Imperial College London

Professor Richard Reynolds, BSc AKC PhD

Faculty of MedicineDepartment of Brain Sciences

Professor of Cellular Neurobiology
 
 
 
//

Contact

 

+44 (0)20 7594 6668r.reynolds

 
 
//

Location

 

E414Burlington DanesHammersmith Campus

//

Summary

 

Publications

Publication Type
Year
to

187 results found

Asimakidou E, Reynolds R, Barron AM, Lo CHet al., 2024, Autolysosomal acidification impairment as a mediator for TNFR1 induced neuronal necroptosis in Alzheimer's disease., Neural Regen Res, Vol: 19, Pages: 1869-1870, ISSN: 1673-5374

Journal article

Elkjaer ML, Hartebrodt A, Oubounyt M, Weber A, Vitved L, Reynolds R, Thomassen M, Rottger R, Baumbach J, Illes Zet al., 2024, Single-Cell Multi-Omics Map of Cell Type-Specific Mechanistic Drivers of Multiple Sclerosis Lesions., Neurol Neuroimmunol Neuroinflamm, Vol: 11

BACKGROUND AND OBJECTIVES: In progressive multiple sclerosis (MS), compartmentalized inflammation plays a pivotal role in the complex pathology of tissue damage. The interplay between epigenetic regulation, transcriptional modifications, and location-specific alterations within white matter (WM) lesions at the single-cell level remains underexplored. METHODS: We examined intracellular and intercellular pathways in the MS brain WM using a novel dataset obtained by integrated single-cell multi-omics techniques from 3 active lesions, 3 chronic active lesions, 3 remyelinating lesions, and 3 control WM of 6 patients with progressive MS and 3 non-neurologic controls. Single-nucleus RNA-seq and ATAC-seq were combined and additionally enriched with newly conducted spatial transcriptomics from 1 chronic active lesion. Functional gene modules were then validated in our previously published bulk tissue transcriptome data obtained from 73 WM lesions of patients with progressive MS and 25 WM of non-neurologic disease controls. RESULTS: Our analysis uncovered an MS-specific oligodendrocyte genetic signature influenced by the KLF/SP gene family. This modulation has potential associations with the autocrine iron uptake signaling observed in transcripts of transferrin and its receptor LRP2. In addition, an inflammatory profile emerged within these oligodendrocytes. We observed unique cellular endophenotypes both at the periphery and within the chronic active lesion. These include a distinct metabolic astrocyte phenotype, the importance of FGF signaling among astrocytes and neurons, and a notable enrichment of mitochondrial genes at the lesion edge populated predominantly by astrocytes. Our study also identified B-cell coexpression networks indicating different functional B-cell subsets with differential location and specific tendencies toward certain lesion types. DISCUSSION: The use of single-cell multi-omics has offered a detailed perspective into the cellular dynamics and interac

Journal article

Muraro P, 2024, Soluble CD27 is an intrathecal biomarker of T-cell-mediated lesion activity in multiple sclerosis, Journal of Neuroinflammation, ISSN: 1742-2094

Journal article

Calabrese M, Preziosa P, Scalfari A, Colato E, Marastoni D, Absinta M, Battaglini M, De Stefano N, Di Filippo M, Hametner S, Howell OW, Inglese M, Lassmann H, Martin R, Nicholas R, Reynolds R, Rocca MA, Tamanti A, Vercellino M, Villar LM, Filippi M, Magliozzi Ret al., 2024, Determinants and Biomarkers of Progression Independent of Relapses in Multiple Sclerosis., Ann Neurol

Clinical, pathological, and imaging evidence in multiple sclerosis (MS) suggests that a smoldering inflammatory activity is present from the earliest stages of the disease and underlies the progression of disability, which proceeds relentlessly and independently of clinical and radiological relapses (PIRA). The complex system of pathological events driving "chronic" worsening is likely linked with the early accumulation of compartmentalized inflammation within the central nervous system as well as insufficient repair phenomena and mitochondrial failure. These mechanisms are partially lesion-independent and differ from those causing clinical relapses and the formation of new focal demyelinating lesions; they lead to neuroaxonal dysfunction and death, myelin loss, glia alterations, and finally, a neuronal network dysfunction outweighing central nervous system (CNS) compensatory mechanisms. This review aims to provide an overview of the state of the art of neuropathological, immunological, and imaging knowledge about the mechanisms underlying the smoldering disease activity, focusing on possible early biomarkers and their translation into clinical practice. ANN NEUROL 2024.

