Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Proc Natl Acad Sci U S A ; 119(24): e2121804119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35666871

ABSTRACT

Neuromyelitis optica spectrum disorders (NMOSDs) are caused by immunoglobulin G (IgG) autoantibodies directed against the water channel aquaporin-4 (AQP4). In NMOSDs, discrete clinical relapses lead to disability and are robustly prevented by the anti-CD20 therapeutic rituximab; however, its mechanism of action in autoantibody-mediated disorders remains poorly understood. We hypothesized that AQP4-IgG production in germinal centers (GCs) was a core feature of NMOSDs and could be terminated by rituximab. To investigate this directly, deep cervical lymph node (dCLN) aspirates (n = 36) and blood (n = 406) were studied in a total of 63 NMOSD patients. Clinical relapses were associated with AQP4-IgM generation or shifts in AQP4-IgG subclasses (odds ratio = 6.0; range of 3.3 to 10.8; P < 0.0001), features consistent with GC activity. From seven dCLN aspirates of patients not administered rituximab, AQP4-IgGs were detected alongside specific intranodal synthesis of AQP4-IgG. AQP4-reactive B cells were isolated from unmutated naive and mutated memory populations in both blood and dCLNs. After rituximab administration, fewer clinical relapses (annual relapse rate of 0.79 to 0; P < 0.001) were accompanied by marked reductions in both AQP4-IgG (fourfold; P = 0.004) and intranodal B cells (430-fold; P < 0.0001) from 11 dCLNs. Our findings implicate ongoing GC activity as a rituximab-sensitive driver of AQP4 antibody production. They may explain rituximab's clinical efficacy in several autoantibody-mediated diseases and highlight the potential value of direct GC measurements across autoimmune conditions.


Subject(s)
Aquaporin 4 , Germinal Center , Immunologic Factors , Neuromyelitis Optica , Rituximab , Aquaporin 4/drug effects , Aquaporin 4/metabolism , Autoantibodies , Germinal Center/pathology , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Lymph Nodes/metabolism , Neuromyelitis Optica/drug therapy , Rituximab/pharmacology , Rituximab/therapeutic use
2.
Ann Neurol ; 88(6): 1244-1250, 2020 12.
Article in English | MEDLINE | ID: mdl-32951275

ABSTRACT

This study aimed to characterise both neuronal autoantibodies and levels of interferon α, two proposed causative agents in neuropsychiatric systemic lupus erythematosus (NPSLE). Cerebrospinal fluid (CSF) and plasma from 35 patients with systemic lupus erythematosus (SLE; 15 with NPSLE) showed no antibodies against natively expressed N-methyl-D-aspartate receptors (NMDARs), or the surface of live hippocampal neurons. By comparison to controls (n = 104), patients with SLE had antibodies that bound to a peptide representing the extracellular domain of NMDARs (p < 0.0001), however, binding was retained against both rearranged peptides and no peptide (r = 0.85 and r = 0.79, respectively, p < 0.0001). In summary, neuronal-surface reactive antibodies were not detected in NPSLE. Further, while interferon α levels were higher in SLE (p < 0.0001), they lacked specificity for NPSLE. Our findings mandate a search for novel biomarkers in this condition. ANN NEUROL 2020;88:1244-1250.


Subject(s)
Autoantibodies/immunology , Lupus Vasculitis, Central Nervous System/immunology , Neurons/immunology , Adolescent , Adult , Aged , Autoantibodies/blood , Autoantibodies/cerebrospinal fluid , Case-Control Studies , Cells, Cultured , Female , Hippocampus/immunology , Humans , Interferon-alpha/blood , Male , Middle Aged , Receptors, N-Methyl-D-Aspartate/immunology , Young Adult
3.
Brain ; 143(6): 1731-1745, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32437528

