Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters











Publication year range
1.
Heliyon ; 10(18): e38070, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39381248

ABSTRACT

Excitotoxicity-induced oxidative stress results in neuronal cell death. Pink lotus essential oil (PLO) is a concentrated volatile oil from lotus blossoms widely used in traditional medicine. This study aimed to explore the possible therapeutic effects of PLO and its underlying mechanisms on kainic acid (KA)-induced oxidative stress and hippocampal cell death in a mouse model of epilepsy. Mice were treated with 100 mg/kg or 200 mg/kg PLO to ameliorate neurodegeneration and seizure-induced behavior induced by KA injection. Pre- and post-treatment of PLO increased antioxidant activities, reduced the seizure score, prevented oxidative stress by increasing GSH and CAT levels, and reduced MDA (malondialdehyde) levels after KA-induced status epilepticus. KA injection created neuronal cell death in the pyramidal layers of CA1 and CA3 subfields of the hippocampus, and affected interneurons in the hilus of the dentate gyrus. PLO treatment notably diminished KA-induced neuronal cell death in these areas through activation of the Akt signaling pathway, increasing reactive astrogliosis, and up-regulation of GDNF expression. Moreover, caspase-3 expression, and microglia activation were significantly decreased in PLO treatments. Taken together, these results suggest that PLO possesses antiepileptic, anti-apoptosic, and neuroprotective effects on KA-induced epileptogenesis indicating that PLO may serve as a dietary supplement option in the treatment of epilepsy or of other neurodegenerative disorders.

2.
Front Immunol ; 14: 1157421, 2023.
Article in English | MEDLINE | ID: mdl-36960056

ABSTRACT

Periodontal Ehlers-Danlos syndrome (pEDS) is an autosomal dominant disorder characterized by early-onset periodontitis leading to premature loss of teeth, lack of attached gingiva and thin and fragile gums leading to gingival recession. Connective tissue abnormalities of pEDS typically include easy bruising, pretibial plaques, distal joint hypermobility, hoarse voice, and less commonly manifestations such as organ or vessel rupture. pEDS is caused by heterozygous missense mutations in C1R and C1S genes of the classical complement C1 complex. Previously we showed that pEDS pathogenic variants trigger intracellular activation of C1r and/or C1s, leading to extracellular presence of activated C1s. However, the molecular link relating activated C1r and C1s proteases to the dysregulated connective tissue homeostasis in pEDS is unknown. Using cell- and molecular-biological assays, we identified activated C1s (aC1s) as an enzyme which degrades collagen I in cell culture and in in vitro assays. Matrix collagen turnover in cell culture was assessed using labelled hybridizing peptides, which revealed fast and comprehensive collagen protein remodeling in patient fibroblasts. Furthermore, collagen I was completely degraded by aC1s when assays were performed at 40°C, indicating that even moderate elevated temperature has a tremendous impact on collagen I integrity. This high turnover is expected to interfere with the formation of a stable ECM and result in tissues with loose compaction a hallmark of the EDS phenotype. Our results indicate that pathogenesis in pEDS is not solely mediated by activation of the complement cascade but by inadequate C1s-mediated degradation of matrix proteins, confirming pEDS as a primary connective tissue disorder.


Subject(s)
Complement C1s , Ehlers-Danlos Syndrome , Humans , Collagen Type I/genetics , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Mutation, Missense , Complement C1s/genetics
3.
F1000Res ; 12: 846, 2023.
Article in English | MEDLINE | ID: mdl-38434672