Journal article

Nicholas R, Magliozzi R, Marastoni D, Howell O, Roncaroli F, Muraro P, Reynolds R, Friede Tet al., 2024, High Levels of Perivascular Inflammation and Active Demyelinating Lesions at Time of Death Associated with Rapidly Progressive Multiple Sclerosis Disease Course: A Retrospective Postmortem Cohort Study., Ann Neurol, Vol: 95, Pages: 706-719

OBJECTIVE: Analysis of postmortem multiple sclerosis (MS) tissues combined with in vivo disease milestones suggests that whereas perivascular white matter infiltrates are associated with demyelinating activity in the initial stages, leptomeningeal immune cell infiltration, enriched in B cells, and associated cortical lesions contribute to disease progression. We systematically examine the association of inflammatory features and white matter demyelination at postmortem with clinical milestones. METHODS: In 269 MS brains, 20 sites were examined using immunohistochemistry for active lesions (ALs) and perivenular inflammation (PVI). In a subset of 22, a detailed count of CD20+ B cells and CD3+ T cells in PVIs was performed. RESULTS: ALs were detected in 22%, whereas high levels of PVI were detected in 52% of cases. ALs were present in 35% of cases with high levels of PVI. Shorter time from onset of progression to death was associated with increased prevalence and higher levels of PVI (both p < 0.0001). Shorter time from onset of progression to wheelchair use was associated with higher prevalence of ALs (odds ratio [OR] = 0.921, 95% confidence interval [CI] = 0.858-0.989, p = 0.0230) and higher level of PVI (OR = 0.932, 95% CI = 0.886-0.981, p = 0.0071). High levels of PVI were associated with meningeal inflammation and increased cortical demyelination and significantly higher levels of B lymphocytes within the PVI. INTERPRETATION: ALs, a feature of early disease stage, persist up to death in a subgroup with high levels of PVI. These features link to a rapid progressive phase and higher levels of meningeal inflammation and B-cell infiltrates, supporting the hypothesis that chronic inflammation drives progression in MS. ANN NEUROL 2024;95:706-719.

Journal article

Knowles S, Middleton R, Cooze B, Farkas I, Leung YY, Allen K, Winslade M, Owen DRJ, Magliozzi R, Reynolds R, Neal JW, Pearson O, Nicholas R, Pickrell WO, Howell OW, UK MS Register Research Groupet al., 2024, Comparing the pathology, clinical, and demographic characteristics of younger and older-onset multiple sclerosis, Annals of Neurology, Vol: 95, Pages: 471-486, ISSN: 0364-5134

OBJECTIVE: Older people with multiple sclerosis (MS) have a less active radiological and clinical presentation, but many still attain significant levels of disability; but what drives worsening disability in this group? METHODS: We used data from the UK MS Register to characterize demographics and clinical features of late-onset multiple sclerosis (LOMS; symptom onset at ≥50 years), compared with adult-onset MS (AOMS; onset 18-49 years). We performed a pathology study of a separate MS cohort with a later onset (n = 18, mean age of onset 54 years) versus AOMS (n = 23, mean age of onset 29 years). RESULTS: In the Register cohort, there were 1,608 (9.4%) with LOMS. When compared with AOMS, there was a lower proportion of women, a higher proportion of primary progressive MS, a higher level of disability at diagnosis (median MS impact scale 36.7 vs. 28.3, p < 0.001), and a higher proportion of gait-related initial symptoms. People with LOMS were less likely to receive a high efficacy disease-modifying treatment and attained substantial disability sooner. Controlling for age of death and sex, neuron density in the thalamus and pons decreased with onset-age, whereas actively demyelinating lesions and compartmentalized inflammation was greatest in AOMS. Only neuron density, and not demyelination or the extent of compartmentalized inflammation, correlated with disability outcomes in older-onset MS patients. INTERPRETATION: The more progressive nature of older-onset MS is associated with significant neurodegeneration, but infrequent inflammatory demyelination. These findings have implications for the assessment and treatment of MS in older people. ANN NEUROL 2023.