ABSTRACT

Autoantibodies against leucine-rich glioma inactivated 1 (LGI1) are found in patients with limbic encephalitis and focal seizures. Here, we generate patient-derived monoclonal antibodies (mAbs) against LGI1. We explore their sequences and binding characteristics, plus their pathogenic potential using transfected HEK293T cells, rodent neuronal preparations, and behavioural and electrophysiological assessments in vivo after mAb injections into the rodent hippocampus. In live cell-based assays, LGI1 epitope recognition was examined with patient sera (n = 31), CSFs (n = 11), longitudinal serum samples (n = 15), and using mAbs (n = 14) generated from peripheral B cells of two patients. All sera and 9/11 CSFs bound both the leucine-rich repeat (LRR) and the epitempin repeat (EPTP) domains of LGI1, with stable ratios of LRR:EPTP antibody levels over time. By contrast, the mAbs derived from both patients recognized either the LRR or EPTP domain. mAbs against both domain specificities showed varied binding strengths, and marked genetic heterogeneity, with high mutation frequencies. LRR-specific mAbs recognized LGI1 docked to its interaction partners, ADAM22 and ADAM23, bound to rodent brain sections, and induced internalization of the LGI1-ADAM22/23 complex in both HEK293T cells and live hippocampal neurons. By contrast, few EPTP-specific mAbs bound to rodent brain sections or ADAM22/23-docked LGI1, but all inhibited the docking of LGI1 to ADAM22/23. After intrahippocampal injection, and by contrast to the LRR-directed mAbs, the EPTP-directed mAbs showed far less avid binding to brain tissue and were consistently detected in the serum. Post-injection, both domain-specific mAbs abrogated long-term potentiation induction, and LRR-directed antibodies with higher binding strengths induced memory impairment. Taken together, two largely dichotomous populations of LGI1 mAbs with distinct domain binding characteristics exist in the affinity matured peripheral autoantigen-specific memory pools of individuals, both of which have pathogenic potential. In human autoantibody-mediated diseases, the detailed characterization of patient mAbs provides a valuable method to dissect the molecular mechanisms within polyclonal populations.


Subject(s)
Antibodies, Monoclonal/metabolism , Autoantibodies/immunology , Intracellular Signaling Peptides and Proteins/immunology , ADAM Proteins/metabolism , Animals , Antibodies, Monoclonal/immunology , Autoantibodies/blood , Autoantibodies/metabolism , Autoantigens/metabolism , Brain/metabolism , Epitopes/immunology , HEK293 Cells , Hippocampus/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Limbic Encephalitis/blood , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Binding/immunology , Protein Domains/immunology
4.
Ann Neurol ; 83(3): 553-561, 2018 03.
Article in English | MEDLINE | ID: mdl-29406578

ABSTRACT

INTRODUCTION: N-methyl-D-aspartate receptor (NMDAR) antibody encephalitis is mediated by immunoglobulin G (IgG) autoantibodies directed against the NR1 subunit of the NMDAR. Around 20% of patients have an underlying ovarian teratoma, and the condition responds to early immunotherapies and ovarian teratoma removal. However, despite clear therapeutic relevance, mechanisms of NR1-IgG production and the contribution of germinal center B cells to NR1-IgG levels are unknown. METHODS: Clinical data and longitudinal paired serum NR1-reactive IgM and IgG levels from 10 patients with NMDAR-antibody encephalitis were determined. Peripheral blood mononuclear cells from these 10 patients, and two available ovarian teratomas, were stimulated with combinations of immune factors and tested for secretion of total IgG and NR1-specific antibodies. RESULTS: In addition to disease-defining NR1-IgG, serum NR1-IgM was found in 6 of 10 patients. NR1-IgM levels were typically highest around disease onset and detected for several months into the disease course. Moreover, circulating patient B cells were differentiated into CD19+ CD27++ CD38++ antibody-secreting cells in vitro and, from 90% of patients, secreted NR1-IgM and NR1-IgG. Secreted levels of NR1-IgG correlated with serum NR1-IgG (p < 0.0001), and this was observed across the varying disease durations, suggestive of an ongoing process. Furthermore, ovarian teratoma tissue contained infiltrating lymphocytes which produced NR1-IgG in culture. INTERPRETATION: Serum NR1-IgM and NR1-IgG, alongside the consistent production of NR1-IgG from circulating B cells and from ovarian teratomas suggest that ongoing germinal center reactions may account for the peripheral cell populations which secrete NR1-IgG. Cells participating in germinal center reactions might be a therapeutic target for the treatment of NMDAR-antibody encephalitis. Ann Neurol 2018;83:553-561.


