Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 4655, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35945193

ABSTRACT

Friedreich's ataxia (FA) is an inherited progressive neurodegenerative disease for which there is no proven disease-modifying treatment. Here we perform an open-label, pilot study of recombinant human granulocyte-colony stimulating factor (G-CSF) administration in seven people with FA (EudraCT: 2017-003084-34); each participant receiving a single course of G-CSF (Lenograstim; 1.28 million units per kg per day for 5 days). The primary outcome is peripheral blood mononuclear cell frataxin levels over a 19-day period. The secondary outcomes include safety, haematopoietic stem cell (HSC) mobilisation, antioxidant levels and mitochondrial enzyme activity. The trial meets pre-specified endpoints. We show that administration of G-CSF to people with FA is safe. Mobilisation of HSCs in response to G-CSF is comparable to that of healthy individuals. Notably, sustained increases in cellular frataxin concentrations and raised PGC-1α and Nrf2 expression are detected. Our findings show potential for G-CSF therapy to have a clinical impact in people with FA.


Subject(s)
Friedreich Ataxia , Granulocyte Colony-Stimulating Factor , Recombinant Proteins , Friedreich Ataxia/drug therapy , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocytes/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Pilot Projects , Recombinant Proteins/adverse effects
2.
Mult Scler ; 28(8): 1179-1188, 2022 07.
Article in English | MEDLINE | ID: mdl-34841955

ABSTRACT

BACKGROUND: Cell-based therapies for multiple sclerosis (MS), including those employing autologous bone marrow-derived mesenchymal stromal cells (MSC) are being examined in clinical trials. However, recent studies have identified abnormalities in the MS bone marrow microenvironment. OBJECTIVE: We aimed to compare the secretome of MSC isolated from control subjects (C-MSC) and people with MS (MS-MSC) and explore the functional relevance of findings. METHODS: We employed high throughput proteomic analysis, enzyme-linked immunosorbent assays and immunoblotting, as well as in vitro assays of enzyme activity and neuroprotection. RESULTS: We demonstrated that, in progressive MS, the MSC secretome has lower levels of mitochondrial fumarate hydratase (mFH). Exogenous mFH restores the in vitro neuroprotective potential of MS-MSC. Furthermore, MS-MSC expresses reduced levels of fumarate hydratase (FH) with downstream reduction in expression of master regulators of oxidative stress. CONCLUSIONS: Our findings are further evidence of dysregulation of the bone marrow microenvironment in progressive MS with respect to anti-oxidative capacity and immunoregulatory potential. Given the clinical utility of the fumaric acid ester dimethyl fumarate in relapsing-remitting MS, our findings have potential implication for understanding MS pathophysiology and personalised therapeutic intervention.


Subject(s)
Fumarate Hydratase , Mesenchymal Stem Cells , Mitochondria , Multiple Sclerosis, Chronic Progressive , Neuroprotection , Fumarate Hydratase/metabolism , Humans , Mitochondria/enzymology , Multiple Sclerosis, Chronic Progressive/metabolism , Proteomics
3.
J Neurol ; 268(6): 2175-2184, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33484325

ABSTRACT

There is increasing interest in the development of multiple sclerosis (MS) biomarkers that reflect central nervous system tissue injury to determine prognosis. We aimed to assess the prognostic value of kinesin superfamily motor protein KIF5A in MS by measuring levels of KIF5A in cerebrospinal fluid (CSF) combined with analysis of single nucleotide polymorphisms (SNPs; rs12368653 and rs703842) located within a MS susceptibility gene locus at chromosome 12q13-14 region. Enzyme-linked immunosorbent assay was used to measure KIF5A in CSF obtained from two independent biobanks comprising non-inflammatory neurological disease controls (NINDC), clinically isolated syndrome (CIS) and MS cases. CSF KIF5A expression was significantly elevated in progressive MS cases compared with NINDCs, CIS and relapsing-remitting MS (RRMS). In addition, levels of KIF5A positively correlated with change in MS disease severity scores (EDSS, MSSS and ARMSSS), in RRMS patients who had documented disease progression at 2-year clinical follow-up. Copies of adenine risk alleles (AG/AA; rs12368653 and rs703842) corresponded with a higher proportion of individuals in relapse at the time of lumbar puncture (LP), higher use of disease-modifying therapies post LP and shorter MS duration. Our study suggests that CSF KIF5A has potential as a predictive biomarker in MS and further studies into the potential prognostic value of analysing MS susceptibility SNPs should be considered.


