Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
2.
Biomedicines ; 11(5)2023 May 05.
Article in English | MEDLINE | ID: mdl-37239045

ABSTRACT

BACKGROUND: Some neurodegenerative diseases have an element of neuroinflammation that is triggered by viral nucleic acids, resulting in the generation of type I interferons. In the cGAS-STING pathway, microbial and host-derived DNA bind and activate the DNA sensor cGAS, and the resulting cyclic dinucleotide, 2'3-cGAMP, binds to a critical adaptor protein, stimulator of interferon genes (STING), which leads to activation of downstream pathway components. However, there is limited work demonstrating the activation of the cGAS-STING pathway in human neurodegenerative diseases. METHODS: Post-mortem CNS tissue from donors with multiple sclerosis (n = 4), Alzheimer's disease (n = 6), Parkinson's disease (n = 3), amyotrophic lateral sclerosis (n = 3) and non-neurodegenerative controls (n = 11) were screened by immunohistochemistry for STING and relevant protein aggregates (e.g., amyloid-ß, α-synuclein, TDP-43). Human brain endothelial cells were cultured and stimulated with the STING agonist palmitic acid (1-400 µM) and assessed for mitochondrial stress (release of mitochondrial DNA into cytosol, increased oxygen consumption), downstream regulator factors, TBK-1/pIRF3 and inflammatory biomarker interferon-ß release and changes in ICAM-1 integrin expression. RESULTS: In neurodegenerative brain diseases, elevated STING protein was observed mainly in brain endothelial cells and neurons, compared to non-neurodegenerative control tissues where STING protein staining was weaker. Interestingly, a higher STING presence was associated with toxic protein aggregates (e.g., in neurons). Similarly high STING protein levels were observed within acute demyelinating lesions in multiple sclerosis subjects. To understand non-microbial/metabolic stress activation of the cGAS-STING pathway, brain endothelial cells were treated with palmitic acid. This evoked mitochondrial respiratory stress up to a ~2.5-fold increase in cellular oxygen consumption. Palmitic acid induced a statistically significant increase in cytosolic DNA leakage from endothelial cell mitochondria (Mander's coefficient; p < 0.05) and a significant increase in TBK-1, phosphorylated transcription factor IFN regulatory factor 3, cGAS and cell surface ICAM. In addition, a dose response in the secretion of interferon-ß was observed, but it failed to reach statistical significance. CONCLUSIONS: The histological evidence shows that the common cGAS-STING pathway appears to be activated in endothelial and neural cells in all four neurodegenerative diseases examined. Together with the in vitro data, this suggests that the STING pathway might be activated via perturbation of mitochondrial stress and DNA leakage, resulting in downstream neuroinflammation; hence, this pathway may be a target for future STING therapeutics.

3.
Nature ; 603(7899): 131-137, 2022 03.
Article in English | MEDLINE | ID: mdl-35197628

ABSTRACT

Variants of UNC13A, a critical gene for synapse function, increase the risk of amyotrophic lateral sclerosis and frontotemporal dementia1-3, two related neurodegenerative diseases defined by mislocalization of the RNA-binding protein TDP-434,5. Here we show that TDP-43 depletion induces robust inclusion of a cryptic exon in UNC13A, resulting in nonsense-mediated decay and loss of UNC13A protein. Two common intronic UNC13A polymorphisms strongly associated with amyotrophic lateral sclerosis and frontotemporal dementia risk overlap with TDP-43 binding sites. These polymorphisms potentiate cryptic exon inclusion, both in cultured cells and in brains and spinal cords from patients with these conditions. Our findings, which demonstrate a genetic link between loss of nuclear TDP-43 function and disease, reveal the mechanism by which UNC13A variants exacerbate the effects of decreased TDP-43 function. They further provide a promising therapeutic target for TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , TDP-43 Proteinopathies , Alternative Splicing , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Codon, Nonsense , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Humans , Nerve Tissue Proteins , Polymorphism, Single Nucleotide/genetics
4.
Acta Neuropathol ; 143(3): 383-401, 2022 03.
Article in English | MEDLINE | ID: mdl-34961893

