Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(1): 111930, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640356

ABSTRACT

Leukocyte recruitment from the vasculature into tissues is a crucial component of the immune system but is also key to inflammatory disease. Chemokines are central to this process but have yet to be therapeutically targeted during inflammation due to a lack of mechanistic understanding. Specifically, CXCL4 (Platelet Factor 4, PF4) has no established receptor that explains its function. Here, we use biophysical, in vitro, and in vivo techniques to determine the mechanism underlying CXCL4-mediated leukocyte recruitment. We demonstrate that CXCL4 binds to glycosaminoglycan (GAG) sugars on proteoglycans within the endothelial extracellular matrix, resulting in increased adhesion of leukocytes to the vasculature, increased vascular permeability, and non-specific recruitment of a range of leukocytes. Furthermore, GAG sulfation confers selectivity onto chemokine localization. These findings present mechanistic insights into chemokine biology and provide future therapeutic targets.


Subject(s)
Platelet Factor 4 , Proteoglycans , Platelet Factor 4/metabolism , Receptors, Chemokine , Chemokines/metabolism , Glycosaminoglycans , Extracellular Matrix/metabolism
2.
Glia ; 70(6): 1068-1083, 2022 06.
Article in English | MEDLINE | ID: mdl-35150591

ABSTRACT

Microglia, resident brain immune cells, are critical in orchestrating responses to central nervous system (CNS) injury. Many microglial functions, such as phagocytosis, motility and chemotaxis, are suggested to rely on chloride channels, including the volume-regulated anion channel (VRAC), but studies to date have relied on the use of pharmacological tools with limited specificity. VRAC has also been proposed as a drug target for acute CNS injury, and its role in microglial function is of considerable interest for developing CNS therapeutics. This study aimed to definitively confirm the contribution of VRAC in microglia function by using conditional LRRC8A-knockout mice, which lacked the essential VRAC subunit LRRC8A in microglia. We demonstrated that while VRAC contributed to cell volume regulation, it had no effect on phagocytic activity, cell migration or P2YR12-dependent chemotaxis. Moreover, loss of microglial VRAC did not affect microglial morphology or the extent of ischemic damage following stroke. We conclude that VRAC does not critically regulate microglial responses to brain injury and could be targetable in other CNS cell types (e.g., astrocytes) without impeding microglial function. Our results also demonstrate a role for VRAC in cell volume regulation but show that VRAC is not involved in several major cellular functions that it was previously thought to regulate, and point to other, alternative mechanisms of chloride transport in innate immunity.


Subject(s)
Microglia , Stroke , Animals , Cell Size , Ion Transport , Membrane Proteins/metabolism , Mice , Microglia/metabolism
3.
Int J Exp Pathol ; 103(2): 34-43, 2022 04.
Article in English | MEDLINE | ID: mdl-35076142

ABSTRACT

Leucocyte recruitment is a critical component of the immune response and is central to our ability to fight infection. Paradoxically, leucocyte recruitment is also a central component of inflammatory-based diseases such as rheumatoid arthritis, atherosclerosis and cancer. The role of the extracellular matrix, in particular proteoglycans, in this process has been largely overlooked. Proteoglycans consist of protein cores with glycosaminoglycan sugar side chains attached. Proteoglycans have been shown to bind and regulate the function of a number of proteins, for example chemokines, and also play a key structural role in the local tissue environment/niche. Whilst they have been implicated in leucocyte recruitment and inflammatory disease, their mechanistic function has yet to be fully understood, precluding therapeutic targeting. This review summarizes what is currently known about the role of proteoglycans in the different stages of leucocyte recruitment and proposes a number of areas where more research is needed. A better understanding of the mechanistic role of proteoglycans during inflammatory disease will inform the development of next-generation therapeutics.