Journal article

Quick JD, Silva C, Wong JH, Lim KL, Reynolds R, Barron AM, Zeng J, Lo CHet al., 2023, Lysosomal acidification dysfunction in microglia: an emerging pathogenic mechanism of neuroinflammation and neurodegeneration, Journal of Neuroinflammation, Vol: 20, ISSN: 1742-2094

Microglia are the resident innate immune cells in the brain with a major role in orchestrating immune responses. They also provide a frontline of host defense in the central nervous system (CNS) through their active phagocytic capability. Being a professional phagocyte, microglia participate in phagocytic and autophagic clearance of cellular waste and debris as well as toxic protein aggregates, which relies on optimal lysosomal acidification and function. Defective microglial lysosomal acidification leads to impaired phagocytic and autophagic functions which result in the perpetuation of neuroinflammation and progression of neurodegeneration. Reacidification of impaired lysosomes in microglia has been shown to reverse neurodegenerative pathology in Alzheimer's disease. In this review, we summarize key factors and mechanisms contributing to lysosomal acidification impairment and the associated phagocytic and autophagic dysfunction in microglia, and how these defects contribute to neuroinflammation and neurodegeneration. We further discuss techniques to monitor lysosomal pH and therapeutic agents that can reacidify impaired lysosomes in microglia under disease conditions. Finally, we propose future directions to investigate the role of microglial lysosomal acidification in lysosome-mitochondria crosstalk and in neuron-glia interaction for more comprehensive understanding of its broader CNS physiological and pathological implications.

Journal article

Magliozzi R, Howell OW, Calabrese M, Reynolds Ret al., 2023, Meningeal inflammation as a driver of cortical grey matter pathology and clinical progression in multiple sclerosis, Nature Reviews Neurology, Vol: 19, Pages: 461-476, ISSN: 1759-4766

Growing evidence from cerebrospinal fluid samples and post-mortem brain tissue from individuals with multiple sclerosis (MS) and rodent models indicates that the meninges have a key role in the inflammatory and neurodegenerative mechanisms underlying progressive MS pathology. The subarachnoid space and associated perivascular spaces between the membranes of the meninges are the access points for entry of lymphocytes, monocytes and macrophages into the brain parenchyma, and the main route for diffusion of inflammatory and cytotoxic molecules from the cerebrospinal fluid into the brain tissue. In addition, the meningeal spaces act as an exit route for CNS-derived antigens, immune cells and metabolites. A number of studies have demonstrated an association between chronic meningeal inflammation and a more severe clinical course of MS, suggesting that the build-up of immune cell aggregates in the meninges represents a rational target for therapeutic intervention. Therefore, understanding the precise cell and molecular mechanisms, timing and anatomical features involved in the compartmentalization of inflammation within the meningeal spaces in MS is vital. Here, we present a detailed review and discussion of the cellular, molecular and radiological evidence for a role of meningeal inflammation in MS, alongside the clinical and therapeutic implications.