Subject(s)
Kinesins , Multiple Sclerosis, Chronic Progressive , Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Biomarkers , Disease Progression , Genotype , Humans , Kinesins/genetics , Multiple Sclerosis/diagnosis , Multiple Sclerosis/genetics , Multiple Sclerosis, Relapsing-Remitting/genetics
4.
J Pathol ; 247(4): 422-434, 2019 04.
Article in English | MEDLINE | ID: mdl-30565681

ABSTRACT

The overall survival for patients with primary glioblastoma is very poor. Glioblastoma contains a subpopulation of glioma stem cells (GSC) that are responsible for tumour initiation, treatment resistance and recurrence. PPARα is a transcription factor involved in the control of lipid, carbohydrate and amino acid metabolism. We have recently shown that PPARα gene and protein expression is increased in glioblastoma and has independent clinical prognostic significance in multivariate analyses. In this work, we report that PPARα is overexpressed in GSC compared to foetal neural stem cells. To investigate the role of PPARα in GSC, we knocked down its expression using lentiviral transduction with short hairpin RNA (shRNA). Transduced GSC were tagged with luciferase and stereotactically xenografted into the striatum of NOD-SCID mice. Bioluminescent and magnetic resonance imaging showed that knockdown (KD) of PPARα reduced the tumourigenicity of GSC in vivo. PPARα-expressing control GSC xenografts formed invasive histological phenocopies of human glioblastoma, whereas PPARα KD GSC xenografts failed to establish viable intracranial tumours. PPARα KD GSC showed significantly reduced proliferative capacity and clonogenic potential in vitro with an increase in cellular senescence. In addition, PPARα KD resulted in significant downregulation of the stem cell factors c-Myc, nestin and SOX2. This was accompanied by downregulation of the PPARα-target genes and key regulators of fatty acid oxygenation ACOX1 and CPT1A, with no compensatory increase in glycolytic flux. These data establish the aberrant overexpression of PPARα in GSC and demonstrate that this expression functions as an important regulator of tumourigenesis, linking self-renewal and the malignant phenotype in this aggressive cancer stem cell subpopulation. We conclude that targeting GSC PPARα expression may be a therapeutically beneficial strategy with translational potential as an adjuvant treatment. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , PPAR alpha/metabolism , RNA, Small Interfering/pharmacology , Animals , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques/methods , Humans , Lentivirus , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction/physiology , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Ann Neurol ; 83(4): 779-793, 2018 04.
Article in English | MEDLINE | ID: mdl-29534309

ABSTRACT

OBJECTIVE: Friedreich's ataxia is an incurable inherited neurological disease caused by frataxin deficiency. Here, we report the neuroreparative effects of myeloablative allogeneic bone marrow transplantation in a humanized murine model of the disease. METHODS: Mice received a transplant of fluorescently tagged sex-mismatched bone marrow cells expressing wild-type frataxin and were assessed at monthly intervals using a range of behavioral motor performance tests. At 6 months post-transplant, mice were euthanized for protein and histological analysis. In an attempt to augment numbers of bone marrow-derived cells integrating within the nervous system and improve therapeutic efficacy, a subgroup of transplanted mice also received monthly subcutaneous infusions of the cytokines granulocyte-colony stimulating factor and stem cell factor. RESULTS: Transplantation caused improvements in several indicators of motor coordination and locomotor activity. Elevations in frataxin levels and antioxidant defenses were detected. Abrogation of disease pathology throughout the nervous system was apparent, together with extensive integration of bone marrow-derived cells in areas of nervous tissue injury that contributed genetic material to mature neurons, satellite-like cells, and myelinating Schwann cells by processes including cell fusion. Elevations in circulating bone marrow-derived cell numbers were detected after cytokine administration and were associated with increased frequencies of Purkinje cell fusion and bone marrow-derived dorsal root ganglion satellite-like cells. Further improvements in motor coordination and activity were evident. INTERPRETATION: Our data provide proof of concept of gene replacement therapy, via allogeneic bone marrow transplantation, that reverses neurological features of Friedreich's ataxia with the potential for rapid clinical translation. Ann Neurol 2018;83:779-793.