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is a group of heterogeneous neurodegenerative disorders affecting the frontal and temporal lobes of the brain. Nuclear loss and cytoplasmic aggregation of the RNA-binding protein TDP-43 represents the major FTLD pathology, known as FTLD-TDP. To date, there is no effective treatment for FTLD-TDP due to an incomplete understanding of the molecular mechanisms underlying disease development. Here we compared postmortem tissue RNA-seq transcriptomes from the frontal cortex, temporal cortex, and cerebellum between 28 controls and 30 FTLD-TDP patients to profile changes in cell-type composition, gene expression and transcript usage. We observed downregulation of neuronal markers in all three regions of the brain, accompanied by upregulation of microglia, astrocytes, and oligodendrocytes, as well as endothelial cells and pericytes, suggesting shifts in both immune activation and within the vasculature. We validate our estimates of neuronal loss using neuropathological atrophy scores and show that neuronal loss in the cortex can be mainly attributed to excitatory neurons, and that increases in microglial and endothelial cell expression are highly correlated with neuronal loss. All our analyses identified a strong involvement of the cerebellum in the neurodegenerative process of FTLD-TDP. Altogether, our data provides a detailed landscape of gene expression alterations to help unravel relevant disease mechanisms in FTLD.


Subject(s)
Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Brain/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endothelial Cells/pathology , Frontotemporal Dementia/pathology , Frontotemporal Lobar Degeneration/pathology , Humans , Transcriptome
5.
J Clin Invest ; 132(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-34874913

ABSTRACT

Oligodendrocytes are the primary target of demyelinating disorders, and progressive neurodegenerative changes may evolve in the CNS. DNA damage and oxidative stress are considered key pathogenic events, but the underlying molecular mechanisms remain unclear. Moreover, animal models do not fully recapitulate human diseases, complicating the path to effective treatments. Here we report that mice with cell-autonomous deletion of the nuclear COP9 signalosome component CSN5 (JAB1) in oligodendrocytes develop DNA damage and defective DNA repair in myelinating glial cells. Interestingly, oligodendrocytes lacking JAB1 expression underwent a senescence-like phenotype that fostered chronic inflammation and oxidative stress. These mutants developed progressive CNS demyelination, microglia inflammation, and neurodegeneration, with severe motor deficits and premature death. Notably, blocking microglia inflammation did not prevent neurodegeneration, whereas the deletion of p21CIP1 but not p16INK4a pathway ameliorated the disease. We suggest that senescence is key to sustaining neurodegeneration in demyelinating disorders and may be considered a potential therapeutic target.


Subject(s)
Aging/metabolism , COP9 Signalosome Complex/deficiency , Gene Deletion , Neurodegenerative Diseases/metabolism , Oligodendroglia/metabolism , Peptide Hydrolases/deficiency , Aging/genetics , Aging/pathology , Animals , COP9 Signalosome Complex/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Oligodendroglia/pathology , Peptide Hydrolases/metabolism
6.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Article in English | MEDLINE | ID: mdl-34183414