Journal article

Reynolds R, Mazarakis N, Picon C, Jayaraman A, Bates RJet al., 2023, Persistent Expression of TNFR1 Ligands in the Meninges as a Cause of Neurodegeneration in MS Cortical Grey Matter, Publisher: SAGE PUBLICATIONS LTD, Pages: NP9-NP9, ISSN: 1352-4585

Conference paper

Wang Q, Zheng J, Pettersson S, Reynolds R, Tan E-Ket al., 2023, The link between neuroinflammation and the neurovascular unit in synucleinopathies., Science Advances, Vol: 9, Pages: 1-16, ISSN: 2375-2548

The neurovascular unit (NVU) is composed of vascular cells, glial cells, and neurons. As a fundamental functional module in the central nervous system, the NVU maintains homeostasis in the microenvironment and the integrity of the blood-brain barrier. Disruption of the NVU and interactions among its components are involved in the pathophysiology of synucleinopathies, which are characterized by the pathological accumulation of α-synuclein. Neuroinflammation contributes to the pathophysiology of synucleinopathies, including Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. This review aims to summarize the neuroinflammatory response of glial cells and vascular cells in the NVU. We also review neuroinflammation in the context of the cross-talk between glial cells and vascular cells, between glial cells and pericytes, and between microglia and astroglia. Last, we discuss how α-synuclein affects neuroinflammation and how neuroinflammation influences the aggregation and spread of α-synuclein and analyze different properties of α-synuclein in synucleinopathies.

Journal article

Elkjaer ML, Simon L, Frisch T, Bente L-M, Kacprowski T, Thomassen M, Reynolds R, Baumbach J, Roettger R, Illes Zet al., 2022, Hypothesis of a potential BrainBiota and its relation to CNS autoimmune inflammation, FRONTIERS IN IMMUNOLOGY, Vol: 13, ISSN: 1664-3224

Journal article

Cooze B, Farkas I, Leung Y, Yadanar H, Gerhards K, Gveric D, Nicholas R, Neal JW, Owen DR, Reynolds R, Howell OWet al., 2022, Neuronal loss in the thalamus and pons correlates with disease severity: Interim analysis of a large digital pathology study, 38th Congress of the European-Committee-for-Treatment-and-Research-in-Multiple-Sclerosis, Publisher: SAGE PUBLICATIONS LTD, Pages: 494-494, ISSN: 1352-4585

Conference paper

Cooze BJ, Dickerson M, Loganathan R, Watkins LM, Grounds E, Pearson BR, Bevan RJ, Morgan BP, Magliozzi R, Reynolds R, Neal JW, Howell OWet al., 2022, The association between neurodegeneration and local complement activation in the thalamus to progressive multiple sclerosis outcome, BRAIN PATHOLOGY, Vol: 32, ISSN: 1015-6305

Journal article

Magliozzi R, Fadda G, Brown RA, Bar-Or A, Howell OW, Hametner S, Marastoni D, Poli A, Nicholas R, Calabrese M, Monaco S, Reynolds Ret al., 2022, "Ependymal-in" gradient of thalamic damage in progressive multiple sclerosis, Annals of Neurology, Vol: 92, Pages: 670-685, ISSN: 0364-5134

Leptomeningeal and perivenular infiltrates are important contributors to cortical grey matter damage and disease progression in multiple sclerosis (MS). Whereas perivenular inflammation induces vasculocentric lesions, leptomeningeal involvement follows a subpial “surface-in” gradient. To determine whether similar gradient of damage occurs in deep grey matter nuclei, we examined the dorsomedial thalamic nuclei and cerebrospinal fluid (CSF) samples from 41 postmortem secondary progressive MS cases compared with 5 non-neurological controls and 12 controls with other neurological diseases. CSF/ependyma-oriented gradient of reduction in NeuN+ neuron density was present in MS thalamic lesions compared to controls, greatest (26%) in subventricular locations at the ependyma/CSF boundary and least with increasing distance (12% at 10 mm). Concomitant graded reduction in SMI31+ axon density was observed, greatest (38%) at 2 mm from the ependyma/CSF boundary and least at 10 mm (13%). Conversely, gradient of major histocompatibility complex (MHC)-II+ microglia density increased by over 50% at 2 mm at the ependyma/CSF boundary and only by 15% at 10 mm and this gradient inversely correlated with the neuronal (R = −0.91, p < 0.0001) and axonal (R = −0.79, p < 0.0001) thalamic changes. Observed gradients were also detected in normal-appearing thalamus and were associated with rapid/severe disease progression; presence of leptomeningeal tertiary lymphoid-like structures; large subependymal infiltrates, enriched in CD20+ B cells and occasionally containing CXCL13+ CD35+ follicular dendritic cells; and high CSF protein expression of a complex pattern of soluble inflammatory/neurodegeneration factors, including chitinase-3-like-1, TNFR1, parvalbumin, neurofilament-light-chains and TNF. Substantial “ependymal-in” gradient of pathological cell alterations, accompanied by presence of in