Subject(s)
Bone Marrow Transplantation/methods , Friedreich Ataxia/surgery , Animals , Body Weight/physiology , Cytokines/metabolism , Disease Models, Animal , Exploratory Behavior/physiology , Friedreich Ataxia/genetics , Ganglia, Spinal/pathology , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/therapeutic use , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Iron-Binding Proteins/genetics , Leukocytes, Mononuclear/pathology , Mice , Mice, Inbred C57BL , Muscle Strength/physiology , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/pathology , Frataxin
6.
J Clin Pathol ; 71(8): 695-701, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29463577

ABSTRACT

AIMS: Histopathological tissue samples are being increasingly used as sources of nucleic acids in molecular pathology translational research. This study investigated the suitability of glioblastoma and control central nervous system (CNS) formalin-fixed paraffin embedded (FFPE) tissue-derived RNA for gene expression analyses. METHODS: Total RNA was extracted from control (temporal lobe resection tissue) and glioblastoma FFPE tissue samples. RNA purity (260/280 ratios) was determined and RNA integrity number (RIN) analysis was performed. RNA was subsequently used for RT-qPCR for two reference genes, 18S and GAPDH. RESULTS: Reference gene expression was equivalent between control and glioblastoma tissue when using RNA extracted from FFPE tissue, which has key implications for biological normalisation for CNS gene expression studies. There was a significant difference between the mean RIN values of control and glioblastoma FFPE tissue. There was no significant correlation between 260/280 or RIN values versus total RNA yield. The age of the tissue blocks did not influence RNA yield, fragmentation or purity. There was no significant correlation between RIN or 260/280 ratios and mean qPCR cycle threshold for either reference gene. CONCLUSIONS: This study showed that routinely available CNS FFPE tissue is suitable for RNA extraction and downstream gene expression studies, even after 60 months of storage. Substantial RNA fragmentation associated with glioblastoma and control FFPE tissue blocks did not preclude downstream RT-qPCR gene expression analyses. Cross validation with both archival and prospectively collated FFPE specimens is required to further demonstrate that CNS tissue blocks can be used in novel translational molecular biomarker studies.


Subject(s)
Brain Neoplasms/genetics , Epilepsy, Temporal Lobe/genetics , Fixatives/chemistry , Formaldehyde/chemistry , Gene Expression Profiling , Glioblastoma/genetics , Paraffin Embedding , RNA Stability , RNA, Neoplasm/genetics , Tissue Fixation/methods , Brain Neoplasms/surgery , Case-Control Studies , Epilepsy, Temporal Lobe/surgery , Gene Expression Profiling/standards , Glioblastoma/surgery , Humans , Paraffin Embedding/standards , Predictive Value of Tests , Quality Control , Reproducibility of Results , Time Factors , Tissue Fixation/standards
7.
J Alzheimers Dis ; 60(4): 1511-1524, 2017.
Article in English | MEDLINE | ID: mdl-29060936

ABSTRACT

Defects in motor protein-mediated neuronal transport mechanisms have been implicated in a number of neurodegenerative disorders but remain relatively little studied in Alzheimer's disease (AD). Our aim in the present study was to assess the expression of the anterograde kinesin superfamily motor proteins KIF5A, KIF1B, and KIF21B, and to examine their relationship to levels of hyperphosphorylated tau, amyloid-ß protein precursor (AßPP), and amyloid-ß (Aß) in human brain tissue. We used a combination of qPCR, immunoblotting, and ELISA to perform these analyses in midfrontal cortex from 49 AD and 46 control brains. Expression of KIF5A, KIF1B, and KIF21B at gene and protein level was significantly increased in AD. KIF5A protein expression correlated inversely with the levels of AßPP and soluble Aß in AD brains. Upregulation of KIFs may be an adaptive response to impaired axonal transport in AD.


Subject(s)
Alzheimer Disease/metabolism , Frontal Lobe/metabolism , Kinesins/metabolism , Aged , Aged, 80 and over , Amyloid beta-Protein Precursor/metabolism , Blotting, Western , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Phosphorylation , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation , tau Proteins/metabolism
8.
Biomark Med ; 11(8): 589-591, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28770630
9.
Histopathology ; 70(7): 1030-1043, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27926792

