Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38147228

ABSTRACT

The blood-brain barrier consists of tightly connected endothelial cells protecting the brain's microenvironment from the periphery. These endothelial cells are characterized by specific tight junction proteins such as Claudin-5 and Occludin, forming the endothelial barrier. Disrupting these cells might lead to blood-brain barrier dysfunction. The Wnt/ß-catenin signaling pathway can regulate the expression of these tight junction proteins and subsequent barrier permeability. The aim of this study was to investigate the in vitro effects of Wnt7a mediated ß-catenin signaling on endothelial barrier integrity. Mouse brain endothelial cells, bEnd.3, were treated with recombinant Wnt7a protein or XAV939, a selective inhibitor of Wnt/ß-catenin mediated transcription to modulate the Wnt signaling pathway. The involvement of Wnt/HIF1α signaling was investigated by inhibiting Hif1α signaling with Hif1α siRNA. Wnt7a stimulation led to activation and nuclear translocation of ß-catenin, which was inhibited by XAV939. Wnt7a stimulation decreased Claudin-5 expression mediated by ß-catenin and decreased endothelial barrier formation. Wnt7a increased Hif1α and Vegfa expression mediated by ß-catenin. However, Hif1α signaling pathway did not regulate tight junction proteins Claudin-5 and Occludin. Our data suggest that Wnt7a stimulation leads to a decrease in tight junction proteins mediated by the nuclear translocation of ß-catenin, which hampers proper endothelial barrier formation. This process might be crucial in initiating endothelial cell proliferation and angiogenesis. Although HIF1α did not modulate the expression of tight junction proteins, it might play a role in brain angiogenesis and underlie pathogenic mechanisms in Wnt/HIF1α signaling in diseases such as cerebral small vessel disease.

2.
Acta Neuropathol Commun ; 11(1): 128, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37550790

ABSTRACT

Cerebral small vessel disease is characterised by decreased cerebral blood flow and blood-brain barrier impairments which play a key role in the development of white matter lesions. We hypothesised that cerebral hypoperfusion causes local hypoxia, affecting oligodendrocyte precursor cell-endothelial cell signalling leading to blood-brain barrier dysfunction as an early mechanism for the development of white matter lesions. Bilateral carotid artery stenosis was used as a mouse model for cerebral hypoperfusion. Pimonidazole, a hypoxic cell marker, was injected prior to humane sacrifice at day 7. Myelin content, vascular density, blood-brain barrier leakages, and hypoxic cell density were quantified. Primary mouse oligodendrocyte precursor cells were exposed to hypoxia and RNA sequencing was performed. Vegfa gene expression and protein secretion was examined in an oligodendrocyte precursor cell line exposed to hypoxia. Additionally, human blood plasma VEGFA levels were measured and correlated to blood-brain barrier permeability in normal-appearing white matter and white matter lesions of cerebral small vessel disease patients and controls. Cerebral blood flow was reduced in the stenosis mice, with an increase in hypoxic cell number and blood-brain barrier leakages in the cortical areas but no changes in myelin content or vascular density. Vegfa upregulation was identified in hypoxic oligodendrocyte precursor cells, which was mediated via Hif1α and Epas1. In humans, VEGFA plasma levels were increased in patients versus controls. VEGFA plasma levels were associated with increased blood-brain barrier permeability in normal appearing white matter of patients. Cerebral hypoperfusion mediates hypoxia induced VEGFA expression in oligodendrocyte precursor cells through Hif1α/Epas1 signalling. VEGFA could in turn increase BBB permeability. In humans, increased VEGFA plasma levels in cerebral small vessel disease patients were associated with increased blood-brain barrier permeability in the normal appearing white matter. Our results support a role of VEGFA expression in cerebral hypoperfusion as seen in cerebral small vessel disease.