Journal article

Bates REJ, Browne E, Schalks R, Jacobs H, Tan L, Parekh P, Magliozzi R, Calabrese M, Mazarakis ND, Reynolds Ret al., 2022, Lymphotoxin-alpha expression in the meninges causes lymphoid tissue formation and neurodegeneration, Brain: a journal of neurology, Vol: 145, Pages: 4287-4307, ISSN: 0006-8950

Organized meningeal immune cell infiltrates are suggested to play an important role in cortical grey matter pathology in the multiple sclerosis brain, but the mechanisms involved are as yet unresolved. Lymphotoxin-alpha plays a key role in lymphoid organ development and cellular cytotoxicity in the immune system and its expression is increased in the CSF of naïve and progressive multiple sclerosis patients and post-mortem meningeal tissue. Here we show that persistently increased levels of lymphotoxin-alpha in the cerebral meninges can give rise to lymphoid-like structures and underlying multiple sclerosis-like cortical pathology. Stereotaxic injections of recombinant lymphotoxin-alpha into the rat meninges led to acute meningeal inflammation and subpial demyelination that resolved after 28 days, with demyelination being dependent on prior subclinical immunization with myelin oligodendrocyte glycoprotein. Injection of a lymphotoxin-alpha lentiviral vector into the cortical meningeal space, to produce chronic localized overexpression of the cytokine, induced extensive lymphoid-like immune cell aggregates, maintained over 3 months, including T-cell rich zones containing podoplanin + fibroblastic reticular stromal cells and B-cell rich zones with a network of follicular dendritic cells, together with expression of lymphoid chemokines and their receptors. Extensive microglial and astroglial activation, subpial demyelination and marked neuronal loss occurred in the underlying cortical parenchyma. Whereas subpial demyelination was partially dependent on previous myelin oligodendrocyte glycoprotein immunization, the neuronal loss was present irrespective of immunization. Conditioned medium from LTα treated microglia was able to induce a reactive phenotype in astrocytes. Our results show that chronic lymphotoxin-alpha overexpression alone is sufficient to induce formation of meningeal lymphoid-like structures and subsequent neurodegeneration, similar to that seen

Journal article

Jayaraman A, Reynolds R, 2022, Diverse pathways to neuronal necroptosis in Alzheimer's disease, European Journal of Neuroscience, Vol: 56, Pages: 5428-5441, ISSN: 0953-816X

Necroptosis, or programmed necrosis, involves the kinase activity of receptor interacting kinases 1 and 3, the activation of the pseudokinase mixed lineage kinase domain-like and formation of a complex called the necrosome. It is one of the non-apoptotic cell death pathways that has gained interest in the recent years, especially as a neuronal cell death pathway occurring in Alzheimer's disease. In this review, we focus our discussion on the various molecular mechanisms that could trigger neuronal death through necroptosis and have been shown to play a role in Alzheimer's disease pathogenesis and neuroinflammation. We describe how each of these pathways, such as tumour necrosis factor signalling, reactive oxygen species, endosomal sorting complex, post-translational modifications and certain individual molecules, is dysregulated or activated in Alzheimer's disease, and how this dysregulation/activation could trigger necroptosis. At the cellular level, many of these molecular mechanisms and pathways may act in parallel to synergize with each other or inhibit one another, and changes in the balance between them may determine different cellular vulnerabilities at different disease stages. However, from a therapeutic standpoint, it remains unclear how best to target one or more of these pathways, given that such diverse pathways could all contribute to necroptotic cell death in Alzheimer's disease.