ABSTRACT

Background: One of the most common neurodegenerative diseases is Parkinson's disease (PD); PD is characterized by a reduction of neurons containing dopamine in the substantia nigra (SN), which leads to a lack of dopamine (DA) in nigrostriatal pathways, resulting in motor function disorders. Oxidative stress is considered as one of the etiologies involved in dopaminergic neuronal loss. Thus, we aimed to investigate the neuroprotective effects of pinostrobin (PB), a bioflavonoid extracted from Boesenbergia rotunda with antioxidative activity in PD. Methods: Rats were treated with 40 mg/kg of PB for seven consecutive days before and after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD. After completing the experiment, the brains including SN and striatum were used for histological studies and biochemical assays. Results: PB treatment demonstrated a reduction of free radicals in the SN as indicated by significantly decreased MDA levels, whereas the antioxidative enzymes (SOD and GSH) were significantly increased. Furthermore, PB treatment significantly increased glial cell line-derived neurotrophic factor (GDNF) immunolabelling which has neurotrophic and neuroprotective effects on the survival of dopaminergic neurons. Furthermore, PB treatment was shown to protect CA1 and CA3 neurons in the hippocampus and dopaminergic neurons in the SN. DA levels in the SN were increased after PB treatment, leading to the improvement of motor function of PD rats. Conclusions: These results imply that PB prevents MPTP-induced neurotoxicity via its antioxidant activities and increases GDNF levels, which may contribute to the therapeutic strategy for PD.


Subject(s)
Flavanones , Neuroprotective Agents , Parkinson Disease , Animals , Rats , Antioxidants/metabolism , Dopamine , Dopaminergic Neurons , Glial Cell Line-Derived Neurotrophic Factor , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Up-Regulation
4.
Cells ; 11(9)2022 05 04.
Article in English | MEDLINE | ID: mdl-35563843

ABSTRACT

Intracellular signal transduction in response to growth factor receptor activation is a fundamental process during the regeneration of the nervous system. In this context, intracellular inhibitors of neuronal growth factor signaling have become of great interest in the recent years. Among them are the prominent signal transduction regulators Sprouty (SPRY) and phosphatase and tensin homolog deleted on chromosome 10 (PTEN), which interfere with major signaling pathways such as extracellular signal-regulated kinase (ERK) or phosphoinositide 3-kinase (PI3K)/Akt in neurons and glial cells. Furthermore, SPRY and PTEN are themselves tightly regulated by ubiquitin ligases such as c-casitas b-lineage lymphoma (c-CBL) or neural precursor cell expressed developmentally down-regulated protein 4 (NEDD4) and by different microRNAs (miRs) including miR-21 and miR-222. SPRY, PTEN and their intracellular regulators play an important role in the developing and the lesioned adult central and peripheral nervous system. This review will focus on the effects of SPRY and PTEN as well as their regulators in various experimental models of axonal regeneration in vitro and in vivo. Targeting these signal transduction regulators in the nervous system holds great promise for the treatment of neurological injuries in the future.


Subject(s)
MicroRNAs , Phosphatidylinositol 3-Kinases , MicroRNAs/genetics , MicroRNAs/pharmacology , Nedd4 Ubiquitin Protein Ligases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism
5.
Ann Anat ; 241: 151890, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35085704

ABSTRACT

As elsewhere, the cadavers of Nazi victims were used at the 'Alpenunversität Innsbruck' for the education of medical students. They were also used by members of the Institute of Anatomy and the Institute of Histology for scientific research and publications. In 2018, over 300 drawers were discovered in a laboratory anteroom of the Innsbruck Histological Institute containing around 15,000 histological slides. After a closer examination, 237 slides were found to have human tissues from victims of the 3rd Reich possibly. These 237 slides were produced between May 1938 and March 1944. All 237 slides were digitized, the labels carefully analysed, and some of the victims were identified. Several specimens come from the tissues of three Nazi victims who were executed in Munich-Stadelheim and whose bodies were brought to the Innsbruck Anatomical Institute. From there, the organs were passed on to the Histological Institute Innsbruck. Inscriptions on other slides such as "Cl[ara]. 40", "hing[erichtet]. Clara" or "Hinger[ichtet]. Cl[ara]." prove that the specimens were most likely sent to the Institute by the histologist Max Clara. At this time, Clara was Director of the Leipzig Anatomical Institute and still had close ties to the Innsbruck Institute, where he had been trained. Based on several sources, some Nazi victims could be identified by name; biographical traces complement this identification. Under what political and sociological conditions future generations will look at the crimes of the Nazi dictatorship is not yet foreseeable. As anatomists and scientists, we must be cautious about removing evidence from this terrible time. Therefore, we will bury all slides where relatives wish to do so or where it is clear that Rabbi Polak's "Vienna Protocol" must be applied. However, the remaining slides will be kept safe for eventual further investigation.