Subject(s)
Cerebral Small Vessel Diseases , Oligodendrocyte Precursor Cells , White Matter , Humans , Mice , Animals , Blood-Brain Barrier/metabolism , Oligodendrocyte Precursor Cells/metabolism , White Matter/pathology , Hypoxia/metabolism , Cerebral Small Vessel Diseases/pathology , Vascular Endothelial Growth Factor A/metabolism
3.
Biol Imaging ; 3: e24, 2023.
Article in English | MEDLINE | ID: mdl-38510175

ABSTRACT

This study aimed to expand our understanding of myelin basic protein (MBP), a key component of central nervous system myelin, by developing a protocol to track and quantifying individual MBP particles during oligodendrocyte (OL) differentiation. MBP particle directionality, confinement, and diffusion were tracked by rapid TIRF and HILO imaging of Dendra2 tagged MBP in three stages of mouse oligodendroglia: OL precursors, early myelinating OLs, and mature myelinating OLs. The directionality and confinement of MBP particles increased at each stage consistent with progressive transport toward, and recruitment into, emerging myelin structures. Unexpectedly, diffusion data presented a more complex pattern with subpopulations of the most diffusive particles disappearing at the transition between the precursor and early myelinating stage, before reemerging in the membrane sheets of mature OLs. This diversity of particle behaviors, which would be undetectable by conventional ensemble-averaged methods, are consistent with a multifunctional view of MBP involving roles in myelin expansion and compaction.

4.
Cells ; 11(11)2022 05 31.
Article in English | MEDLINE | ID: mdl-35681504

ABSTRACT

The subventricular zone (SVZ) is the largest and most active germinal zone in the adult forebrain. Neural stem cells (NSCs) of the SVZ generate olfactory interneurons throughout life and retain the intrinsic ability to generate oligodendrocytes (OLs), the myelinating cells of the central nervous system. OLs and myelin are targets in demyelinating diseases such as multiple sclerosis (MS). Remyelination is dependent on the ability of oligodendrocyte progenitor cells (OPCs) to proliferate, migrate, and terminally differentiate into myelinating OLs. During aging, there is a gradual decrease in the regenerative capacity of OPCs, and the consequent loss of OLs and myelin is a contributing factor in cognitive decline and the failure of remyelination in MS and other pathologies with aging contexts, including Alzheimer's disease (AD) and stroke. The age-related decrease in oligodendrogenesis has not been fully characterised but is known to reflect changes in intrinsic and environmental factors affecting the ability of OPCs to respond to pro-differentiation stimuli. Notably, SVZ-derived OPCs are an important source of remyelinating OLs in addition to parenchymal OPCs. In this mini-review, we briefly discuss differences between SVZ-derived and parenchymal OPCs in their responses to demyelination and highlight challenges associated with their study in vivo and how they can be targeted for regenerative therapies in the aged brain.


Subject(s)
Multiple Sclerosis , Myelin Sheath , Aged , Brain/pathology , Humans , Lateral Ventricles , Multiple Sclerosis/pathology , Myelin Sheath/pathology , Oligodendroglia
5.
Front Cell Dev Biol ; 9: 688283, 2021.
Article in English | MEDLINE | ID: mdl-34504841

ABSTRACT

Embryonic stem cells (ESC) have the potential to generate homogeneous immature cells like stem/progenitor cells, which appear to be difficult to isolate and expand from primary tissue samples. In this study, we developed a simple method to generate homogeneous immature oligodendrocyte (OL) lineage cells from mouse ESC-derived neural stem cell (NSC). NSC converted to NG2+/OLIG2+double positive progenitors (NOP) after culturing in serum-free media for a week. NOP expressed Prox1, but not Gpr17 gene, highlighting their immature phenotype. Interestingly, FACS analysis revealed that NOP expressed proteins for NG2, but not PDGFRɑ, distinguishing them from primary OL progenitor cells (OPC). Nevertheless, NOP expressed various OL lineage marker genes including Cspg4, Pdgfrα, Olig1/2, and Sox9/10, but not Plp1 genes, and, when cultured in OL differentiation conditions, initiated transcription of Gpr17 and Plp1 genes, and expression of PDGFRα proteins, implying that NOP converted into a matured OPC phenotype. Unexpectedly, NOP remained multipotential, being able to differentiate into neurons as well as astrocytes under appropriate conditions. Moreover, NOP-derived OPC myelinated axons with a lower efficiency when compared with primary OPC. Taken together, these data demonstrate that NOP are an intermediate progenitor cell distinguishable from both NSC and primary OPC. Based on this profile, NOP may be useful for modeling mechanisms influencing the earliest stages of oligogenesis, and exploring the cellular and molecular responses of the earliest OL progenitors to conditions that impair myelination in the developing nervous system.