ABSTRACT

AIMS: PPARα agonists are in current clinical use as hypolipidaemic agents and show significant antineoplastic effects in human glioblastoma models. To date however, the expression of PPARα in large-scale glioblastoma datasets has not been examined. We aimed to investigate the expression of the transcription factor PPARα in primary glioblastoma, the relationship between PPARα expression and patients' clinicopathological features and other molecular markers associated with gliomagenesis. METHODS AND RESULTS: With protein immunoblotting techniques and reverse transcription quantitative real-time PCR, PPARα was found to be significantly overexpressed in glioblastoma compared with control brain tissue (P = 0.032 and P = 0.005). PPARA gene expression was found to be enriched in the classical glioblastoma subtype within The Cancer Genome Atlas (TCGA) dataset. Although not associated with overall survival when assessed by immunohistochemistry, cross-validation with the TCGA dataset and multivariate analyses identified PPARA gene expression as an independent prognostic marker for overall survival (P = 0.042). Finally, hierarchical clustering revealed novel, significant associations between high PPARA expression and a putative set of glioblastoma molecular mediators including EMX2, AQP4, and NTRK2. CONCLUSIONS: PPARα is overexpressed in primary glioblastoma and high PPARA expression functions as an independent prognostic marker in the glioblastoma TCGA dataset. Further studies are required to explore genetic associations with high PPARA expression and to analyse the predictive role of PPARα expression in glioblastoma models in response to PPARα agonists.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/pathology , Glioblastoma/pathology , PPAR alpha/biosynthesis , Adult , Aged , Aged, 80 and over , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Child , Female , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Isocitrate Dehydrogenase/genetics , Kaplan-Meier Estimate , Male , Middle Aged , PPAR alpha/analysis , Prognosis , Proportional Hazards Models
10.
Ann Neurol ; 81(2): 212-226, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28009062

ABSTRACT

OBJECTIVES: Friedreich's ataxia is a devastating neurological disease currently lacking any proven treatment. We studied the neuroprotective effects of the cytokines, granulocyte-colony stimulating factor (G-CSF) and stem cell factor (SCF) in a humanized murine model of Friedreich's ataxia. METHODS: Mice received monthly subcutaneous infusions of cytokines while also being assessed at monthly time points using an extensive range of behavioral motor performance tests. After 6 months of treatment, neurophysiological evaluation of both sensory and motor nerve conduction was performed. Subsequently, mice were sacrificed for messenger RNA, protein, and histological analysis of the dorsal root ganglia, spinal cord, and cerebellum. RESULTS: Cytokine administration resulted in significant reversal of biochemical, neuropathological, neurophysiological, and behavioural deficits associated with Friedreich's ataxia. Both G-CSF and SCF had pronounced effects on frataxin levels (the primary molecular defect in the pathogenesis of the disease) and a regulators of frataxin expression. Sustained improvements in motor coordination and locomotor activity were observed, even after onset of neurological symptoms. Treatment also restored the duration of sensory nerve compound potentials. Improvements in peripheral nerve conduction positively correlated with cytokine-induced increases in frataxin expression, providing a link between increases in frataxin and neurophysiological function. Abrogation of disease-related pathology was also evident, with reductions in inflammation/gliosis and increased neural stem cell numbers in areas of tissue injury. INTERPRETATION: These experiments show that cytokines already clinically used in other conditions offer the prospect of a novel, rapidly translatable, disease-modifying, and neuroprotective treatment for Friedreich's ataxia. Ann Neurol 2017;81:212-226.


Subject(s)
Behavior, Animal/drug effects , Friedreich Ataxia/drug therapy , Granulocyte Colony-Stimulating Factor/pharmacology , Iron-Binding Proteins/metabolism , Neural Conduction/drug effects , Neuroprotective Agents/pharmacology , Peripheral Nerves/drug effects , Stem Cell Factor/pharmacology , Animals , Disease Models, Animal , Friedreich Ataxia/metabolism , Friedreich Ataxia/physiopathology , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroprotective Agents/administration & dosage , Stem Cell Factor/administration & dosage , Frataxin
11.
Biomark Med ; 10(8): 889-902, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27416337

ABSTRACT

AIM: To provide an up-to-date review of oxidative stress biomarkers in multiple sclerosis and thus identify candidate molecules with greatest promise as biomarkers of diagnosis, disease activity or prognosis. METHOD: A semi-systematic literature search using PubMed and other databases. RESULTS: Nitric oxide metabolites, superoxide dismutase, catalase, glutathione reductase, inducible nitric oxide synthase, protein carbonyl, 3-nitrotyrosine, isoprostanes, malondialdehyde and products of DNA oxidation have been identified across multiple studies as having promise as diagnostic, therapeutic or prognostic markers in MS. CONCLUSION: Heterogeneity of study design, particularly patient selection, limits comparability across studies. Further cohort studies are needed, and we would recommend promising markers be incorporated into future clinical trials to prospectively validate their potential.