Subject(s)
Autoantibodies/blood , Germinal Center/metabolism , Immunoglobulin G/blood , Immunoglobulin M/blood , Receptors, N-Methyl-D-Aspartate/blood , Adolescent , Adult , Aged , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/blood , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/diagnosis , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/immunology , Autoantibodies/immunology , Female , Germinal Center/immunology , HEK293 Cells , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Longitudinal Studies , Middle Aged , Ovarian Neoplasms/blood , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/immunology , Prospective Studies , Receptors, N-Methyl-D-Aspartate/immunology , Teratoma/blood , Teratoma/diagnosis , Teratoma/immunology , Young Adult
5.
Neurobiol Dis ; 65: 160-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24423643

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by progressive loss of motor neurons (MNs) and astrogliosis. Recent evidence suggests that factors secreted by activated astrocytes might contribute to degeneration of MNs. We focused on endothelin-1 (ET-1), a peptide which is strongly up-regulated in reactive astrocytes under different pathological conditions. We show that ET-1 is abundantly expressed by reactive astrocytes in the spinal cord of the SOD1-G93A mouse model and sporadic ALS patients. To test if ET-1 might play a role in degeneration of MNs, we investigated its effect on MN survival in an in vitro model of mixed rat spinal cord cultures (MSCs) enriched of astrocytes exhibiting a reactive phenotype. ET-1 exerted a toxic effect on MNs in a time- and concentration-dependent manner, with an exposure to 100-200nM ET-1 for 48h resulting in 40-50% MN cell death. Importantly, ET-1 did not induce MN degeneration when administered on cultures treated with AraC (5µM) or grown in a serum-free medium that did not favor astrocyte proliferation and reactivity. We found that both ETA and ETB receptors are enriched in astrocytes in MSCs. The ET-1 toxic effect was mimicked by ET-3 (100nM) and sarafotoxin S6c (10nM), two selective agonists of endothelin-B receptors, and was not additive with that of ET-3 suggesting the involvement of ETB receptors. Surprisingly, however, the ET-1 effect persisted in the presence of the ETB receptor antagonist BQ-788 (200nM-2µM) and was slightly reversed by the ETA receptor antagonist BQ-123 (2µM), suggesting an atypical pharmacological profile of the astrocytic receptors responsible for ET-1 toxicity. The ET-1 effect was not undone by the ionotropic glutamate receptor AMPA antagonist GYKI 52466 (20µM), indicating that it is not caused by an increased glutamate release. Conversely, a 48-hour ET-1 treatment increased MN cell death induced by acute exposure to AMPA (50µM), which is indicative of two distinct pathways leading to neuronal death. Altogether these results indicate that ET-1 exerts a toxic effect on cultured MNs through mechanisms mediated by reactive astrocytes and suggest that ET-1 may contribute to MN degeneration in ALS. Thus, a treatment aimed at lowering ET-1 levels or antagonizing its effect might be envisaged as a potential therapeutic strategy to slow down MN degeneration in this devastating disease.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Endothelin-1/pharmacology , Motor Neurons/drug effects , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/genetics , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Embryo, Mammalian , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Pregnancy , Rats , Rats, Wistar , Spinal Cord/cytology , Superoxide Dismutase/genetics , Time Factors
6.
Nat Commun ; 14(1): 8354, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102121