Subject(s)
Anatomists , Anatomy , Academies and Institutes , Cadaver , Germany , Histological Techniques , History, 20th Century , Humans , Male , National Socialism
6.
J Cereb Blood Flow Metab ; 42(2): 219-236, 2022 02.
Article in English | MEDLINE | ID: mdl-34427147

ABSTRACT

Due to the limited therapeutic options after ischemic stroke, gene therapy has emerged as a promising choice, especially with recent advances in viral vector delivery systems. Therefore, we aimed to provide the current state of the art of lentivirus (LV) and adeno-associated virus (AAV) mediated gene interventions in preclinical ischemic stroke models. A systematic analysis including qualitative and quantitative syntheses of studies published until December 2020 was performed. Most of the 87 selected publications used adult male rodents and the preferred stroke model was transient middle cerebral artery occlusion. LV and AAV vectors were equally used for transgene delivery, however loads of AAVs were higher than LVs. Serotypes having broad cell tropism, the use of constitutive promoters, and virus delivery before the stroke induction via stereotaxic injection in the cortex and striatum were preferred in the analyzed studies. The meta-analysis based on infarct volume as the primary outcome confirmed the efficacy of the preclinical interventions. The quality assessment exposed publication bias and setbacks in regard to risks of bias and study relevance. The translational potential could increase by using specific cell targeting, post-stroke interventions, non-invasive systematic delivery, and use of large animals.


Subject(s)
Cerebral Cortex , Corpus Striatum , Dependovirus , Genetic Therapy , Genetic Vectors , Ischemic Stroke , Lentivirus , Animals , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Corpus Striatum/blood supply , Corpus Striatum/metabolism , Disease Models, Animal , Humans , Ischemic Stroke/genetics , Ischemic Stroke/metabolism , Ischemic Stroke/therapy
7.
Mol Neurobiol ; 58(8): 3884-3902, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33860438

ABSTRACT

Fibroblast growth factors (FGFs) act as key signalling molecules in brain development, maintenance, and repair. They influence the intricate relationship between myelinating cells and axons as well as the association of astrocytic and microglial processes with neuronal perikarya and synapses. Advances in molecular genetics and imaging techniques have allowed novel insights into FGF signalling in recent years. Conditional mouse mutants have revealed the functional significance of neuronal and glial FGF receptors, not only in tissue protection, axon regeneration, and glial proliferation but also in instant behavioural changes. This review provides a summary of recent findings regarding the role of FGFs and their receptors in the nervous system and in the pathogenesis of major neurological and psychiatric disorders.


Subject(s)
Fibroblast Growth Factors/metabolism , Nerve Regeneration/physiology , Nervous System Diseases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/physiology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Fibroblast Growth Factors/genetics , Humans , Microglia/metabolism , Microglia/pathology , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Receptors, Fibroblast Growth Factor/genetics
8.
Front Cell Neurosci ; 13: 332, 2019.
Article in English | MEDLINE | ID: mdl-31396054

ABSTRACT

Restoration of neuronal connectivity after lesion of the central nervous system, such as spinal cord injury, is one of the biggest challenges in modern medicine. In particular, the accumulation of axon growth inhibitory factors at the site of injury constitutes a major obstacle to structural and thus functional repair. We previously investigated a group of prenylflavonoids derived from hops for their capacity to promote neuroregeneration. We identified a molecule called ENDF1 that was very potent to enhance regrowth and branching of neurites from dorsal root ganglion neurons in culture on growth promoting substrates. In the present study, we investigated ENDF1's capacity to promote regeneration of rat dorsal root ganglion neurons in vitro in the presence of three main components of the extracellular matrix acting as axon growth inhibitors: Semaphorin 3A, Ephrin A4 and mixed chondroitin sulfate proteoglycans. We report that ENDF1 application significantly promoted the percentages of sensory neurons able to regrow their neurites regardless of the presence of those inhibitors, and this to an extent similar to the one obtained after NGF treatment. Moreover, ENDF1 strongly enhanced the total neurite length and the complexity of neurites extending from neurons challenged with axon growth inhibitors. Although the impact of NGF and ENDF1 on the regeneration of neurons was similar, the activity of ENDF1 was not mediated by signaling through the TrkA receptor, indicating that each molecule act through different signaling pathways. In addition, ENDF1 did not decrease the phosphorylation of cofilin, a downstream effector of the regeneration-associated RhoA/ROCK signaling pathway. Hence, ENDF1 is a potent pro-neuroregenerative factors that could help in identifying new efficient targets for regenerative therapies of the nervous system.