Subject(s)
Extracellular Matrix , Proteoglycans , Extracellular Matrix/metabolism , Glycosaminoglycans , Proteoglycans/metabolism
5.
Nat Commun ; 12(1): 211, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431878

ABSTRACT

Graphene active sensors have demonstrated promising capabilities for the detection of electrophysiological signals in the brain. Their functional properties, together with their flexibility as well as their expected stability and biocompatibility have raised them as a promising building block for large-scale sensing neural interfaces. However, in order to provide reliable tools for neuroscience and biomedical engineering applications, the maturity of this technology must be thoroughly studied. Here, we evaluate the performance of 64-channel graphene sensor arrays in terms of homogeneity, sensitivity and stability using a wireless, quasi-commercial headstage and demonstrate the biocompatibility of epicortical graphene chronic implants. Furthermore, to illustrate the potential of the technology to detect cortical signals from infra-slow to high-gamma frequency bands, we perform proof-of-concept long-term wireless recording in a freely behaving rodent. Our work demonstrates the maturity of the graphene-based technology, which represents a promising candidate for chronic, wide frequency band neural sensing interfaces.


Subject(s)
Brain/physiology , Graphite/chemistry , Wireless Technology , Animals , Behavior, Animal , Gamma Rhythm/physiology , Materials Testing , Rats, Long-Evans , Signal Processing, Computer-Assisted , Sleep/physiology , Time Factors , Transistors, Electronic
6.
Dis Model Mech ; 13(5)2020 05 26.
Article in English | MEDLINE | ID: mdl-32152060

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA), also known as Kennedy's Disease, is a late-onset X-linked progressive neuromuscular disease, which predominantly affects males. The pathological hallmarks of the disease are selective loss of spinal and bulbar motor neurons, accompanied by weakness, atrophy and fasciculations of bulbar and limb muscles. SBMA is caused by a CAG repeat expansion in the gene that encodes the androgen receptor (AR) protein. Disease manifestation is androgen dependent and results principally from a toxic gain of AR function. There are currently no effective treatments for this debilitating disease. It is important to understand the course of the disease in order to target therapeutics to key pathological stages. This is especially relevant in disorders such as SBMA, for which disease can be identified before symptom onset, through family history and genetic testing. To fully characterise the role of muscle in SBMA, we undertook a longitudinal physiological and histological characterisation of disease progression in the AR100 mouse model of SBMA. Our results show that the disease first manifests in skeletal muscle, before any motor neuron degeneration, which only occurs in late-stage disease. These findings reveal that alterations in muscle function, including reduced muscle force and changes in contractile characteristics, are early pathological events in SBMA mice and suggest that muscle-targeted therapeutics may be effective in SBMA.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/pathology , Bulbo-Spinal Atrophy, X-Linked/physiopathology , Muscle Contraction , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Animals , Biomechanical Phenomena , Body Weight , Cell Survival , Disease Progression , Hindlimb/innervation , Hindlimb/physiopathology , Mice , Motor Activity/physiology , Motor Neurons/pathology , Muscle Fatigue , Muscle, Skeletal/innervation , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Oxidation-Reduction
7.
Hum Gene Ther ; 30(9): 1052-1066, 2019 09.
Article in English | MEDLINE | ID: mdl-31020862