Subject(s)
Biomarkers/analysis , Multiple Sclerosis/diagnosis , Oxidative Stress , Biomarkers/blood , Biomarkers/cerebrospinal fluid , DNA/chemistry , Humans , Malondialdehyde/blood , Malondialdehyde/cerebrospinal fluid , Multiple Sclerosis/pathology , Nitric Oxide/blood , Nitric Oxide/cerebrospinal fluid , Nitric Oxide Synthase Type II/blood , Nitric Oxide Synthase Type II/cerebrospinal fluid , Protein Carbonylation , Superoxide Dismutase/blood , Superoxide Dismutase/cerebrospinal fluid
12.
Brain Res ; 1642: 452-460, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27086975

ABSTRACT

Cerebellar dysfunction is a significant contributor to disability in multiple sclerosis (MS). Both white matter (WM) and grey matter (GM) injury occurs within MS cerebellum and, within GM, demyelination, inflammatory cell infiltration and neuronal injury contribute to on-going pathology. The precise nature of cerebellar GM injury is, however, unknown. Oxidative stress pathways with ultimate lipid peroxidation and cell membrane injury occur extensively in MS and the purpose of this study was to investigate these processes in MS cerebellar GM. Post-mortem human cerebellar GM from MS and control subjects was analysed immunohistochemically, followed by semi-quantitative analysis of markers of cellular injury, lipid peroxidation and anti-oxidant enzyme expression. We have shown evidence for reduction in myelin and neuronal markers in MS GM, coupled to an increase in expression of a microglial marker. We also show that the lipid peroxidation product 4-hydroxynonenal co-localises with myelin and its levels negatively correlate to myelin basic protein levels. Furthermore, superoxide dismutase (SOD1 and 2) enzymes, localised within cerebellar neurons, are up-regulated, yet the activation of subsequent enzymes responsible for the detoxification of hydrogen peroxide, catalase and glutathione peroxidase are relatively deficient. These studies provide evidence for oxidative injury in MS cerebellar GM and further help define disease mechanisms within the MS brain.


Subject(s)
Cerebellum/metabolism , Cerebellum/pathology , Gray Matter/metabolism , Gray Matter/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Adult , Aged , Aged, 80 and over , Aldehydes/metabolism , Female , Humans , Lipid Peroxidation/physiology , Male , Microglia/metabolism , Microglia/pathology , Middle Aged , Myelin Basic Protein/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neurons/metabolism , Neurons/pathology , Oxidative Stress/physiology , RNA, Messenger/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism , White Matter/metabolism , White Matter/pathology
13.
Methods Mol Biol ; 1313: 165-84, 2015.
Article in English | MEDLINE | ID: mdl-25947664

ABSTRACT

It has emerged that cells which typically reside in the bone marrow have the capacity to cross the blood brain barrier and contribute genetic material to a range of neuronal cell types within the central nervous system. One such mechanism to account for this phenomenon is cellular fusion, occurring between migrating bone marrow-derived stem cells and neuronal cells in-situ. Biologically, the significance as to why cells from distinct lineages fuse with cells of the central nervous system is, as yet, unclear. Growing evidence however suggests that these cell fusion events could provide an efficient means of rescuing the highly complex and differentiated neuronal cell types that cannot be replaced in adulthood. To facilitate further understanding of cell fusion within the central nervous system, we describe here a technique to establish chimeric mice that are stably reconstituted with green fluorescent protein expressing sex-mismatched bone marrow. These chimeric mice are known to represent an excellent model for studying bone marrow cell migration and infiltration throughout the body, while in parallel, as will be described here, also provide a means to neatly analyze both bone marrow-derived cell fusion and trans-differentiation events within the central nervous system.


Subject(s)
Bone Marrow Cells/cytology , Cell Fusion , Central Nervous System/cytology , Chimerism , Hybrid Cells , Animals , Cell Differentiation , Cell Fusion/methods , Cell Separation , Female , Fluorescent Antibody Technique , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence , Male , Mice , Microscopy, Confocal
14.
J Neurosci Res ; 93(6): 882-92, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25639260