ABSTRACT

Demyelination is a key pathogenic feature of multiple sclerosis (MS). Here, we evaluated the astrocyte contribution to myelin loss and focused on the neurotrophin receptor TrkB, whose up-regulation on the astrocyte finely demarcated chronic demyelinated areas in MS and was paralleled by neurotrophin loss. Mice lacking astrocyte TrkB were resistant to demyelination induced by autoimmune or toxic insults, demonstrating that TrkB signaling in astrocytes fostered oligodendrocyte damage. In vitro and ex vivo approaches highlighted that astrocyte TrkB supported scar formation and glia proliferation even in the absence of neurotrophin binding, indicating TrkB transactivation in response to inflammatory or toxic mediators. Notably, our neuropathological studies demonstrated copper dysregulation in MS and model lesions and TrkB-dependent expression of copper transporter (CTR1) on glia cells during neuroinflammation. In vitro experiments evidenced that TrkB was critical for the generation of glial intracellular calcium flux and CTR1 up-regulation induced by stimuli distinct from neurotrophins. These events led to copper uptake and release by the astrocyte, and in turn resulted in oligodendrocyte loss. Collectively, these data demonstrate a pathogenic demyelination mechanism via the astrocyte release of copper and open up the possibility of restoring copper homeostasis in the white matter as a therapeutic target in MS.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Copper/metabolism , Multiple Sclerosis/metabolism , Animals , Biological Transport , Chronic Disease , Cicatrix/pathology , Cuprizone , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Humans , Inflammation/pathology , Ligands , Membrane Transport Proteins/metabolism , Mice, Knockout , Myelin Sheath/metabolism , Nerve Growth Factors/metabolism , Receptor, trkB/metabolism , Up-Regulation , White Matter/pathology
7.
Brain Pathol ; 31(4): e12937, 2021 07.
Article in English | MEDLINE | ID: mdl-33576079

ABSTRACT

Histopathological analysis of tissue sections is invaluable in neurodegeneration research. However, cell-to-cell variation in both the presence and severity of a given phenotype is a key limitation of this approach, reducing the signal to noise ratio and leaving unresolved the potential of single-cell scoring for a given disease attribute. Here, we tested different machine learning methods to analyse high-content microscopy measurements of hundreds of motor neurons (MNs) from amyotrophic lateral sclerosis (ALS) post-mortem tissue sections. Furthermore, we automated the identification of phenotypically distinct MN subpopulations in VCP- and SOD1-mutant transgenic mice, revealing common morphological cellular phenotypes. Additionally we established scoring metrics to rank cells and tissue samples for both disease probability and severity. By adapting this paradigm to human post-mortem tissue, we validated our core finding that morphological descriptors robustly discriminate ALS from control healthy tissue at single cell resolution. Determining disease presence, severity and unbiased phenotypes at single cell resolution might prove transformational in our understanding of ALS and neurodegeneration more broadly.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Spinal Cord/pathology , Animals , Mice , Mice, Transgenic , Mitochondria/pathology , Motor Neurons/metabolism , Phenotype , Superoxide Dismutase/metabolism
8.
FASEB J ; 34(12): 16662-16675, 2020 12.
Article in English | MEDLINE | ID: mdl-33124722

ABSTRACT

We previously showed that calnexin (Canx)-deficient mice are desensitized to experimental autoimmune encephalomyelitis (EAE) induction, a model that is frequently used to study inflammatory demyelinating diseases, due to increased resistance of the blood-brain barrier to immune cell transmigration. We also discovered that Fabp5, an abundant cytoplasmic lipid-binding protein found in brain endothelial cells, makes protein-protein contact with the cytoplasmic C-tail domain of Canx. Remarkably, both Canx-deficient and Fabp5-deficient mice commonly manifest resistance to EAE induction. Here, we evaluated the importance of Fabp5/Canx interactions on EAE pathogenesis and on the patency of a model blood-brain barrier to T-cell transcellular migration. The results demonstrate that formation of a complex comprised of Fabp5 and the C-tail domain of Canx dictates the permeability of the model blood-brain barrier to immune cells and is also a prerequisite for EAE pathogenesis.


