prepared from mouse eyes. Oxidative stress was induced by different concentrations of t-butylhydroperoxide. Mitogen-activated protein kinases (MAPKs) were blocked by commercially available inhibitors (SB203580, U0126, SP600125). Cell viability was determined by MTT assay. ERK1/2 expression and activation were assessed by Western blotting. RESULTS: Oxidative stress induced concentration dependent cell death, which occurred at lower concentrations in Nrf2 knock-out RPE. Western blot analysis displayed a biphasic activation of ERK1/2 in murine wild-type RPE and the inhibition of late, but not early activation of ERK1/2 exerted protection in wild-type murine RPE cells. The biphasic activation of ERK1/2 is lost in Nrf2 knock-out mice, and inhibition of ERK1/2 was generally protective. The inhibition of MAPK JNK or p38 exerted no protection, irrespective of Nrf2. CONCLUSION: RPE cells display a biphasic activation of ERK1/2 after oxidative insult, of which the late activation is pro-apoptotic. The biphasic activation is lost in Nrf2 knock-outs, suggesting that early ERK1/2 activation may be connected to Nrf2 signaling. In addition, ERK1/2 activation in Nrf2 knock-outs mediates oxidative stress-induced cell death.


Subject(s)
MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidative Stress , Retinal Pigment Epithelium/enzymology , Animals , Apoptosis , Blotting, Western , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Mice, Knockout , NF-E2-Related Factor 2/physiology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , tert-Butylhydroperoxide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...