Journal article

Vanderdonckt P, Aloisi F, Comi G, de Bruyn A, Hartung H-P, Huitinga I, Kuhlmann T, Lucchinetti CF, Metz I, Reynolds R, Lassmann Het al., 2022, Tissue donations for multiple sclerosis research: current state and suggestions for improvement, BRAIN COMMUNICATIONS, Vol: 4

Journal article

Tu H, Zhang ZW, Qiu L, Lin Y, Jiang M, Chia S-Y, Wei Y, Ng ASL, Reynolds R, Tan E-K, Zeng Let al., 2022, Increased expression of pathological markers in Parkinson's disease dementia post-mortem brains compared to dementia with Lewy bodies., BMC Neurosci, Vol: 23, Pages: 3-3

BACKGROUND: Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are common age-related neurodegenerative diseases comprising Lewy body spectrum disorders associated with cortical and subcortical Lewy body pathology. Over 30% of PD patients develop PD dementia (PDD), which describes dementia arising in the context of established idiopathic PD. Furthermore, Lewy bodies frequently accompany the amyloid plaque and neurofibrillary tangle pathology of Alzheimer's disease (AD), where they are observed in the amygdala of approximately 60% of sporadic and familial AD. While PDD and DLB share similar pathological substrates, they differ in the temporal onset of motor and cognitive symptoms; however, protein markers to distinguish them are still lacking. METHODS: Here, we systematically studied a series of AD and PD pathogenesis markers, as well as mitochondria, mitophagy, and neuroinflammation-related indicators, in the substantia nigra (SN), temporal cortex (TC), and caudate and putamen (CP) regions of human post-mortem brain samples from individuals with PDD and DLB and condition-matched controls. RESULTS: We found that p-APPT668 (TC), α-synuclein (CP), and LC3II (CP) are all increased while the tyrosine hydroxylase (TH) (CP) is decreased in both PDD and DLB compared to control. Also, the levels of Aβ42 and DD2R, IBA1, and p-LRRK2S935 are all elevated in PDD compared to control. Interestingly, protein levels of p-TauS199/202 in CP and DD2R, DRP1, and VPS35 in TC are all increased in PDD compared to DLB. CONCLUSIONS: Together, our comprehensive and systematic study identified a set of signature proteins that will help to understand the pathology and etiology of PDD and DLB at the molecular level.

Journal article

Bouman P, Pitt D, Reich D, Schneider J, Bennett D, Nagra R, Reynolds R, Corboy J, De Jager Pet al., 2021, Towards a new resource for the MS brain: a cross-brain bank proteomic atlas of non-lesional neocortex, Publisher: SAGE PUBLICATIONS LTD, Pages: 333-334, ISSN: 1352-4585

Conference paper

Picon C, Robertas A, Wojewska M, Pezzini F, Magliozzi R, Nicholas R, Reynolds Ret al., 2021, Endosomal sorting complex III is dysregulated in cortical neurons in progressive MS and associated with necroptosis activation, Publisher: SAGE PUBLICATIONS LTD, Pages: 52-52, ISSN: 1352-4585

Conference paper

Magliozzi R, Tamanti A, Castellaro M, Nicholas R, Howell O, Bazan D, Pizzini FB, Mainero C, Hametner S, Lassmann H, Reynolds R, Monaco S, Calabrese Met al., 2021, Combined MRI and neuropathology pattern characterization of subpial cortical lesion activity in multiple sclerosis, Publisher: SAGE PUBLICATIONS LTD, Pages: 79-80, ISSN: 1352-4585

Conference paper

Jayaraman A, Htike TT, James R, Picon C, Reynolds Ret al., 2021, TNF-mediated neuroinflammation is linked to neuronal necroptosis in Alzheimer's disease hippocampus., Acta Neuropathologica Communications, Vol: 9, Pages: 159-159, ISSN: 2051-5960