9.
Sci Rep ; 9(1): 10095, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31300753

ABSTRACT

While axons within the central nervous system (CNS) do not regenerate following injury, those in the peripheral nervous system (PNS) do, although not in a clinically satisfactory manner as only a small proportion of axons exhibit long-distance regeneration. Moreover, functional recovery is hampered by excessive axonal sprouting and aberrant reinnervation of target tissue. In order to investigate the mechanisms governing the regrowth of axons following injury, previous studies have used lesion paradigms of peripheral nerves in rat or mouse models, and reagents or cells have been administered to the lesion site through nerve conduits, aiming to improve early-stage regeneration. Morphological analysis of such in vivo experiments has however been limited by the incompatibility of synthetic nerve conduits with existing tissue-clearing and imaging techniques. We present herein a novel experimental approach that allows high-resolution imaging of individual axons within nerve conduits, together with quantitative assessment of fiber growth. We used a GFP-expressing mouse strain in a lesion model of the sciatic nerve to describe a strategy that combines nerve clearing, chemical treatment of chitosan nerve conduits, and long working distance confocal microscopy with image processing and analysis. This novel experimental setup provides a means of documenting axon growth within the actual conduit during the critical initial stage of regeneration. This will greatly facilitate the development and evaluation of treatment regimens to improve axonal regeneration following nerve damage.


Subject(s)
Axons/physiology , Image Processing, Computer-Assisted/methods , Nerve Regeneration/physiology , Optical Imaging/methods , Sciatic Nerve/physiology , Animals , Biocompatible Materials/chemistry , Chitosan/chemistry , Female , Mice , Microscopy, Confocal/methods , Peripheral Nerve Injuries/pathology , Prostheses and Implants , Recovery of Function/physiology , Sciatic Nerve/cytology
11.
Anat Rec (Hoboken) ; 302(8): 1261-1267, 2019 08.
Article in English | MEDLINE | ID: mdl-30951263

ABSTRACT

Peripherally projecting neurons undergo significant morphological changes during development and regeneration. This neuroplasticity is controlled by growth factors, which bind specific membrane bound kinase receptors that in turn activate two major intracellular signal transduction cascades. Besides the PI3 kinase/AKT pathway, activated extracellular signal-regulated kinase (ERK) plays a key role in regulating the mode and speed of peripheral axon outgrowth in the adult stage. Cell culture studies and animal models revealed that ERK signaling is mainly involved in elongative axon growth in vitro and long-distance nerve regeneration in vivo. Here, we review ERK dependent morphological plasticity in adult peripheral neurons and evaluate the therapeutic potential of interfering with regulators of ERK signaling to promote nerve regeneration. Anat Rec, 302:1261-1267, 2019. © 2019 Wiley Periodicals, Inc.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Nerve Regeneration , Peripheral Nerve Injuries/therapy , Peripheral Nerves/cytology , Animals , Humans , Peripheral Nerves/metabolism
12.
Anat Rec (Hoboken) ; 302(8): 1268-1275, 2019 08.
Article in English | MEDLINE | ID: mdl-30950230