Subject(s)
Calnexin/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fatty Acid-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Line , Cell Movement/physiology , Disease Models, Animal , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Permeability
10.
J Clin Invest ; 130(11): 6080-6092, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32790644

ABSTRACT

No treatment for frontotemporal dementia (FTD), the second most common type of early-onset dementia, is available, but therapeutics are being investigated to target the 2 main proteins associated with FTD pathological subtypes: TDP-43 (FTLD-TDP) and tau (FTLD-tau). Testing potential therapies in clinical trials is hampered by our inability to distinguish between patients with FTLD-TDP and FTLD-tau. Therefore, we evaluated truncated stathmin-2 (STMN2) as a proxy of TDP-43 pathology, given the reports that TDP-43 dysfunction causes truncated STMN2 accumulation. Truncated STMN2 accumulated in human induced pluripotent stem cell-derived neurons depleted of TDP-43, but not in those with pathogenic TARDBP mutations in the absence of TDP-43 aggregation or loss of nuclear protein. In RNA-Seq analyses of human brain samples from the NYGC ALS cohort, truncated STMN2 RNA was confined to tissues and disease subtypes marked by TDP-43 inclusions. Last, we validated that truncated STMN2 RNA was elevated in the frontal cortex of a cohort of patients with FTLD-TDP but not in controls or patients with progressive supranuclear palsy, a type of FTLD-tau. Further, in patients with FTLD-TDP, we observed significant associations of truncated STMN2 RNA with phosphorylated TDP-43 levels and an earlier age of disease onset. Overall, our data uncovered truncated STMN2 as a marker for TDP-43 dysfunction in FTD.


Subject(s)
DNA-Binding Proteins/metabolism , Frontal Lobe/metabolism , Frontotemporal Dementia/metabolism , Induced Pluripotent Stem Cells/metabolism , Stathmin/metabolism , Biomarkers/metabolism , DNA-Binding Proteins/genetics , Female , Frontal Lobe/pathology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Male , Middle Aged , Mutation , Stathmin/genetics
11.
Mult Scler Relat Disord ; 43: 102084, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32442882

ABSTRACT

BACKGROUND: Thousands of people with multiple sclerosis (MS) have used self-administered oxygen therapy in the UK. Clinical trials have been performed, with scant evidence that people with MS have been consulted to explore how they benefit from or how to optimize this treatment. The conventional MS disease disability scores used in trials seldom reflect the effects individuals report when using oxygen therapy to treat their symptoms. METHODS: Three people with MS and the manager of an MS Centre formed a public involvement group and collaborated with clinicians and scientists to inform a lab-based study to investigate the physiological effects of oxygen therapy on microvascular brain endothelial cells. RESULTS: People with MS often use oxygen therapy at a later stage when their symptoms worsen and only after using other treatments. The frequency of oxygen therapy sessions and hyperbaric pressure is individualized and varies for people with MS. Despite direct comparisons of efficacy proving difficult, most individuals are exposed to 100% O2 at 1.5 atmosphere absolute (ATA; 1140 mmHg absolute) for 60 min. In a laboratory-based study human brain endothelial cells were exposed in vitro to 152 mmHg O2 for 60 min with and without pressure, as this equates to 20% O2 achievable via hyperbarics, which was then replicated at atmospheric pressure. A significant reduction in endothelial cells ICAM-1 (CD54) implicated in inflammatory cell margination across the blood brain barrier was observed under oxygen treatment. CONCLUSIONS: By collaborating with people living with MS, we were able to design laboratory-based experimental protocols that replicate their treatment regimens to advance our understanding of the physiological effects of hyperbaric oxygen treatment on brain cells and their role in neuroinflammation.


Subject(s)
Hyperbaric Oxygenation , Multiple Sclerosis , Brain , Endothelial Cells , Humans , Multiple Sclerosis/therapy , Oxygen
12.
Brain ; 143(2): 430-440, 2020 02 01.
Article in English | MEDLINE | ID: mdl-32040555