The pathogenetic mechanisms underlying neuronal death and dysfunction in Alzheimer's disease (AD) remain unclear. However, chronic neuroinflammation has been implicated in stimulating or exacerbating neuronal damage. The tumor necrosis factor (TNF) superfamily of cytokines are involved in many systemic chronic inflammatory and degenerative conditions and are amongst the key mediators of neuroinflammation. TNF binds to the TNFR1 and TNFR2 receptors to activate diverse cellular responses that can be either neuroprotective or neurodegenerative. In particular, TNF can induce programmed necrosis or necroptosis in an inflammatory environment. Although activation of necroptosis has recently been demonstrated in the AD brain, its significance in AD neuron loss and the role of TNF signaling is unclear. We demonstrate an increase in expression of multiple proteins in the TNF/TNF receptor-1-mediated necroptosis pathway in the AD post-mortem brain, as indicated by the phosphorylation of RIPK3 and MLKL, predominantly observed in the CA1 pyramidal neurons. The density of phosphoRIPK3 + and phosphoMLKL + neurons correlated inversely with total neuron density and showed significant sexual dimorphism within the AD cohort. In addition, apoptotic signaling was not significantly activated in the AD brain compared to the control brain. Exposure of human iPSC-derived glutamatergic neurons to TNF increased necroptotic cell death when apoptosis was inhibited, which was significantly reversed by small molecule inhibitors of RIPK1, RIPK3, and MLKL. In the post-mortem AD brain and in human iPSC neurons, in response to TNF, we show evidence of altered expression of proteins of the ESCRT III complex, which has been recently suggested as an antagonist of necroptosis and a possible mechanism by which cells can survive after necroptosis has been triggered. Taken together, our results suggest that neuronal loss in AD is due to TNF-mediated necroptosis rather than apoptos

Journal article

Chew EGY, Heng YJ, Lian M, Tandiono M, Dempster E, Policicchio S, Mill J, Reynolds R, Foo JNet al., 2021, Interrogating Parkinson's disease associated mutations at single cell resolution, Publisher: WILEY, Pages: S307-S307, ISSN: 0885-3185

Conference paper

Pienaar IS, Mohammed R, Courtley R, Gledson MR, Reynolds R, Nicholas R, Elson JLet al., 2021, Investigation of the correlation between mildly deleterious mtDNA Variations and the clinical progression of multiple sclerosis, MULTIPLE SCLEROSIS AND RELATED DISORDERS, Vol: 53, ISSN: 2211-0348

Journal article

Jayaraman A, Htike TT, James R, Picon C, Reynolds Ret al., 2021, TNF-mediated neuroinflammation is linked to neuronal necroptosis in Alzheimer’s disease hippocampus

<jats:title>Abstract</jats:title><jats:p>The pathogenetic mechanisms underlying neuronal death and dysfunction in Alzheimer’s disease (AD) remain unclear. However, chronic neuroinflammation has been implicated in stimulating or exacerbating neuronal damage. The tumor necrosis factor (TNF) superfamily of cytokines are involved in many systemic chronic inflammatory and degenerative conditions and are amongst the key mediators of neuroinflammation. TNF binds to the TNFR1 and TNFR2 receptors to activate diverse cellular responses that can be either neuroprotective or neurodegenerative. In particular, TNF can induce programmed necrosis or necroptosis in an inflammatory environment. Although activation of necroptosis has recently been demonstrated in the AD brain, its significance in AD neuron loss and the role of TNF signaling is unclear. We demonstrate an increase in expression of multiple proteins in the TNF/TNF receptor-1-mediated necroptosis pathway in the AD post-mortem brain, as indicated by the phosphorylation of RIPK3 and MLKL, predominantly observed in the CA1 pyramidal neurons. The density of phosphoRIPK3+ and phosphoMLKL+ neurons correlated inversely with total neuron density and showed significant sexual dimorphism within the AD cohort. In addition, apoptotic signaling was not significantly activated in the AD brain compared to the control brain. Exposure of human iPSC-derived glutamatergic neurons to TNF increased necroptotic cell death when apoptosis was inhibited, which was significantly reversed by small molecule inhibitors of RIPK1, RIPK3, and MLKL. In the post-mortem AD brain and in human iPSC neurons to TNF, we show evidence of altered expression of proteins of the ESCRT III complex, which has been recently suggested as an antagonist of necroptosis and a possible mechanism by which cells can survive after necroptosis has been triggered. Taken together, our results suggest that neuronal loss in AD is due to TNF-mediated necroptos