ABSTRACT

In the course of peripheral nerve regeneration, axons encounter different extracellular growth factors secreted by non-neuronal cells at the injury site and retrogradely transported after binding to neuronal membrane receptor tyrosine kinases. The present study reviews the role of receptor transport in peripheral axon outgrowth and provides novel data on trafficking of fibroblast growth factor receptor type 1 (FGFR1). Differences in receptor transport are determined by different numbers of lysine residues acting as ubiquitination sites in the intracellular receptor domain. We previously demonstrated that overexpression of mutant FGFR1-25R (25 out of 29 intracellular lysines replaced with arginine) results in enhanced receptor recycling as compared to wild-type FGFR1 followed by strong stimulation of elongative axon growth in vitro. Here, the effects of lysine-deficient FGFR1 (FGFR1-29R lacking all 29 cytoplasmic lysine residues) or of only 15 lysine mutations (FGFR1-15R) on axon outgrowth and concomitant changes in signal pathway activation were investigated by immunocytochemistry and morphometry of cultured primary neurons. Overexpression of FGFR1-15R in adult sensory neurons resulted in enhanced receptor recycling, which was accompanied by increased axon elongation without stimulating axon branching. By contrast, FGFR1-29R was neither endocytosed nor axon outgrowth affected. Although overexpression of FGFR1-15R or FGFR1-25Ra strongly promoted elongation, we did not detect increased signal pathway activation (ERK, AKT, PLC, or STAT3) in neurons expressing mutant FGFR1 as compared with wild-type neurons raising the possibility that other signaling pathways or signaling independent mechanisms may be involved in the axon outgrowth effects of recycled FGF receptors. Anat Rec, 302:1268-1275, 2019. © 2019 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Subject(s)
Axons/physiology , Dopaminergic Neurons/cytology , Endocytosis , Neurogenesis , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Sensory Receptor Cells/cytology , Animals , Dopaminergic Neurons/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Lysine/chemistry , Lysine/genetics , Mutation , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, Fibroblast Growth Factor, Type 1/genetics , Sensory Receptor Cells/physiology , Signal Transduction
13.
Front Mol Neurosci ; 12: 73, 2019.
Article in English | MEDLINE | ID: mdl-30983969

ABSTRACT

Sprouty proteins act ubiquitously as signaling integrators and inhibitors of receptor tyrosine kinase (RTK) activated pathways. Among the four Sprouty isoforms, Sprouty2 is a key regulator of growth factor signaling in several neurological disorders. High protein levels correlate with reduced survival of glioma patients. We recently demonstrated that abrogating its function inhibits tumor growth by overstimulation of ERK and induction of DNA replication stress. The important role of Sprouty2 in the proliferation of malignant glioma cells prompted us to investigate its subcellular localization applying super-resolution fluorescence and immunoelectron microscopy. We found that cytoplasmic Sprouty2 is not homogenously distributed but localized to small spots (<100 nm) partly attached to vimentin filaments and co-localized with activated ERK. The protein is associated with early, late and recycling endosomes in response to but also independently of growth factor stimulation. The subcellular localization of Sprouty2 in all areas exhibiting strong RTK activities may reflect a protective response of glioma cells to limit excessive ERK activation and to prevent cellular senescence and apoptosis.

14.
Cells ; 8(3)2019 03 14.
Article in English | MEDLINE | ID: mdl-30875802

ABSTRACT

The intracellular transport of receptor tyrosine kinases results in the differential activation of various signaling pathways. In this study, optogenetic stimulation of fibroblast growth factor receptor type 1 (FGFR1) was performed to study the effects of subcellular targeting of receptor kinases on signaling and neurite outgrowth. The catalytic domain of FGFR1 fused to the algal light-oxygen-voltage-sensing (LOV) domain was directed to different cellular compartments (plasma membrane, cytoplasm and nucleus) in human embryonic kidney (HEK293) and pheochromocytoma (PC12) cells. Blue light stimulation elevated the pERK and pPLCγ1 levels in membrane-opto-FGFR1-transfected cells similarly to ligand-induced receptor activation; however, no changes in pAKT levels were observed. PC12 cells transfected with membrane-opto-FGFR1 exhibited significantly longer neurites after light stimulation than after growth factor treatment, and significantly more neurites extended from their cell bodies. The activation of cytoplasmic FGFR1 kinase enhanced ERK signaling in HEK293 cells but not in PC12 cells and did not induce neuronal differentiation. The stimulation of FGFR1 kinase in the nucleus also did not result in signaling changes or neurite outgrowth. We conclude that FGFR1 kinase needs to be associated with membranes to induce the differentiation of PC12 cells mainly via ERK activation.