ABSTRACT

The self-templating nature of prions plays a central role in prion pathogenesis and is associated with infectivity and transmissibility. Since propagation of proteopathic seeds has now been acknowledged a principal pathogenic process in many types of dementia, more insight into the molecular mechanism of prion replication is vital to delineate specific and common disease pathways. By employing highly discriminatory anti-PrP antibodies and conversion-tolerant PrP chimera, we here report that de novo PrP conversion and formation of fibril-like PrP aggregates are distinct in mechanistic and kinetic terms. De novo PrP conversion occurs within minutes after infection at two subcellular locations, while fibril-like PrP aggregates are formed exclusively at the plasma membrane, hours after infection. Phenotypically distinct pools of abnormal PrP at perinuclear sites and the plasma membrane show differences in N-terminal processing, aggregation state and fibril formation and are linked by exocytic transport via synaptic and large-dense core vesicles.


Subject(s)
Prion Diseases , Prions , Humans , Prion Proteins , Prions/metabolism , Cell Line , Cell Membrane/metabolism , Muscle Fibers, Skeletal/metabolism , Prion Diseases/metabolism
7.
Ann Indian Acad Neurol ; 25(3): 473-478, 2022.
Article in English | MEDLINE | ID: mdl-35936609

ABSTRACT

Background: Prevalence of antibody-mediated autoimmune encephalitis (AE) is reported to be comparable to infectious encephalitis in Western populations. We evaluated the frequency and significance of AE and neuronal autoantibodies in comparison to infectious etiologies among patients presenting with encephalitis in a South Asian population. Methods: Ninety-nine consecutive patients with a clinical diagnosis of encephalitis/meningoencephalitis admitted to two of the largest tertiary-care hospitals in Sri Lanka were studied. PCR and ELISA were used to screen viruses while Gram stain and culture were used to screen bacteria. Sera were tested for antibodies binding to primary embryonic rat hippocampal neuronal cultures and cell-based assays for antibodies to NMDAR, LGI1, CASPR2, Contactin2, AMPAR, GABAAR, GABABR, aquaporin-4 and MOG. Results: Patient ages ranged from 1 month to 73 years (mean = 24.91; SD = 21.33) with a male: female ratio of 1.75:1. A viral etiology was identified in 27.3% and bacterial meningoencephalitis was diagnosed in 17.1%. Sera of nine patients had antibodies binding to live primary neurons, but only five had specific antibodies to CASPR2 (n = 1), NMDAR (n = 2) or GABABR-antibodies (n = 2). Moreover, the patients with CASPR2 antibodies and NMDAR-antibodies were also positive for dengue antibodies. Only the two patients with NMDAR-antibodies had features and responses to immunotherapy consistent with AE. Conclusions: Identified infectious forms of meningoencephalitis (44.4%) greatly exceeded the occurrence of neuronal autoantibodies (9.1%) and AE (2%) in Sri Lanka, and this may be common in those regions where infections are prevalent.

8.
Neurobiol Dis ; 42(3): 252-64, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21232601

ABSTRACT

The causes of amyotrophic lateral sclerosis (ALS) are mostly undefined; however, excitotoxic injury and astrogliosis may contribute to motor neuron (MN) degeneration. Group I metabotropic glutamate (mGlu) receptors are over-expressed in reactive astrocytes in ALS, but the functional significance of this over-expression is presently unknown. We examined the role of group I mGlu receptors on excitotoxic death of spinal cord MNs grown in cultures enriched of astrocytes bearing a reactive phenotype. A prolonged exposure to the selective non-competitive mGlu5 receptor antagonist MPEP reduced AMPA-mediated toxicity and cobalt uptake in MNs. Expression levels of the GluR1 (but not GluR2) AMPA receptor subunit and levels of brain-derived neurotrophic factor (BDNF) were reduced in mixed spinal cord cultures pretreated with MPEP. In addition, neuroprotection by MPEP was less than additive with that produced by a neutralizing anti-BDNF antibody and a treatment with exogenous BDNF masked the protective effect of MPEP, suggesting that mGlu5 receptors and BDNF converge in facilitating excitotoxic MN death. The protective effect of MPEP was absent in cultures with a reduced number of astrocytes. We suggest that blocking astrocytic mGlu5 receptors is a potential therapeutic strategy in ALS.