ABSTRACT

Axonal injury is often characterized by axonal transport defects and abnormal accumulation of intra-axonal components. Nitric oxide (NO) has a key role in mediating inflammatory axonopathy in many neurodegenerative diseases, but little is known about how nitrosative/oxidative stress affects axonal transport or whether reductions in kinesin superfamily protein (KIF) expression correlate with axon pathology. KIFs are molecular motors that have a key role in axonal and dendritic transport, and impairment of these mechanisms has been associated with a number of neurological disorders. This study shows that rat cortical neurons exposed to NO display both a time-dependent decrease in KIF gene/protein expression and neurofilament phosphorylation in addition to a reduction in axonal length and neuronal survival. Because mesenchymal stem cells (MSCs) represent a promising therapeutic candidate for neuronal/axonal repair, this study analyzes the capacity of MSCs to protect neurons and axonal transport mechanisms from NO damage. Results show that coculture of MSCs with NO-exposed neurons results in the preservation of KIF expression, axonal length, and neuronal survival. Altogether, these results suggest a potential mechanism involved in the disruption of axonal transport and abnormal accumulation of proteins in axons during nitrosative insult. We hypothesize that impaired axonal transport contributes, per se, to progression of injury and provide further evidence of the therapeutic potential of MSCs for neurodegenerative disorders.


Subject(s)
Axons/pathology , Free Radical Scavengers/toxicity , Gene Expression Regulation/drug effects , Kinesins/metabolism , Neurons/drug effects , Nitric Oxide/toxicity , Analysis of Variance , Animals , Antigens, CD/metabolism , Axons/drug effects , Bone Marrow Cells/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Embryo, Mammalian , Humans , Kinesins/genetics , Neurofilament Proteins/metabolism , Rats , Time Factors
15.
Brain Pathol ; 25(6): 692-700, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25411024

ABSTRACT

Cerebellar ataxia commonly occurs in multiple sclerosis, particularly in chronic progressive disease. Previous reports have highlighted both white matter and grey matter pathological changes within the cerebellum; and demyelination and inflammatory cell infiltrates appear commonly. As Purkinje cell axons are the sole output of the cerebellar cortex, understanding pathologic processes within these cells is crucial to develop strategies to prevent their loss and thus reduce ataxia. We studied pathologic changes occurring within Purkinje cells of the cerebellum. Using immunohistochemic techniques, we found changes in neurofilament phosphorylation states within Purkinje cells, including loss of dephosphorylated neurofilament and increased phosphorylated and hyperphosphorylated neurofilament. We also found Purkinje axonal spheroids and Purkinje cell loss, both of which occurred predominantly within areas of leucocortical demyelination within the cerebellar cortex. These changes have important implications for the study of cerebellar involvement in multiple sclerosis and may help design therapies to reduce the burden of ataxia in the condition.


Subject(s)
Cerebellum/pathology , Multiple Sclerosis/pathology , Purkinje Cells/pathology , Aged , Aged, 80 and over , Axons/immunology , Axons/metabolism , Axons/pathology , Cell Death , Cerebellum/immunology , Cerebellum/metabolism , Cohort Studies , Female , Fluorescent Antibody Technique , Humans , Intermediate Filaments/immunology , Intermediate Filaments/metabolism , Male , Middle Aged , Multiple Sclerosis/metabolism , Phosphorylation , Purkinje Cells/immunology , Purkinje Cells/metabolism
16.
Brain Res ; 1559: 55-64, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24602691

ABSTRACT

Chronic demyelination, on-going inflammation, axonal loss and grey matter neuronal injury are likely pathological processes that contribute to disease progression in multiple sclerosis (MS). Although the precise contribution of each process and their aetiological substrates is not fully known, recent evidence has implicated oxidative damage as a major cause of tissue injury in MS. The degree of tissue injury caused by oxidative molecules, such as reactive oxygen species (ROS), is balanced by endogenous anti-oxidant enzymes which detoxify ROS. Understanding endogenous mechanisms which protect the brain against oxidative injury in MS is important, since enhancing anti-oxidant responses is a major therapeutic strategy for preventing irreversible tissue injury in the disease. Our aims were to determine expression and activity levels of the hydrogen peroxide-reducing enzyme catalase in MS grey matter (GM). In MS GM, a catalase enzyme activity was elevated compared to control GM. We measured catalase protein expression by immune dot-blotting and catalase mRNA by a real-time polymerase chain reaction (RT-PCR). Protein analysis studies showed a strong positive correlation between catalase and microglial marker IBA-1 in MS GM. In addition, calibration of catalase mRNA level with reference to the microglial-specific transcript AIF-1 revealed an increase in this transcript in MS. This was reflected by the extent of HLA-DR immunolabeling in MS GM which was significantly elevated compared to control GM. Collectively, these observations provide evidence that microglial catalase activity is elevated in MS grey matter and may be an important endogenous anti-oxidant defence mechanism in MS.