Subject(s)
Cell Differentiation , Cell Membrane/metabolism , Cell Nucleus/metabolism , Neurons/cytology , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cell Differentiation/radiation effects , Cell Membrane/radiation effects , Cell Nucleus/radiation effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Ligands , Light , Neurites/metabolism , Neurites/radiation effects , Neurons/radiation effects , Optogenetics , PC12 Cells , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction
15.
Front Cell Neurosci ; 13: 583, 2019.
Article in English | MEDLINE | ID: mdl-32038175

ABSTRACT

Sprouty2 (Spry2) and phosphatase and tensin homolog deleted on chromosome 10 (PTEN) are both well-established regulators of receptor tyrosine kinase (RTK) signaling, and knockdown of Spry2 or PTEN enhances axon regeneration of dorsal root ganglia (DRG) neurons. The major role of Spry2 is the inhibition of the rat sarcoma RAS/extracellular signal-regulated kinase (ERK) pathway, whereas PTEN acts mainly as an inhibitor of the phosphoinositide 3-kinase (PI3K)/Akt pathway. In non-neuronal cells, Spry2 increases the expression and activity of PTEN, and PTEN enhances the amount of Spry2 by the inhibition of the microRNA-21 (miR-21) that downregulates Spry2. Applying dissociated DRG neuron cultures from wild-type (WT) or Spry2 deficient mice, we demonstrate that PTEN protein was reduced after 72 h during rapid axonal outgrowth on the laminin substrate. Furthermore, PTEN protein was decreased in DRG cultures obtained from homozygous Spry2-/- knockout mice. Vice versa, Spry2 protein was reduced by PTEN siRNA in WT and heterozygous Spry2+/- neurons. Knockdown of PTEN in DRG cultures obtained from homozygous Spry2-/- knockout mice promoted axon elongation without increasing axonal branching. Activation of Akt, but not ERK, was stronger in response to PTEN knockdown in homozygous Spry2-/- DRG neurons than in WT neurons. Together, our study confirms the important role of the signaling modulators Spry2 and PTEN in axon growth of adult DRG neurons. Both function as endogenous inhibitors of neuronal growth factor signaling and their simultaneous knockdown promotes axon elongation more efficiently than the single knockdown of each inhibitor. Furthermore, Spry2 and PTEN are reciprocally downregulated in adult DRG neuron cultures. Axon growth is influenced by multiple factors and our results demonstrate that the endogenous inhibitors of axon growth, Spry2 and PTEN, are co-regulated in adult DRG neuron cultures. Together, our data demonstrate that combined approaches may be more useful to improve nerve regeneration than targeting one single inhibitor of axon growth.

16.
Mol Neurobiol ; 56(6): 3897-3903, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30225774

ABSTRACT

Clinical trials applying growth factors to alleviate symptoms of patients with neurological disorders have largely been unsuccessful in the past. As an alternative approach, growth factor receptors or components of their signal transduction machinery may be targeted directly. In recent years, the search for intracellular signaling integrator downstream of receptor tyrosine kinases provided valuable novel substrates. Among them are the Sprouty proteins which mainly act as inhibitors of growth factor-dependent neuronal and glial signaling pathways. In this review, we summarize the role of Sprouties in the lesioned central and peripheral nervous system with particular reference to Sprouty2 that is upregulated in various experimental models of neuronal degeneration and regeneration. Increased synthesis under pathological conditions makes Sprouty2 an attractive pharmacological target to enhance intracellular signaling activities, notably the ERK pathway, in affected neurons or activated astrocytes. Interestingly, high Sprouty2 levels are also found in malignant glioma cells. We recently demonstrated that abrogating Sprouty2 function strongly inhibits intracranial tumor growth and leads to significantly prolonged survival of glioblastoma bearing mice by induction of ERK-dependent DNA replication stress. On the contrary, knockdown of Sprouty proteins increases proliferation of activated astrocytes and, consequently, reduces secondary brain damage in neuronal lesion models such as kainic acid-induced epilepsy or endothelin-induced ischemia. Furthermore, downregulation of Sprouty2 improves nerve regeneration in the lesioned peripheral nervous system. Taken together, targeting Sprouties as intracellular inhibitors of the ERK pathway holds great promise for the treatment of various neurological disorders including gliomas. Since the protein lacks enzymatic activities, it will be difficult to develop chemical compounds capable to directly and specifically modulate Sprouty functions. However, interfering with Sprouty expression by gene therapy or siRNA treatment provides a realistic approach to evaluate the therapeutic potential of indirectly stimulating ERK activities in neurological disease.