Subject(s)
Cell Death/drug effects , Motor Neurons/drug effects , Receptors, Metabotropic Glutamate/metabolism , Spinal Cord/drug effects , Analysis of Variance , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Chromones/pharmacology , Enzyme-Linked Immunosorbent Assay , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Immunohistochemistry , Motor Neurons/cytology , Motor Neurons/metabolism , Neuroprotective Agents/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Spinal Cord/cytology , Spinal Cord/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
9.
Nucleic Acids Res ; 37(4): 1269-79, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19136466

ABSTRACT

FRAXE is a form of mild to moderate mental retardation due to the silencing of the FMR2 gene. The cellular function of FMR2 protein is presently unknown. By analogy with its homologue AF4, FMR2 was supposed to have a role in transcriptional regulation, but robust evidences supporting this hypothesis are lacking. We observed that FMR2 co-localizes with the splicing factor SC35 in nuclear speckles, the nuclear regions where splicing factors are concentrated, assembled and modified. Similarly to what was reported for splicing factors, blocking splicing or transcription leads to the accumulation of FMR2 in enlarged, rounded speckles. FMR2 is also localized in the nucleolus when splicing is blocked. We show here that FMR2 is able to specifically bind the G-quartet-forming RNA structure with high affinity. Remarkably, in vivo, in the presence of FMR2, the ESE action of the G-quartet situated in mRNA of an alternatively spliced exon of a minigene or of the putative target FMR1 appears reduced. Interestingly, FMR1 is silenced in the fragile X syndrome, another form of mental retardation. All together, our findings strongly suggest that FMR2 is an RNA-binding protein, which might be involved in alternative splicing regulation through an interaction with G-quartet RNA structure.


Subject(s)
Alternative Splicing , G-Quadruplexes , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , RNA/chemistry , Animals , Cell Line , Cell Nucleus Structures/chemistry , Cells, Cultured , Fragile X Mental Retardation Protein/genetics , Humans , Mice , Nuclear Proteins/analysis , Nuclear Proteins/chemistry , Protein Structure, Tertiary , RNA/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/chemistry
10.
Transl Stroke Res ; 12(1): 72-86, 2021 02.
Article in English | MEDLINE | ID: mdl-32253702

ABSTRACT

The extracellular matrix fragment perlecan domain V is neuroprotective and functionally restorative following experimental stroke. As neurogenesis is an important component of chronic post-stroke repair, and previous studies have implicated perlecan in developmental neurogenesis, we hypothesized that domain V could have a broad therapeutic window by enhancing neurogenesis after stroke. We demonstrated that domain V is chronically increased in the brains of human stroke patients, suggesting that it is present during post-stroke neurogenic periods. Furthermore, perlecan deficient mice had significantly less neuroblast precursor cells after experimental stroke. Seven-day delayed domain V administration enhanced neurogenesis and restored peri-infarct excitatory synaptic drive to neocortical layer 2/3 pyramidal neurons after experimental stroke. Domain V's effects were inhibited by blockade of α2ß1 integrin, suggesting the importance of α2ß1 integrin to neurogenesis and domain V neurogenic effects. Our results demonstrate that perlecan plays a previously unrecognized role in post-stroke neurogenesis and that delayed DV administration after experimental stroke enhances neurogenesis and improves recovery in an α2ß1 integrin-mediated fashion. We conclude that domain V is a clinically relevant neuroprotective and neuroreparative novel stroke therapy with a broad therapeutic window.