Subject(s)
Catalase/metabolism , Gray Matter/enzymology , Microglia/enzymology , Multiple Sclerosis/enzymology , Adult , Aged , Aged, 80 and over , Calcium-Binding Proteins , DNA-Binding Proteins/metabolism , Female , Gene Expression , Gray Matter/pathology , HLA-DR Antigens/metabolism , Humans , Male , Microfilament Proteins , Microglia/pathology , Middle Aged , Multiple Sclerosis/pathology , Multiple Sclerosis, Chronic Progressive/enzymology , Multiple Sclerosis, Chronic Progressive/pathology , Multiple Sclerosis, Relapsing-Remitting/enzymology , Multiple Sclerosis, Relapsing-Remitting/pathology , RNA, Messenger/metabolism
17.
Mult Scler ; 20(6): 651-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24067896

ABSTRACT

BACKGROUND: Peroxisomes are organelles in eukaryotic cells with multiple functions including the detoxification of reactive oxygen species, plasmalogen synthesis and ß-oxidation of fatty acids. Recent evidence has implicated peroxisomal dysfunction in models of multiple sclerosis (MS) disease progression. OBJECTIVES: Our aims were to determine whether there are changes in peroxisomes in MS grey matter (GM) compared to control GM. METHODS: We analysed cases of MS and control GM immunocytochemically to assess peroxisomal membrane protein (PMP70) and neuronal proteins. We examined the expression of ABCD3 (the gene that encodes PMP70) in MS and control GM. Analyses of very long chain fatty acid (VLCFA) levels in GM were performed. RESULTS: PMP70 immunolabelling of neuronal somata was significantly lower in MS GM compared to control. Calibration of ABCD3 gene expression with reference to glyceraldehyde 3-phsophate dehydrogenase (GAPDH) revealed overall decreases in expression in MS compared to controls. Mean PMP70 counts in involved MS GM negatively correlated to disease duration. Elevations in C26:0 (hexacosanoic acid) were found in MS GM. CONCLUSIONS: Collectively, these observations provide evidence that there is an overall reduction in peroxisomal gene expression and peroxisomal proteins in GM neurons in MS. Changes in peroxisomal function may contribute to neuronal dysfunction and degeneration in MS.


Subject(s)
Gray Matter/pathology , Membrane Proteins/metabolism , Multiple Sclerosis/pathology , Neurons/pathology , Peroxisomes/pathology , ATP-Binding Cassette Transporters/metabolism , Adult , Aged , Aged, 80 and over , Female , Gene Expression/physiology , Gray Matter/metabolism , Humans , Male , Middle Aged , Neurons/metabolism
18.
Mult Scler ; 19(2): 153-61, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22723571

ABSTRACT

BACKGROUND: Axonal loss and grey matter neuronal injury are pathological processes that contribute to disease progression in multiple sclerosis (MS). Axon damage has been associated with changes in the phosphorylation state of neurofilaments and the presence of axonal spheroids. Perikaryal accumulation of abnormally phosphorylated neurofilament proteins has been reported in some neurodegenerative diseases. OBJECTIVES: The objective of this article is to determine whether abnormally phosphorylated neurofilament accumulates in neuronal perikarya in demyelinated MS cortex. METHODS: We used an antibody to hyperphosphorylated neurofilament-H (SMI-34) to assess the level and distribution of this antigen in paraffin sections of cerebral cortex from cases of neuropathologically confirmed MS and controls. We also examined the relationship of neurofilament phosphorylation to cortical demyelination. RESULTS: The number of SMI-34-positive neuronal somata was significantly higher in the MS cortex than the control cortex. As a proportion of the total number of neurons present (i.e. taking account of neuronal loss), the proportion of SMI-34-positive neurons was also significantly higher in the demyelinated and non-demyelinated MS cortex than the control cortex. CONCLUSIONS: MS is associated with the widespread accumulation of hyperphosphorylated neurofilament protein in neuronal somata, with the most marked accumulation in regions of cortical demyelination. This aberrant localisation of hyperphosphorylated neurofilament protein may contribute to neuronal dysfunction and degeneration in MS patients.