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disease caused by motor neuron loss, resulting in muscle wasting, paralysis and eventual death. A key pathological feature of ALS is cytoplasmically mislocalized and aggregated TDP-43 protein in >95% of cases, which is considered to have prion-like properties. Historical studies have predominantly focused on genetic forms of ALS, which represent ∼10% of cases, leaving the remaining 90% of sporadic ALS relatively understudied. Additionally, the role of astrocytes in ALS and their relationship with TDP-43 pathology is also not currently well understood. We have therefore used highly enriched human induced pluripotent stem cell (iPSC)-derived motor neurons and astrocytes to model early cell type-specific features of sporadic ALS. We first demonstrate seeded aggregation of TDP-43 by exposing human iPSC-derived motor neurons to serially passaged sporadic ALS post-mortem tissue (spALS) extracts. Next, we show that human iPSC-derived motor neurons are more vulnerable to TDP-43 aggregation and toxicity compared with their astrocyte counterparts. We demonstrate that these TDP-43 aggregates can more readily propagate from motor neurons into astrocytes in co-culture paradigms. We next found that astrocytes are neuroprotective to seeded aggregation within motor neurons by reducing (mislocalized) cytoplasmic TDP-43, TDP-43 aggregation and cell toxicity. Furthermore, we detected TDP-43 oligomers in these spALS spinal cord extracts, and as such demonstrated that highly purified recombinant TDP-43 oligomers can reproduce this observed cell-type specific toxicity, providing further support to a protein oligomer-mediated toxicity hypothesis in ALS. In summary, we have developed a human, clinically relevant, and cell-type specific modelling platform that recapitulates key aspects of sporadic ALS and uncovers both an initial neuroprotective role for astrocytes and the cell type-specific toxic effect of TDP-43 oligomers.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , DNA-Binding Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Cell Death/genetics , Cytoplasm/metabolism , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Spinal Cord/metabolism , Spinal Cord/pathology
13.
Brain ; 142(9): 2572-2580, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31368485

ABSTRACT

Mutations causing amyotrophic lateral sclerosis (ALS) clearly implicate ubiquitously expressed and predominantly nuclear RNA binding proteins, which form pathological cytoplasmic inclusions in this context. However, the possibility that wild-type RNA binding proteins mislocalize without necessarily becoming constituents of cytoplasmic inclusions themselves remains relatively unexplored. We hypothesized that nuclear-to-cytoplasmic mislocalization of the RNA binding protein fused in sarcoma (FUS), in an unaggregated state, may occur more widely in ALS than previously recognized. To address this hypothesis, we analysed motor neurons from a human ALS induced-pluripotent stem cell model caused by the VCP mutation. Additionally, we examined mouse transgenic models and post-mortem tissue from human sporadic ALS cases. We report nuclear-to-cytoplasmic mislocalization of FUS in both VCP-mutation related ALS and, crucially, in sporadic ALS spinal cord tissue from multiple cases. Furthermore, we provide evidence that FUS protein binds to an aberrantly retained intron within the SFPQ transcript, which is exported from the nucleus into the cytoplasm. Collectively, these data support a model for ALS pathogenesis whereby aberrant intron retention in SFPQ transcripts contributes to FUS mislocalization through their direct interaction and nuclear export. In summary, we report widespread mislocalization of the FUS protein in ALS and propose a putative underlying mechanism for this process.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Induced Pluripotent Stem Cells/metabolism , RNA-Binding Protein FUS/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Biomarkers/metabolism , Cell Nucleus/chemistry , Cell Nucleus/genetics , Cytoplasm/chemistry , Cytoplasm/genetics , Female , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA-Binding Protein FUS/analysis , RNA-Binding Protein FUS/genetics
14.
Nat Commun ; 9(1): 2010, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29789581