ABSTRACT

Patients with the lysosomal storage disease mucopolysaccharidosis IIIA (MPSIIIA) lack the lysosomal enzyme N-sulfoglucosamine sulfohydrolase (SGSH), one of the many enzymes involved in degradation of heparan sulfate. Build-up of un-degraded heparan sulfate results in severe progressive neurodegeneration for which there is currently no treatment. Experimental gene therapies based on gene addition are currently being explored. Following preclinical evaluation in MPSIIIA mice, an adeno-associated virus vector of serotype rh10 designed to deliver SGSH and sulfatase modifying factor 1 (SAF301) was trialed in four MPSIIIA patients, showing good tolerance and absence of adverse events with some improvements in neurocognitive measures. This study aimed to improve SAF301 further by removing sulfatase modifying factor 1 (SUMF1) and assessing if expression of this gene is needed to increase the SGSH enzyme activity (SAF301b). Second, the murine phosphoglycerate kinase (PGK) promotor was exchanged with a chicken beta actin/CMV composite (CAG) promotor (SAF302) to see if SGSH expression levels could be boosted further. The three different vectors were administered to MPSIIIA mice via intracranial injection, and SGSH expression levels were compared 4 weeks post treatment. Removal of SUMF1 resulted in marginal reductions in enzyme activity. However, promotor exchange significantly increased the amount of SGSH expressed in the brain, leading to superior therapeutic correction with SAF302. Biodistribution of SAF302 was further assessed using green fluorescent protein (GFP), indicating that vector spread was limited to the area around the injection tract. Further modification of the injection strategy to a single depth with higher injection volume increased vector distribution, leading to more widespread GFP distribution and sustained expression, suggesting this approach should be adopted in future trials.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/physiopathology , Animals , Biomarkers , Corpus Striatum/metabolism , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Fluorescent Antibody Technique , Gene Expression , Gene Order , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/isolation & purification , Hydrolases/genetics , Mice , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/therapy , Neurons/metabolism , Organ Specificity/genetics , Transduction, Genetic , Transgenes , Treatment Outcome
8.
Brain ; 137(Pt 7): 1894-906, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24898351

ABSTRACT

Spinal and bulbar muscular atrophy is an X-linked degenerative motor neuron disease caused by an abnormal expansion in the polyglutamine encoding CAG repeat of the androgen receptor gene. There is evidence implicating endoplasmic reticulum stress in the development and progression of neurodegenerative disease, including polyglutamine disorders such as Huntington's disease and in motor neuron disease, where cellular stress disrupts functioning of the endoplasmic reticulum, leading to induction of the unfolded protein response. We examined whether endoplasmic reticulum stress is also involved in the pathogenesis of spinal and bulbar muscular atrophy. Spinal and bulbar muscular atrophy mice that carry 100 pathogenic polyglutamine repeats in the androgen receptor, and develop a late-onset neuromuscular phenotype with motor neuron degeneration, were studied. We observed a disturbance in endoplasmic reticulum-associated calcium homeostasis in cultured embryonic motor neurons from spinal and bulbar muscular atrophy mice, which was accompanied by increased endoplasmic reticulum stress. Furthermore, pharmacological inhibition of endoplasmic reticulum stress reduced the endoplasmic reticulum-associated cell death pathway. Examination of spinal cord motor neurons of pathogenic mice at different disease stages revealed elevated expression of markers for endoplasmic reticulum stress, confirming an increase in this stress response in vivo. Importantly, the most significant increase was detected presymptomatically, suggesting that endoplasmic reticulum stress may play an early and possibly causal role in disease pathogenesis. Our results therefore indicate that the endoplasmic reticulum stress pathway could potentially be a therapeutic target for spinal and bulbar muscular atrophy and related polyglutamine diseases.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Muscular Disorders, Atrophic/pathology , Muscular Disorders, Atrophic/physiopathology , Age Factors , Androgens/pharmacology , Androgens/therapeutic use , Animals , Anterior Horn Cells/physiopathology , Apoptosis/drug effects , Apoptosis/genetics , Cells, Cultured , Dihydrotestosterone/pharmacology , Dihydrotestosterone/therapeutic use , Disease Models, Animal , Embryo, Mammalian , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscular Disorders, Atrophic/drug therapy , Muscular Disorders, Atrophic/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Spinal Cord/pathology , Thapsigargin/therapeutic use
9.
PLoS One ; 8(8): e73944, 2013.
Article in English | MEDLINE | ID: mdl-24023695