Subject(s)
Membrane Proteins/metabolism , Molecular Targeted Therapy , Nervous System/metabolism , Animals , Humans , Models, Biological , Nervous System/pathology , Neurons/metabolism , Neurons/pathology , Signal Transduction
17.
Biomaterials ; 176: 106-121, 2018 09.
Article in English | MEDLINE | ID: mdl-29879652

ABSTRACT

The blocking of specific protein-protein interactions using nanoparticles is an emerging alternative to small molecule-based therapeutic interventions. However, the nanoparticles designed as "artificial proteins" generally require modification of their surface with (bio)organic molecules and/or polymers to ensure their selectivity and specificity of action. Here, we show that nanosized diamond crystals (nanodiamonds, NDs) without any synthetically installed (bio)organic interface enable the specific and efficient targeting of the family of extracellular signalling molecules known as fibroblast growth factors (FGFs). We found that low nanomolar solutions of detonation NDs with positive ζ-potential strongly associate with multiple FGF ligands present at sub-nanomolar concentrations and effectively neutralize the effects of FGF signalling in cells without interfering with other growth factor systems and serum proteins unrelated to FGFs. We identified an evolutionarily conserved FGF recognition motif, ∼17 amino acids long, that contributes to the selectivity of the ND-FGF interaction. In addition, we inserted this motif into a de novo constructed chimeric protein, which significantly improved its interaction with NDs. We demonstrated that the interaction of NDs, as purely inorganic nanoparticles, with proteins can mitigate pathological FGF signalling and promote the restoration of cartilage growth in a mouse limb explant model. Based on our observations, we foresee that NDs may potentially be applied as nanotherapeutics to neutralize disease-related activities of FGFs in vivo.


Subject(s)
Fibroblast Growth Factors/metabolism , Nanodiamonds/chemistry , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Motifs , Animals , Cartilage/physiology , Cell Line , Cell Proliferation , Cell Survival , Embryo, Mammalian , Humans , Ligands , Mice , Protein Binding , Signal Transduction , Tibia/physiology , Tissue Culture Techniques
18.
Neuro Oncol ; 20(8): 1044-1054, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29635363

ABSTRACT

Background: Sprouty2 (SPRY2), a feedback regulator of receptor tyrosine kinase (RTK) signaling, has been shown to be associated with drug resistance and cell proliferation in glioblastoma (GBM), but the underlying mechanisms are still poorly defined. Methods: SPRY2 expression and survival patterns of patients with gliomas were analyzed using publicly available databases. Effects of RNA interference targeting SPRY2 on cellular proliferation in established GBM or patient-derived GBM stemlike cells were examined. Loss- or gain-of-function of SPRY2 to regulate the tumorigenic capacity was assessed in both intracranial and subcutaneous xenografts. Results: SPRY2 was found to be upregulated in GBM, which correlated with reduced survival in GBM patients. SPRY2 knockdown significantly impaired proliferation of GBM cells but not of normal astrocytes. Silencing of SPRY2 increased epidermal growth factor-induced extracellular signal-regulated kinase (ERK) and Akt activation causing premature onset of DNA replication, increased DNA damage, and impaired proliferation, suggesting that SPRY2 suppresses DNA replication stress. Abrogating SPRY2 function strongly inhibited intracranial tumor growth and led to significantly prolonged survival of U87 xenograft-bearing mice. In contrast, SPRY2 overexpression promoted tumor propagation of low-tumorigenic U251 cells. Conclusions: The present study highlights an antitumoral effect of SPRY2 inhibition that is based on excessive activation of ERK signaling and DNA damage response, resulting in reduced cell proliferation and increased cytotoxicity, proposing SPRY2 as a promising pharmacological target in GBM patients.