ABSTRACT

Mutations causing amyotrophic lateral sclerosis (ALS) strongly implicate ubiquitously expressed regulators of RNA processing. To understand the molecular impact of ALS-causing mutations on neuronal development and disease, we analysed transcriptomes during in vitro differentiation of motor neurons (MNs) from human control and patient-specific VCP mutant induced-pluripotent stem cells (iPSCs). We identify increased intron retention (IR) as a dominant feature of the splicing programme during early neural differentiation. Importantly, IR occurs prematurely in VCP mutant cultures compared with control counterparts. These aberrant IR events are also seen in independent RNAseq data sets from SOD1- and FUS-mutant MNs. The most significant IR is seen in the SFPQ transcript. The SFPQ protein binds extensively to its retained intron, exhibits lower nuclear abundance in VCP mutant cultures and is lost from nuclei of MNs in mouse models and human sporadic ALS. Collectively, we demonstrate SFPQ IR and nuclear loss as molecular hallmarks of familial and sporadic ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Motor Neurons/metabolism , PTB-Associated Splicing Factor/genetics , RNA Splicing , Spinal Cord/metabolism , Valosin Containing Protein/genetics , Aged , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Differentiation , Disease Models, Animal , Exons , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Introns , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Motor Neurons/pathology , PTB-Associated Splicing Factor/metabolism , Primary Cell Culture , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Sequence Analysis, RNA , Spinal Cord/pathology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Valosin Containing Protein/metabolism
15.
JCI Insight ; 3(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29515033

ABSTRACT

In multiple sclerosis (MS), a demyelinating inflammatory disease of the CNS, and its animal model (experimental autoimmune encephalomyelitis; EAE), circulating immune cells gain access to the CNS across the blood-brain barrier to cause inflammation, myelin destruction, and neuronal damage. Here, we discovered that calnexin, an ER chaperone, is highly abundant in human brain endothelial cells of MS patients. Conversely, mice lacking calnexin exhibited resistance to EAE induction, no evidence of immune cell infiltration into the CNS, and no induction of inflammation markers within the CNS. Furthermore, calnexin deficiency in mice did not alter the development or function of the immune system. Instead, the loss of calnexin led to a defect in brain endothelial cell function that resulted in reduced T cell trafficking across the blood-brain barrier. These findings identify calnexin in brain endothelial cells as a potentially novel target for developing strategies aimed at managing or preventing the pathogenic cascade that drives neuroinflammation and destruction of the myelin sheath in MS.


Subject(s)
Blood-Brain Barrier/immunology , Calnexin/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/pathology , Calnexin/genetics , Calnexin/immunology , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/immunology , Endothelial Cells/pathology , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Middle Aged , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , T-Lymphocytes/metabolism , Up-Regulation , White Matter/diagnostic imaging , White Matter/immunology , White Matter/pathology
16.
Nat Commun ; 8(1): 1164, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29079839

ABSTRACT

Astrocyte responses to neuronal injury may be beneficial or detrimental to neuronal recovery, but the mechanisms that determine these different responses are poorly understood. Here we show that ephrin type-B receptor 1 (EphB1) is upregulated in injured motor neurons, which in turn can activate astrocytes through ephrin-B1-mediated stimulation of signal transducer and activator of transcription-3 (STAT3). Transcriptional analysis shows that EphB1 induces a protective and anti-inflammatory signature in astrocytes, partially linked to the STAT3 network. This is distinct from the response evoked by interleukin (IL)-6 that is known to induce both pro inflammatory and anti-inflammatory processes. Finally, we demonstrate that the EphB1-ephrin-B1 pathway is disrupted in human stem cell derived astrocyte and mouse models of amyotrophic lateral sclerosis (ALS). Our work identifies an early neuronal help-me signal that activates a neuroprotective astrocytic response, which fails in ALS, and therefore represents an attractive therapeutic target.


Subject(s)
Astrocytes/cytology , Interleukin-6/metabolism , Neurons/metabolism , Receptor, EphB1/metabolism , STAT3 Transcription Factor/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Astrocytes/metabolism , Axons/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Motor Neurons/metabolism , Neuroprotection , Sciatic Nerve/metabolism , Signal Transduction , Transcriptome
17.
Ann Clin Transl Neurol ; 4(9): 663-679, 2017 09.
Article in English | MEDLINE | ID: mdl-28904988