Subject(s)
Brain/metabolism , Heparan Sulfate Proteoglycans/biosynthesis , Neurogenesis/physiology , Neuroprotection/physiology , Stroke/metabolism , Animals , Brain/drug effects , Brain/pathology , Cells, Cultured , Heparan Sulfate Proteoglycans/administration & dosage , Humans , Male , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Neuroprotection/drug effects , Organ Culture Techniques , Protein Domains , Stroke/pathology , Stroke/prevention & control
11.
J Neuroimmunol ; 340: 577149, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31951874

ABSTRACT

Since approximately 50% of patients with Lambert-Eaton myasthenic syndrome (LEMS) subsequently develop small-cell lung cancer (SCLC), it is important to be able to predict cancer occurrence in these patients at neurological presentation. We aimed to determine whether circulating biomarkers were effective and objective predictors of cancer development in LEMS. We found that the presence of either SOX2, N-type voltage gated calcium channel or GABAb antibodies at LEMS diagnosis was highly sensitive (84%) and specific (87%) for the detection of SCLC. Screening for SOX2 and neuronal antibodies is a useful adjunct to clinical predictive scoring tools in predicting SCLC in LEMS.


Subject(s)
Autoantibodies/blood , Biomarkers, Tumor/blood , Lambert-Eaton Myasthenic Syndrome/complications , Lung Neoplasms/immunology , Small Cell Lung Carcinoma/immunology , Adult , Aged , Aged, 80 and over , Autoantibodies/immunology , Autoantigens/immunology , Female , Humans , Lambert-Eaton Myasthenic Syndrome/immunology , Lung Neoplasms/complications , Lung Neoplasms/diagnosis , Male , Middle Aged , Neurons/immunology , Small Cell Lung Carcinoma/complications , Small Cell Lung Carcinoma/diagnosis
12.
Neurochem Res ; 33(12): 2436-43, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18438710

ABSTRACT

Glutamate is the major excitatory neurotransmitter in the mammalian central nervous system (CNS) and exerts its actions via a number of ionotropic glutamate receptors/channels and metabotropic glutamate (mGlu) receptors. In addition to being expressed in neurons, glutamate receptors are expressed in different types of glial cells including astrocytes, oligodendrocytes, and microglia. Astrocytes are now recognized as dynamic signaling elements actively integrating neuronal inputs. Synaptic activity can evoke calcium signals in astrocytes, resulting in the release of gliotransmitters, such as glutamate, ATP, and D-serine, which in turn modulate neuronal excitability and synaptic transmission. In addition, astrocytes, and microglia may play an important role in pathology such as brain trauma and neurodegeneration, limiting or amplifying the pathologic process leading to neuronal death. The present review will focus on recent advances on the role of mGlu receptors expressed in glial cells under physiologic and pathologic conditions.


Subject(s)
Neuroglia/metabolism , Receptors, Metabotropic Glutamate/physiology , Animals , Humans , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction
13.
Sci Rep ; 7(1): 241, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28325900

ABSTRACT

Insulin-like growth factor-I (IGF-I) is involved in the maturation and maintenance of neurons, and impaired IGF-I signaling has been shown to play a role in various neurological diseases including stroke. The aim of the present study was to investigate the efficacy of an optimized IGF-I variant by adding a 40 kDa polyethylene glycol (PEG) chain to IGF-I to form PEG-IGF-I. We show that PEG-IGF-I has a slower clearance which allows for twice-weekly dosing to maintain steady-state serum levels in mice. Using a photothrombotic model of focal stroke, dosing from 3 hrs post-stroke dose-dependently (0.3-1 mg/kg) decreases the volume of infarction and improves motor behavioural function in both young 3-month and aged 22-24 month old mice. Further, PEG-IGF-I treatment increases GFAP expression when given early (3 hrs post-stroke), increases Synaptophysin expression and increases neurogenesis in young and aged. Finally, neurons (P5-6) cultured in vitro on reactive astrocytes in the presence of PEG-IGF-I showed an increase in neurite length, indicating that PEG-IGF-I can aid in sprouting of new connections. This data suggests a modulatory role of IGF-I in both protective and regenerative processes, and indicates that therapeutic approaches using PEG-IGF-I should be given early and where the endogenous regenerative potential is still high.