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by the selective loss of motor neurons in the spinal cord, brain stem, and motor cortex. Mutations in superoxide dismutase (SOD1) are associated with familial ALS and lead to SOD1 protein misfolding and aggregation. Here we show that the molecular chaperone, HSJ1 (DNAJB2), mutations in which cause distal hereditary motor neuropathy, can reduce mutant SOD1 aggregation and improve motor neuron survival in mutant SOD1 models of ALS. Overexpression of human HSJ1a (hHSJ1a) in vivo in motor neurons of SOD1(G93A) transgenic mice ameliorated disease. In particular, there was a significant improvement in muscle force, increased motor unit number and enhanced motor neuron survival. hHSJ1a was present in a complex with SOD1(G93A) and led to reduced SOD1 aggregation at late stages of disease progression. We also observed altered ubiquitin immunoreactivity in the double transgenic animals, suggesting that ubiquitin modification might be important for the observed improvements. In a cell model of SOD1(G93A) aggregation, HSJ1a preferentially bound to mutant SOD1, enhanced SOD1 ubiquitylation and reduced SOD1 aggregation in a J-domain and ubiquitin interaction motif (UIM) dependent manner. Collectively, the data suggest that HSJ1a acts on mutant SOD1 through a combination of chaperone, co-chaperone and pro-ubiquitylation activity. These results show that targeting SOD1 protein misfolding and aggregation in vivo can be neuroprotective and suggest that manipulation of DnaJ molecular chaperones might be useful in the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , HSP40 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Neuroprotective Agents/metabolism , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Body Weight , Cattle , Cell Survival , Disease Models, Animal , Disease Progression , Female , Humans , Longevity , Male , Mice , Mice, Transgenic , Models, Biological , Motor Neurons/pathology , Muscles/physiopathology , Organ Size , Protein Structure, Quaternary , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Ubiquitination
10.
Brain ; 136(Pt 3): 926-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23393146

ABSTRACT

Spinal and bulbar muscular atrophy, also known as Kennedy's disease, is an adult-onset hereditary neurodegenerative disorder caused by an expansion of the polyglutamine repeat in the first exon in the androgen receptor gene. Pathologically, the disease is defined by selective loss of spinal and bulbar motor neurons causing bulbar, facial and limb weakness. Although the precise disease pathophysiology is largely unknown, it appears to be related to abnormal accumulation of the pathogenic androgen receptor protein within the nucleus, leading to disruption of cellular processes. Using a mouse model of spinal and bulbar muscular atrophy that exhibits many of the characteristic features of the human disease, in vivo physiological assessment of muscle function revealed that mice with the pathogenic expansion of the androgen receptor develop a motor deficit characterized by a reduction in muscle force, abnormal muscle contractile characteristics, loss of functional motor units and motor neuron degeneration. We have previously shown that treatment with arimoclomol, a co-inducer of the heat shock stress response, delays disease progression in the mutant superoxide dismutase 1 mouse model of amyotrophic lateral sclerosis, a fatal motor neuron disease. We therefore evaluated the therapeutic potential of arimoclomol in mice with spinal and bulbar muscular atrophy. Arimoclomol was administered orally, in drinking water, from symptom onset and the effects established at 18 months of age, a late stage of disease. Arimoclomol significantly improved hindlimb muscle force and contractile characteristics, rescued motor units and, importantly, improved motor neuron survival and upregulated the expression of the vascular endothelial growth factor which possess neurotrophic activity. These results provide evidence that upregulation of the heat shock response by treatment with arimoclomol may have therapeutic potential in the treatment of spinal and bulbar muscular atrophy and may also be a possible approach for the treatment of other neurodegenerative diseases.


Subject(s)
Heat-Shock Proteins/metabolism , Heat-Shock Response/drug effects , Hydroxylamines/pharmacology , Muscular Disorders, Atrophic/metabolism , Neuroprotective Agents/pharmacology , Animals , Blotting, Western , Disease Models, Animal , Disease Progression , Heat-Shock Response/physiology , Male , Mice , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...