ABSTRACT

OBJECTIVE: Conventional magnetic resonance imaging (MRI) of the multiple sclerosis spinal cord is limited by low specificity regarding the underlying pathological processes, and new MRI metrics assessing microscopic damage are required. We aim to show for the first time that neurite orientation dispersion (i.e., variability in axon/dendrite orientations) is a new biomarker that uncovers previously undetected layers of complexity of multiple sclerosis spinal cord pathology. Also, we validate against histology a clinically viable MRI technique for dispersion measurement (neurite orientation dispersion and density imaging, NODDI), to demonstrate the strong potential of the new marker. METHODS: We related quantitative metrics from histology and MRI in four post mortem spinal cord specimens (two controls; two progressive multiple sclerosis cases). The samples were scanned at high field, obtaining maps of neurite density and orientation dispersion from NODDI and routine diffusion tensor imaging (DTI) indices. Histological procedures provided markers of astrocyte, microglia, myelin and neurofilament density, as well as neurite dispersion. RESULTS: We report from both NODDI and histology a trend toward lower neurite dispersion in demyelinated lesions, indicative of reduced neurite architecture complexity. Also, we provide unequivocal evidence that NODDI-derived dispersion matches its histological counterpart (P < 0.001), while DTI metrics are less specific and influenced by several biophysical substrates. INTERPRETATION: Neurite orientation dispersion detects a previously undescribed and potentially relevant layer of microstructural complexity of multiple sclerosis spinal cord pathology. Clinically feasible techniques such as NODDI may play a key role in clinical trial and practice settings, as they provide histologically meaningful dispersion indices.

18.
Stem Cells Transl Med ; 6(10): 1840-1851, 2017 10.
Article in English | MEDLINE | ID: mdl-28941240

ABSTRACT

Vascular changes, including blood brain barrier destabilization, are common pathological features in multiple sclerosis (MS) lesions. Blood vessels within adult organs are reported to harbor mesenchymal stromal cells (MSCs) with phenotypical and functional characteristics similar to pericytes. We performed an immunohistochemical study of MSCs/pericytes in brain tissue from MS and healthy persons. Post-mortem brain tissue from patients with early progressive MS (EPMS), late stage progressive MS (LPMS), and healthy persons were analyzed for the MSC and pericyte markers CD146, platelet-derived growth factor receptor beta (PDGFRß), CD73, CD271, alpha-smooth muscle actin, and Ki67. The MS samples included active, chronic active, chronic inactive lesions, and normal-appearing white matter. MSC and pericyte marker localization were detected in association with blood vessels, including subendothelial CD146+ PDGFRß+ Ki67+ cells and CD73+ CD271+ PDGFRß+ Ki67- cells within the adventitia and perivascular areas. Both immunostained cell subpopulations were termed mesenchymal perivascular cells (MPCs). Quantitative analyses of immunostainings showed active lesions containing increased regions of CD146+ PDGFRß+ Ki67+ and CD73+ CD271+ PDGFRß+ Ki67- MPC subpopulations compared to inactive lesions. Chronic lesions presented with decreased levels of CD146+ PDGFRß+ Ki67+ MPC cells compared to control tissue. Furthermore, LPMS lesions displayed increased numbers of blood vessels harboring greatly enlarged CD73+ CD271+ adventitial and perivascular areas compared to control and EPMS tissue. In conclusion, we demonstrate the presence of MPC subgroups in control human brain vasculature, and their phenotypic changes in MS brain, which correlated with inflammation, demyelination and MS disease duration. Our findings demonstrate that brain-derived MPCs respond to pathologic mechanisms involved in MS disease progression and suggest that vessel-targeted therapeutics may benefit patients with progressive MS. Stem Cells Translational Medicine 2017;6:1840-1851.