7.
Life (Basel) ; 11(5)2021 May 01.
Article in English | MEDLINE | ID: mdl-34062792

ABSTRACT

Migraine is a highly prevalent and disabling primary headache disorder, however its pathophysiology remains unclear, hindering successful treatment. A number of key secondary headache disorders have headaches that mimic migraine. Evidence has suggested a role of mitochondrial dysfunction and an imbalance between energetic supply and demand that may contribute towards migraine susceptibility. Targeting these deficits with nutraceutical supplementation may provide an additional adjunctive therapy. Neuroimaging techniques have demonstrated a metabolic phenotype in migraine similar to mitochondrial cytopathies, featuring reduced free energy availability and increased metabolic rate. This is reciprocated in vivo when modelling a fundamental mechanism of migraine aura, cortical spreading depression. Trials assessing nutraceuticals successful in the treatment of mitochondrial cytopathies including magnesium, coenzyme q10 and riboflavin have also been conducted in migraine. Although promising results have emerged from nutraceutical trials in patients with levels of minerals or vitamins below a critical threshold, they are confounded by lacking control groups or cohorts that are not large enough to be representative. Energetic imbalance in migraine may be relevant in driving the tissue towards maximum metabolic capacity, leaving the brain lacking in free energy. Personalised medicine considering an individual's deficiencies may provide an approach to ameliorate migraine.

8.
Sci Rep ; 11(1): 9588, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953273

ABSTRACT

Neuronal activity is established as a driver of oligodendrocyte (OL) differentiation and myelination. The concept of activity-dependent myelin plasticity, and its role in cognition and disease, is gaining support. Methods capable of resolving changes in the morphology of individual myelinating OL would advance our understanding of myelin plasticity and injury, thus we adapted a labelling approach involving Semliki Forest Virus (SFV) vectors to resolve and quantify the 3-D structure of OL processes and internodes in cerebellar slice cultures. We first demonstrate the utility of the approach by studying changes in OL morphology after complement-mediated injury. SFV vectors injected into cerebellar white matter labelled transitional OL (TOL), whose characteristic mixture of myelinating and non-myelinating processes exhibited significant degeneration after complement injury. The method was also capable of resolving finer changes in morphology related to neuronal activity. Prolonged suppression of neuronal activity, which reduced myelination, selectively decreased the length of putative internodes, and the proportion of process branches that supported them, while leaving other features of process morphology unaltered. Overall this work provides novel information on the morphology of TOL, and their response to conditions that alter circuit function or induce demyelination.


Subject(s)
Cerebellum/physiology , Myelin Sheath/physiology , Oligodendroglia/physiology , White Matter/physiology , Animals , Cell Shape/physiology , Cerebellum/pathology , Mice , Myelin Sheath/pathology , Neurogenesis/physiology , Neurons/pathology , Neurons/physiology , Oligodendroglia/pathology , White Matter/pathology
10.
Cells ; 9(6)2020 06 25.
Article in English | MEDLINE | ID: mdl-32630426