Subject(s)
Insulin-Like Growth Factor I/administration & dosage , Ischemia/therapy , Stroke/therapy , Animals , Disease Models, Animal , Insulin-Like Growth Factor I/pharmacokinetics , Locomotion , Mice , Serum/chemistry , Treatment Outcome
15.
Sci Rep ; 6: 21896, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26902390

ABSTRACT

Although recovery following a stroke is limited, undamaged neurons under the right conditions can establish new connections and take on-board lost functions. Sonic hedgehog (Shh) signaling is integral for developmental axon growth, but its role after injury has not been fully examined. To investigate the effects of Shh on neuronal sprouting after injury, we used an in vitro model of glial scar, whereby cortical astrocytes were mechanically traumatized to mimic reactive astrogliosis observed after stroke. This mechanical trauma impaired neurite outgrowth from post-natal cortical neurons plated on top of reactive astrocytes. Addition of Shh to the media, however, resulted in a concentration-dependent increase in neurite outgrowth. This response was inhibited by cyclopamine and activated by oxysterol 20(S)-hydroxycholesterol, both of which modulate the activity of the Shh co-receptor Smoothened (Smo), demonstrating that Shh-mediated neurite outgrowth is Smo-dependent. In addition, neurite outgrowth was not associated with an increase in Gli-1 transcription, but could be inhibited by PP2, a selective inhibitor of Src family kinases. These results demonstrate that neurons exposed to the neurite growth inhibitory environment associated with a glial scar can be stimulated by Shh, with signaling occurring through a non-canonical pathway, to overcome this suppression and stimulate neurite outgrowth.


Subject(s)
Astrocytes/drug effects , Gliosis/metabolism , Hedgehog Proteins/pharmacology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Coculture Techniques , Gene Expression Regulation , Gliosis/genetics , Gliosis/pathology , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hydroxycholesterols/pharmacology , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/cytology , Neurons/metabolism , Pressure , Primary Cell Culture , Pyrimidines/pharmacology , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Stress, Mechanical , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/metabolism
16.
Expert Rev Neurother ; 14(11): 1335-44, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25319267

ABSTRACT

Stroke remains the leading cause of long-term disability with no pharmacological approaches available to limit the degree of damage or aid in recovery. Considerable effort has been made to minimize neuronal damage using neuroprotective compounds. However, attempts have so far failed to translate into the clinic. Brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-related kinase type B are actively produced throughout the brain and are involved in regulating neuronal activity and normal day-to-day function. Further, BDNF has been shown to play a role in both protection and recovery of functions after stroke. This review focuses on the endogenous release of BDNF as well as activity-induced (pharmacological and physical) elevation in BDNF, and the role this plays during both acute (hours to days) and subacute (days to weeks) periods after stroke. Exogenous administration has previously been shown not to cross the blood-brain barrier; therefore, we have focused this review on approaches that allow us to directly stimulate, using pharmacological therapies and mimetics, physical activity and potential drug delivery systems that can be used to administer BDNF. Finally, we also discuss the role of BDNF polymorphisms and the influence of epigenetic regulation of BDNF on post-stroke recovery.


Subject(s)
Brain Ischemia/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Receptor, trkB/metabolism , Recovery of Function/physiology , Stroke/metabolism , Brain Ischemia/rehabilitation , Humans , Neurons/metabolism , Stroke Rehabilitation
SELECTION OF CITATIONS
SEARCH DETAIL