Subject(s)
Blood Vessels/pathology , Brain/pathology , Mesenchymal Stem Cells/metabolism , Multiple Sclerosis/pathology , Pericytes/metabolism , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adult , Aged , Aged, 80 and over , Blood Vessels/metabolism , Brain/blood supply , CD146 Antigen/genetics , CD146 Antigen/metabolism , Female , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Mesenchymal Stem Cells/pathology , Middle Aged , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pericytes/pathology , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
19.
Neurobiol Dis ; 96: 236-247, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27590623

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is the most common form of motor neuron disease (MND), and >95% of familial and sporadic cases involve the deposition of insoluble aggregated, phosphorylated and cleaved TDP-43 protein. Accumulating clinical and biological evidence now indicates that ALS bears a number of similarities to the prion diseases, with TDP-43 acting as a misfolded 'prion-like' protein demonstrating similar underlying pathobiology. Here we systematically address the hypothesis that ALS is a prion-like disorder. First we demonstrate that TDP-43 demonstrates seeded polymerisation in vitro directly from both ALS brain and spinal cord. We next show that the seeding of TDP-43 results in the formation of characteristic insoluble, aggregated, and phosphorylated TDP-43 pathology that directly recapitulates the morphological diversity of TDP-43 inclusions detected in ALS patient CNS tissue. We next demonstrate that this reaction can be serially propagated to produce increasing amounts of phosphorylated TDP-43 pathology, and that aggregates can spread from cell to cell in an analogous fashion to that seen in the prion diseases. Finally, we reproduced our findings in a murine motor neuron-like cell line (NSC-34), where the seeding of TDP-43 induces the formation of TDP-43 oligomers and reduced cell viability. These findings may guide therapeutic strategies in this rapidly progressive and invariably fatal disease.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Brain/metabolism , DNA-Binding Proteins/metabolism , Prion Proteins/metabolism , Spinal Cord/metabolism , Animals , Autopsy , Brain/pathology , Coculture Techniques , DNA-Binding Proteins/genetics , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Motor Neurons/metabolism , Neuroblastoma/pathology , Spinal Cord/pathology , Transfection
20.
Glia ; 64(4): 553-69, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26649511

ABSTRACT

The mechanisms whereby human glial cells modulate local immune responses are not fully understood. Interleukin-27 (IL-27), a pleiotropic cytokine, has been shown to dampen the severity of experimental autoimmune encephalomyelitis, but it is still unresolved whether IL-27 plays a role in the human disease multiple sclerosis (MS). IL-27 contribution to local modulation of immune responses in the brain of MS patients was investigated. The expression of IL-27 subunits (EBI3 and p28) and its cognate receptor IL-27R (the gp130 and TCCR chains) was elevated within post-mortem MS brain lesions compared with normal control brains. Moreover, astrocytes (GFAP(+) cells) as well as microglia and macrophages (Iba1(+) cells) were important sources of IL-27. Brain-infiltrating CD4 and CD8 T lymphocytes expressed the IL-27R specific chain (TCCR) implying that these cells could respond to local IL-27 sources. In primary cultures of human astrocytes inflammatory cytokines increased IL-27 production, whereas myeloid cell inflammatory M1 polarization and inflammatory cytokines enhanced IL-27 expression in microglia and macrophages. Astrocytes in postmortem tissues and in vitro expressed IL-27R. Moreover, IL-27 triggered the phosphorylation of the transcription regulator STAT1, but not STAT3 in human astrocytes; indeed IL-27 up-regulated MHC class I expression on astrocytes in a STAT1-dependent manner. These findings demonstrated that IL-27 and its receptor were elevated in MS lesions and that local IL-27 can modulate immune properties of astrocytes and infiltrating immune cells. Thus, therapeutic strategies targeting IL-27 may influence not only peripheral but also local inflammatory responses within the brain of MS patients.


Subject(s)
Astrocytes/immunology , Brain/immunology , Interleukins/metabolism , Multiple Sclerosis/immunology , Myeloid Cells/immunology , Receptors, Interleukin/metabolism , Adult , Aged , Astrocytes/pathology , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Female , Humans , Macrophages/immunology , Macrophages/pathology , Male , Microglia/immunology , Microglia/pathology , Middle Aged , Multiple Sclerosis/pathology , Myeloid Cells/pathology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...