ABSTRACT

Key pathological features of cerebral small vessel disease (cSVD) include impairment of the blood brain barrier (BBB) and the progression of white matter lesions (WMLs) amongst other structural lesions, leading to the clinical manifestations of cSVD. The function of endothelial cells (ECs) is of major importance to maintain a proper BBB. ECs interact with several cell types to provide structural and functional support to the brain. Oligodendrocytes (OLs) myelinate axons in the central nervous system and are crucial in sustaining the integrity of white matter. The interplay between ECs and OLs and their precursor cells (OPCs) has received limited attention yet seems of relevance for the study of BBB dysfunction and white matter injury in cSVD. Emerging evidence shows a crosstalk between ECs and OPCs/OLs, mediated by signaling through the Wingless and Int-1 (WNT)/ß-catenin pathway. As the latter is involved in EC function (e.g., angiogenesis) and oligodendrogenesis, we reviewed the role of WNT/ß-catenin signaling for both cell types and performed a systematic search to identify studies describing a WNT-mediated interplay between ECs and OPCs/OLs. Dysregulation of this interaction may limit remyelination of WMLs and render the BBB leaky, thereby initiating a vicious neuroinflammatory cycle. A better understanding of the role of this signaling pathway in EC-OL crosstalk is essential in understanding cSVD development.


Subject(s)
Cerebral Small Vessel Diseases/genetics , Endothelial Cells/metabolism , Oligodendroglia/metabolism , Wnt Signaling Pathway/genetics , Humans
11.
Int J Mol Sci ; 20(10)2019 May 17.
Article in English | MEDLINE | ID: mdl-31108947

ABSTRACT

Glia form a central component of the nervous system whose varied activities sustain an environment that is optimised for healthy development and neuronal function. Alpha-amino-3-hydroxy-5-methyl-4-isoxazole (AMPA)-type glutamate receptors (AMPAR) are a central mediator of glutamatergic excitatory synaptic transmission, yet they are also expressed in a wide range of glial cells where they influence a variety of important cellular functions. AMPAR enable glial cells to sense the activity of neighbouring axons and synapses, and as such many aspects of glial cell development and function are influenced by the activity of neural circuits. However, these AMPAR also render glia sensitive to elevations of the extracellular concentration of glutamate, which are associated with a broad range of pathological conditions. Excessive activation of AMPAR under these conditions may induce excitotoxic injury in glial cells, and trigger pathophysiological responses threatening other neural cells and amplifying ongoing disease processes. The aim of this review is to gather information on AMPAR function from across the broad diversity of glial cells, identify their contribution to pathophysiological processes, and highlight new areas of research whose progress may increase our understanding of nervous system dysfunction and disease.


Subject(s)
Nervous System Diseases/metabolism , Neuroglia/metabolism , Receptors, AMPA/metabolism , Animals , Gene Expression Regulation , Glutamic Acid/metabolism , Humans , Synaptic Transmission
12.
Glia ; 66(9): 1896-1914, 2018 09.
Article in English | MEDLINE | ID: mdl-29704264

ABSTRACT

Glutamate receptor subunit 4 (GluA4) is highly expressed by neural cells sensitive to excitotoxicity, and is the predominant subunit expressed by oligodendrocyte precursor cells (OPC) during a key period of vulnerability to hypoxic-ischemic injury. Therefore, transcriptional networks downstream of excitotoxic GluA4 activation represent a promising area for therapeutic intervention. In this work, we identify the CCAAT binding transcription factor NF-Yb as a novel transcriptional regulator of Gria4 (GluA4 gene), and a controller of excitotoxic death in the oligodendroglial lineage. We describe a novel regulatory region within Gria4 containing CCAAT sequences whose binding by NF-Yb is regulated by excitotoxicity. Excitotoxicity-induced alterations in NF-Yb binding are associated with changes in Gria4 transcription, while knockdown of NF-Yb alters the transcription of reporter constructs containing this regulatory region. Data from immortalized and primary OPC reveal that RNAi and pharmacological disruption of NF-Yb alter Gria4 transcription, with the latter inducing apoptosis and influencing a set of apoptotic genes similarly regulated during excitotoxicity. These data provide the first definition of a trans-acting mechanism regulating Gria4, and identify the NF-Y network as a potential source of pharmacological targets for promoting OPC survival.