Subject(s)
Cerebral Cortex/metabolism , Multiple Sclerosis/metabolism , Neurodegenerative Diseases/pathology , Neurofilament Proteins/metabolism , Adult , Aged , Aged, 80 and over , Axons/pathology , Blotting, Western , Cerebral Cortex/pathology , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Multiple Sclerosis/pathology , Neurofilament Proteins/immunology , Neurons/pathology , Paraffin Embedding , Phosphorylation , Tissue Banks
19.
Stem Cells Dev ; 21(11): 2026-35, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22132904

ABSTRACT

The discovery that mesenchymal stem cells (MSCs) secrete SOD3 may help explain studies in which MSCs have direct antioxidant activities both in vivo and in vitro. SOD3 is an antioxidant enzyme that dismutes toxic free radicals produced during inflammatory processes. Therefore, MSC production and secretion of active and therapeutically significant levels of SOD3 would further support the use of MSCs as a cellular based antioxidant therapy. The aim of this study was therefore to investigate in vitro if MSC differentiation down the adipogenic, chondrogenic, and osteogenic lineages influences the expression of the antioxidant molecule SOD3. Human bone marrow MSCs and their differentiated progeny were cultured under standard conditions and both the SOD3 gene and protein expression examined. Following adipogenesis, cultures demonstrated that both SOD3 protein and gene expression are significantly increased, and conversely, following chondrogenesis SOD3 protein and gene expression is significantly decreased. Following osteogenesis there were no significant changes in SOD3 protein or gene expression. This in vitro study describes the initial characterization of SOD3 expression and secretion by differentiated MSCs. This should help guide further in vivo work establishing the therapeutic and antioxidative potential of MSC and their differentiated progeny.


Subject(s)
Bone Marrow/metabolism , Cell Differentiation , Mesenchymal Stem Cells/enzymology , Superoxide Dismutase/metabolism , Adipogenesis , Antioxidants/metabolism , Cell Lineage , Cells, Cultured , Chondrogenesis , Enzyme Activation , Enzyme Assays , Gene Expression Regulation, Enzymologic , Humans , Mesenchymal Stem Cells/cytology , Osteogenesis , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics
20.
PLoS One ; 6(10): e26098, 2011.
Article in English | MEDLINE | ID: mdl-22016819

ABSTRACT

Dramatic advances in recent decades in understanding the genetics of Friedreich ataxia (FRDA)--a GAA triplet expansion causing greatly reduced expression of the mitochondrial protein frataxin--have thus far yielded no therapeutic dividend, since there remain no effective treatments that prevent or even slow the inevitable progressive disability in affected individuals. Clinical interventions that restore frataxin expression are attractive therapeutic approaches, as, in theory, it may be possible to re-establish normal function in frataxin deficient cells if frataxin levels are increased above a specific threshold. With this in mind several drugs and cytokines have been tested for their ability to increase frataxin levels. Cell transplantation strategies may provide an alternative approach to this therapeutic aim, and may also offer more widespread cellular protective roles in FRDA. Here we show a direct link between frataxin expression in fibroblasts derived from FRDA patients with both decreased expression of hydrogen peroxide scavenging enzymes and increased sensitivity to hydrogen peroxide-mediated toxicity. We demonstrate that normal human mesenchymal stem cells (MSCs) induce both an increase in frataxin gene and protein expression in FRDA fibroblasts via secretion of soluble factors. Finally, we show that exposure to factors produced by human MSCs increases resistance to hydrogen peroxide-mediated toxicity in FRDA fibroblasts through, at least in part, restoring the expression of the hydrogen peroxide scavenging enzymes catalase and glutathione peroxidase 1. These findings suggest, for the first time, that stem cells may increase frataxin levels in FRDA and transplantation of MSCs may offer an effective treatment for these patients.


Subject(s)
Fibroblasts/enzymology , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Gene Expression Regulation , Hydrogen Peroxide/metabolism , Iron-Binding Proteins/metabolism , Mesenchymal Stem Cells/cytology , Bone Marrow Cells/cytology , Case-Control Studies , Catalase/metabolism , Culture Media, Conditioned/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Free Radical Scavengers/metabolism , Friedreich Ataxia/enzymology , Friedreich Ataxia/surgery , Gene Expression Regulation/drug effects , Glutathione Peroxidase/metabolism , Humans , Hydrogen Peroxide/toxicity , Iron-Binding Proteins/genetics , Mesenchymal Stem Cell Transplantation , Glutathione Peroxidase GPX1 , Frataxin
SELECTION OF CITATIONS
SEARCH DETAIL
...