Working paper

Magliozzi R, Pezzini F, Pucci M, Rossi S, Facchiano F, Marastoni D, Montagnana M, Lippi G, Reynolds R, Calabrese Met al., 2021, Changes in Cerebrospinal Fluid Balance of TNF and TNF Receptors in Naive Multiple Sclerosis Patients: Early Involvement in Compartmentalised Intrathecal Inflammation, CELLS, Vol: 10

Journal article

van Olst Y, Rodriguez-Mogeda C, Picon C, Kiljan S, James RE, Kamermans A, van der Pol SMA, Knoop L, Michailidou I, Drost E, Franssen M, Schenk GJ, Geurts JJG, Amor S, Mazarakis ND, van Horssen J, de Vries HE, Reynolds R, Witte MEet al., 2021, Meningeal inflammation in multiple sclerosis induces phenotypic changes in cortical microglia that differentially associate with neurodegeneration, ACTA NEUROPATHOLOGICA, Vol: 141, Pages: 881-899, ISSN: 0001-6322

Journal article

James RE, Farquharson AC, Jacobs HC, Farkas IC, Mazarakis N, Reynolds Ret al., 2021, The Role of Neuronal CXCL13 Chemokine Expression in Multiple Sclerosis Pathology, Publisher: SAGE PUBLICATIONS LTD, Pages: 104-105, ISSN: 1352-4585

Conference paper

Pardini M, Brown JWL, Magliozzi R, Reynolds R, Chard DTet al., 2021, Surface-in pathology in multiple sclerosis: a new view on pathogenesis?, BRAIN, Vol: 144, Pages: 1646-1654, ISSN: 0006-8950

Journal article

Wang Q, Luo Y, Chaudhuri KR, Reynolds R, Tan E-K, Pettersson Set al., 2021, The role of gut dysbiosis in Parkinson's disease: mechanistic insights and therapeutic options., Brain: a journal of neurology, Vol: 144, Pages: 2571-2593, ISSN: 0006-8950

Parkinson's disease is a common neurodegenerative disease in which gastrointestinal symptoms may appear prior to motor symptoms. The gut microbiota of patients with Parkinson's disease shows unique changes, which may be used as early biomarkers of disease. Alteration in gut microbiota composition may be related to the cause or effect of motor or non-motor symptoms, but the specific pathogenic mechanisms are unclear. The gut microbiota and its metabolites have been suggested to be involved in the pathogenesis of Parkinson's disease by regulating neuroinflammation, barrier function and neurotransmitter activity. There is bidirectional communication between the enteric nervous system and the central nervous system, and the microbiota-gut-brain axis may provide a pathway for the transmission of α-synuclein. We highlight recent discoveries and alterations of the gut microbiota in Parkinson's disease, and highlight current mechanistic insights on the microbiota-gut-brain axis in disease pathophysiology. We discuss the interactions between production and transmission of α-synuclein and gut inflammation and neuroinflammation. In addition, we also draw attention to diet modification, use of probiotics and prebiotics and fecal microbiota transplantation as potential therapeutic approaches that may lead to a new treatment paradigm for Parkinson's disease.

Journal article

This data is extracted from the Web of Science and reproduced under a licence from Thomson Reuters. You may not copy or re-distribute this data in whole or in part without the written consent of the Science business of Thomson Reuters.

Request URL: http://wlsprd.imperial.ac.uk:80/respub/WEB-INF/jsp/search-html.jsp Request URI: /respub/WEB-INF/jsp/search-html.jsp Query String: respub-action=search.html&id=00165819&limit=30&person=true