Subject(s)
CCAAT-Binding Factor/metabolism , Cell Survival/physiology , Oligodendroglia/metabolism , Receptors, AMPA/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , CCAAT-Binding Factor/genetics , Cell Line , Cell Survival/drug effects , Cells, Cultured , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/drug effects , Neocortex/metabolism , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , Receptors, AMPA/genetics , Regulatory Sequences, Nucleic Acid , Terpenes/pharmacology , Transcription, Genetic
13.
Neural Regen Res ; 12(8): 1247-1251, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28966634

ABSTRACT

Leucine rich repeat proteins have gained considerable interest as therapeutic targets due to their expression and biological activity within the central nervous system. LINGO-1 has received particular attention since it inhibits axonal regeneration after spinal cord injury in a RhoA dependent manner while inhibiting leucine rich repeat and immunoglobulin-like domain-containing protein 1 (LINGO-1) disinhibits neuron outgrowth. Furthermore, LINGO-1 suppresses oligodendrocyte precursor cell maturation and myelin production. Inhibiting the action of LINGO-1 encourages remyelination both in vitro and in vivo. Accordingly, LINGO-1 antagonists show promise as therapies for demyelinating diseases. An analogous protein to LINGO-1, amphoterin-induced gene and open reading frame-3 (AMIGO3), exerts the same inhibitory effect on the axonal outgrowth of central nervous system neurons, as well as interacting with the same receptors as LINGO-1. However, AMIGO3 is upregulated more rapidly after spinal cord injury than LINGO-1. We speculate that AMIGO3 has a similar inhibitory effect on oligodendrocyte precursor cell maturation and myelin production as with axogenesis. Therefore, inhibiting AMIGO3 will likely encourage central nervous system axonal regeneration as well as the production of myelin from local oligodendrocyte precursor cell, thus providing a promising therapeutic target and an area for future investigation.

15.
Neuropharmacology ; 110(Pt B): 594-604, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26407765

ABSTRACT

The formation and repair of myelin involves alterations in the molecular and physical properties of oligodendrocytes, and highly coordinated interactions with their target axons. Characterising the nature and timing of these events at the molecular and cellular levels illuminates the fundamental events underlying myelin formation, and provides opportunities for the development of therapies to replace myelin lost through traumatic injury and inflammation. The dynamic nature of these events requires that live-imaging methods be used to capture this information accurately and completely. Developments in imaging technologies, and model systems suitable for their application to myelination, have advanced the study of myelin formation, injury and repair. Similarly, new techniques for single molecule imaging, and novel imaging probes, are providing opportunities to resolve the dynamics of myelin proteins during myelination. Here, we explore these developments in the context of myelin formation and injury, identify unmet needs within the field where progress can be advanced through live-imaging approaches, identify technical challenges that are limiting this progress, and highlight practical applications for these approaches that could lead to therapies for the protection of oligodendrocytes and myelin from injury, and restore myelin lost through injury and disease. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.


Subject(s)
Central Nervous System , Inflammation , Myelin Sheath/pathology , Oligodendroglia/physiology , Single Molecule Imaging/methods , Animals , Central Nervous System/diagnostic imaging , Central Nervous System/pathology , Central Nervous System/physiopathology , Humans , Inflammation/diagnostic imaging , Inflammation/pathology , Inflammation/physiopathology
16.
Neurobiol Dis ; 85: 99-110, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26459109

ABSTRACT

Recent research has suggested that the growth of central nervous system (CNS) axons during development is mediated through the PI3K/Akt/mammalian target of rapamycin (mTOR) intracellular signalling axis and that suppression of activity in this pathway occurs during maturity as levels of the phosphatase and tensin homologue (PTEN) rise and inhibit PI3K activation of mTOR, accounting for the failure of axon regeneration in the injured adult CNS. This hypothesis is supported by findings confirming that suppression of PTEN in experimental adult animals promotes impressive axon regeneration in the injured visual and corticospinal motor systems. This review focuses on these recent developments, discussing the therapeutic potential of a mTOR-based treatment aimed at promoting functional recovery in CNS trauma patients, recognising that to fulfil this ambition, the new therapy should aim to promote not only axon regeneration but also remyelination of regenerated axons, neuronal survival and re-innervation of denervated targets through accurate axonal guidance and synaptogenesis, all with minimal adverse effects. The translational challenges presented by the implementation of this new axogenic therapy are also discussed.


Subject(s)
Nerve Regeneration/physiology , TOR Serine-Threonine Kinases/metabolism , Trauma, Nervous System/physiopathology , Trauma, Nervous System/therapy , Animals , Axons/physiology , Humans
17.
Glia ; 63(6): 1021-35, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25739948

ABSTRACT

Myelination is initiated when oligodendrocyte precursor cells (OPC) contact target axons. Neuronal activity promotes myelination through actions that may involve OPC AMPA and NMDA glutamate receptors (AMPAR, NMDAR). Therefore, activity and AMPAR/NMDAR activation are predicted to promote the morphological development of OPC. AMPAR can regulate OPC development, but this analysis was not performed in situ and the role of action potentials was not examined. Hence, the influence of activity and AMPAR on OPC morphology and development remain untested in the CNS where axon-glial interactions are preserved. Data on NMDAR are mixed with conflicting results from in vitro and in vivo work. To gain a fuller understanding of activity-dependent OPC development in situ, we explored the role of AMPAR and NMDAR in cerebellar slice cultures that permit the study of endogenous OPC development and myelination. The structure of individual OPC was resolved from cells labeled with membrane targeted GFP. Morphological data were then validated against assays of OPC development. Blocking either activity or AMPAR impaired the morphological development of OPC and promoted proliferation and differentiation. Increasing the pool of oligodendrocytes by blocking activity or AMPAR failed to promote myelination. Instead both myelination and the expression of myelin basic protein were reduced by these treatments suggesting that full differentiation to a myelinating phenotype did not occur. Blocking NMDAR left OPC proliferation, differentiation and morphology unchanged. These data indicate an important role for AMPAR but not NMDAR in mediating the activity-dependent signals that regulate OPC morphology, development and myelination.


Subject(s)
Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurons/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Receptors, AMPA/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/physiology , Female , Male , Mice, Inbred C57BL , Myelin Sheath/drug effects , Myelin Sheath/physiology , Neural Stem Cells/drug effects , Neurons/drug effects , Oligodendroglia/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Tissue Culture Techniques
18.
J Anat ; 224(2): 216-27, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24252088

ABSTRACT

Expression of the transmembrane NG2 chondroitin sulphate proteoglycan (CSPG) defines a distinct population of NG2-glia. NG2-glia serve as a regenerative pool of oligodendrocyte progenitor cells in the adult central nervous system (CNS), which is important for demyelinating diseases such as multiple sclerosis, and are a major component of the glial scar that inhibits axon regeneration after CNS injury. In addition, NG2-glia form unique neuron-glial synapses with unresolved functions. However, to date it has proven difficult to study the importance of NG2-glia in any of these functions using conventional transgenic NG2 'knockout' mice. To overcome this, we aimed to determine whether NG2-glia can be targeted using an immunotoxin approach. We demonstrate that incubation in primary anti-NG2 antibody in combination with secondary saporin-conjugated antibody selectively kills NG2-expressing cells in vitro. In addition, we provide evidence that the same protocol induces the loss of NG2-glia without affecting astrocyte or neuronal numbers in cerebellar brain slices from postnatal mice. This study shows that targeting the NG2 CSPG with immunotoxins is an effective and selective means for killing NG2-glia, which has important implications for studying the functions of these enigmatic cells both in the normal CNS, and in demyelination and degeneration.


Subject(s)
Ablation Techniques/methods , Cerebellum/metabolism , Cerebellum/surgery , Chondroitin Sulfate Proteoglycans/metabolism , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Cell Count , Cell Line , Cell Survival , Humans , Mice , Organ Culture Techniques
19.
PLoS One ; 8(9): e75223, 2013.
Article in English | MEDLINE | ID: mdl-24098685

ABSTRACT

Isoamylase-type debranching enzymes (ISAs) play an important role in determining starch structure. Amylopectin - a branched polymer of glucose - is the major component of starch granules and its architecture underlies the semi-crystalline nature of starch. Mutants of several species lacking the ISA1-subclass of isoamylase are impaired in amylopectin synthesis. Consequently, starch levels are decreased and an aberrant soluble glucan (phytoglycogen) with altered branch lengths and branching pattern accumulates. Here we use TAP (tandem affinity purification) tagging to provide direct evidence in Arabidopsis that ISA1 interacts with its homolog ISA2. No evidence for interaction with other starch biosynthetic enzymes was found. Analysis of the single mutants shows that each protein is destabilised in the absence of the other. Co-expression of both ISA1 and ISA2 Escherichia coli allowed the formation of the active recombinant enzyme and we show using site-directed mutagenesis that ISA1 is the catalytic subunit. The presence of the active isoamylase alters glycogen biosynthesis in E. coli, resulting in colonies that stain more starch-like with iodine. However, analysis of the glucans reveals that rather than producing an amylopectin like substance, cells expressing the active isoamylase still accumulate small amounts of glycogen together with a population of linear oligosaccharides that stain strongly with iodine. We conclude that for isoamylase to promote amylopectin synthesis it needs to act on a specific precursor (pre-amylopectin) generated by the combined actions of plant starch synthase and branching enzyme isoforms and when presented with an unsuitable substrate (i.e. E. coli glycogen) it simply degrades it.


Subject(s)
Arabidopsis/enzymology , Glycogen Debranching Enzyme System/metabolism , Isoamylase/metabolism , Multiprotein Complexes/metabolism , Base Sequence , Chromatography, Gel , Chromatography, Ion Exchange , DNA Primers/genetics , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Genetic Complementation Test , Glycogen/biosynthesis , Glycogen/metabolism , Isoamylase/genetics , Isoamylase/isolation & purification , Microscopy, Electron, Transmission , Molecular Sequence Data , Protein Stability , Protein Subunits/metabolism , Sequence Alignment , Sequence Analysis, DNA , Starch/biosynthesis , Substrate Specificity
20.
Dev Neurosci ; 33(2): 170-84, 2011.
Article in English | MEDLINE | ID: mdl-21912090

ABSTRACT

Prior studies suggest that non-canonical proteolipid protein (PLP) gene expression occurs during development in non-myelinating neurons as well as myelinating oligodendroglia in mammalian brain. To assess this possibility in neostriatum, a region of uncertain PLP gene expression in neurons, morphological and electrophysiological tools were used to determine phenotypes of cells with activation of a PLP promoter transgene during the early postnatal period in mice. PLP gene expression is evident in both neuronal and oligodendroglial phenotypes in developing neostriatum, a conclusion based on three novel observations: (1) An enhanced green fluorescent protein (EGFP) reporter of PLP promoter activation was localized in two distinct populations of cells, which exhibit collective, developmental differences of morphological and electrophysiological characteristics in accord with neuronal and oligodendroglial phenotypes of neostriatal cells found during the early postnatal period in both transgenic and wild-type mice. (2) The EGFP reporter of PLP promoter activation was appropriately positioned to serve as a regulator of PLP gene expression. It colocalized with native PLP proteins in both neuronal and oligodendroglial phenotypes; however, only soma-restricted PLP protein isoforms were found in the neuronal phenotype, while classic and soma-restricted PLP protein isoforms were found in the oligodendroglial phenotype. (3) As shown by EGFP reporter, PLP promoter activation was placed to regulate PLP gene expression in only one neuronal phenotype among the several that constitute neostriatum. It was localized in medium spiny neurons, but not large aspiny neurons. These outcomes have significant implications for the non-canonical functional roles of PLP gene expression in addition to myelinogenesis in mammalian brain, and are consistent with potentially independent pathologic loci in neurons during the course of human mutational disorders of PLP gene expression.


Subject(s)
Myelin Proteolipid Protein , Neostriatum/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Protein Isoforms/metabolism , Animals , Animals, Newborn , Brain/growth & development , Brain/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins , Humans , Mice , Mice, Transgenic , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/metabolism , Oligodendroglia/cytology , Protein Isoforms/genetics , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...