Subject(s)
Brain Neoplasms/pathology , Carcinogenesis/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Prognosis , RNA, Small Interfering/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Up-Regulation , Xenograft Model Antitumor Assays
19.
Exp Neurol ; 279: 137-148, 2016 May.
Article in English | MEDLINE | ID: mdl-26940083

ABSTRACT

After peripheral nerve injury, recovery of motor performance negatively correlates with the poly-innervation of neuromuscular junctions (NMJ) due to excessive sprouting of the terminal Schwann cells. Denervated muscles produce short-range diffusible sprouting stimuli, of which some are neurotrophic factors. Based on recent data that vibrissal whisking is restored perfectly during facial nerve regeneration in blind rats from the Sprague Dawley (SD)/RCS strain, we compared the expression of brain derived neurotrophic factor (BDNF), fibroblast growth factor-2 (FGF2), insulin growth factors 1 and 2 (IGF1, IGF2) and nerve growth factor (NGF) between SD/RCS and SD-rats with normal vision but poor recovery of whisking function after facial nerve injury. To establish which trophic factors might be responsible for proper NMJ-reinnervation, the transected facial nerve was surgically repaired (facial-facial anastomosis, FFA) for subsequent analysis of mRNA and proteins expressed in the levator labii superioris muscle. A complicated time course of expression included (1) a late rise in BDNF protein that followed earlier elevated gene expression, (2) an early increase in FGF2 and IGF2 protein after 2 days with sustained gene expression, (3) reduced IGF1 protein at 28 days coincident with decline of raised mRNA levels to baseline, and (4) reduced NGF protein between 2 and 14 days with maintained gene expression found in blind rats but not the rats with normal vision. These findings suggest that recovery of motor function after peripheral nerve injury is due, at least in part, to a complex regulation of lesion-associated neurotrophic factors and cytokines in denervated muscles. The increase of FGF-2 protein and concomittant decrease of NGF (with no significant changes in BDNF or IGF levels) during the first week following FFA in SD/RCS blind rats possibly prevents the distal branching of regenerating axons resulting in reduced poly-innervation of motor endplates.


Subject(s)
Facial Muscles/metabolism , Facial Muscles/pathology , Facial Nerve Injuries/metabolism , Facial Nerve Injuries/pathology , Facial Paralysis/metabolism , Facial Paralysis/pathology , Nerve Growth Factors/biosynthesis , Animals , Behavior, Animal , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Facial Muscles/innervation , Female , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor II/genetics , Nerve Growth Factor/biosynthesis , Nerve Growth Factors/genetics , Nerve Regeneration , Neuromuscular Junction/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Recovery of Function/genetics , Vibrissae/innervation
20.
Eur J Neurosci ; 43(3): 309-17, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26222895

ABSTRACT

Peripheral axonal regeneration requires surface-expanding membrane addition. The continuous incorporation of new membranes into the axolemma allows the pushing force of elongating microtubules to drive axonal growth cones forwards. Hence, a constant supply of membranes and cytoskeletal building blocks is required, often for many weeks. In human peripheral nerves, axonal tips may be more than 1 m away from the neuronal cell body. Therefore, in the initial phase of regeneration, membranes are derived from pre-existing vesicles or synthesised locally. Only later stages of axonal regeneration are supported by membranes and proteins synthesised in neuronal cell bodies, considering that the fastest anterograde transport mechanisms deliver cargo at 20 cm/day. Whereas endocytosis and exocytosis of membrane vesicles are balanced in intact axons, membrane incorporation exceeds membrane retrieval during regeneration to compensate for the loss of membranes distal to the lesion site. Physiological membrane turnover rates will not be established before the completion of target reinnervation. In this review, the current knowledge on membrane traffic in axonal outgrowth is summarised, with a focus on endosomal vesicles as the providers of membranes and carriers of growth factor receptors required for initiating signalling pathways to promote the elongation and branching of regenerating axons in lesioned peripheral nerves.


Subject(s)
Axonal Transport , Axons/metabolism , Cell Membrane/metabolism , Nerve Regeneration , Receptors, Neurotransmitter/metabolism , Animals , Axons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL