147 results on '"Waldvogel HJ"'
Search Results
2. The histamine H4 receptor is functionally expressed on neurons in the mammalian CNS
- Author
-
Connelly, WM, primary, Shenton, FC, additional, Lethbridge, N, additional, Leurs, R, additional, Waldvogel, HJ, additional, Faull, RLM, additional, Lees, G, additional, and Chazot, PL, additional
- Published
- 2009
- Full Text
- View/download PDF
3. Cell loss in the motor and cingulate cortex correlates with symptomatology in Huntington's disease.
- Author
-
Thu DC, Oorschot DE, Tippett LJ, Nana AL, Hogg VM, Synek BJ, Luthi-Carter R, Waldvogel HJ, and Faull RL
- Published
- 2010
- Full Text
- View/download PDF
4. Localization of LRRK2 to membranous and vesicular structures in mammalian brain.
- Author
-
Biskup S, Moore DJ, Celsi F, Higashi S, West AB, Andrabi SA, Kurkinen K, Yu SW, Savitt JM, Waldvogel HJ, Faull RL, Emson PC, Torp R, Ottersen OP, Dawson TM, Dawson VL, Biskup, Saskia, Moore, Darren J, Celsi, Fulvio, and Higashi, Shinji
- Published
- 2006
5. Chemical neuroanatomy of the substantia nigra in the ovine brain
- Author
-
Murray, Samantha, Black, BL, Reid, SJ, Rudiger, SR, Bawden, CS, Snell, RG, Waldvogel, HJ, and Faull, RLM
- Published
- 2019
- Full Text
- View/download PDF
6. Single nuclei RNA-seq reveals a medium spiny neuron glutamate excitotoxicity signature prior to the onset of neuronal death in an ovine Huntington's disease model.
- Author
-
Jiang A, You L, Handley RR, Hawkins V, Reid SJ, Jacobsen JC, Patassini S, Rudiger SR, Mclaughlan CJ, Kelly JM, Verma PJ, Bawden CS, Gusella JF, MacDonald ME, Waldvogel HJ, Faull RLM, Lehnert K, and Snell RG
- Subjects
- Animals, Sheep, RNA-Seq, Receptors, AMPA genetics, Receptors, AMPA metabolism, Cell Death genetics, Corpus Striatum metabolism, Corpus Striatum pathology, Animals, Genetically Modified, Huntingtin Protein genetics, Huntingtin Protein metabolism, Humans, Transcriptome genetics, Receptors, Kainic Acid genetics, Receptors, Kainic Acid metabolism, Cell Nucleus metabolism, Cell Nucleus genetics, Medium Spiny Neurons, Huntington Disease genetics, Huntington Disease metabolism, Huntington Disease pathology, Disease Models, Animal, Neurons metabolism, Neurons pathology, Glutamic Acid metabolism, Receptors, N-Methyl-D-Aspartate genetics, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
Huntington's disease (HD) is a neurodegenerative genetic disorder caused by an expansion in the CAG repeat tract of the huntingtin (HTT) gene resulting in behavioural, cognitive, and motor defects. Current knowledge of disease pathogenesis remains incomplete, and no disease course-modifying interventions are in clinical use. We have previously reported the development and characterisation of the OVT73 transgenic sheep model of HD. The 73 polyglutamine repeat is somatically stable and therefore likely captures a prodromal phase of the disease with an absence of motor symptomatology even at 5-years of age and no detectable striatal cell loss. To better understand the disease-initiating events we have undertaken a single nuclei transcriptome study of the striatum of an extensively studied cohort of 5-year-old OVT73 HD sheep and age matched wild-type controls. We have identified transcriptional upregulation of genes encoding N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and kainate receptors in medium spiny neurons, the cell type preferentially lost early in HD. Further, we observed an upregulation of astrocytic glutamate uptake transporters and medium spiny neuron GABAA receptors, which may maintain glutamate homeostasis. Taken together, these observations support the glutamate excitotoxicity hypothesis as an early neurodegeneration cascade-initiating process but the threshold of toxicity may be regulated by several protective mechanisms. Addressing this biochemical defect early may prevent neuronal loss and avoid the more complex secondary consequences precipitated by cell death., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
7. Alzheimer's Disease-associated Region-specific Decrease of Vesicular Glutamate Transporter Immunoreactivity inthe Medial Temporal Lobe and Superior Temporal Gyrus.
- Author
-
Wood OWG, Walby J, Yeung JH, Ke S, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Humans, Male, Aged, Female, Aged, 80 and over, Middle Aged, Immunohistochemistry, Alzheimer Disease metabolism, Alzheimer Disease pathology, Temporal Lobe metabolism, Temporal Lobe pathology, Vesicular Glutamate Transport Protein 1 metabolism, Vesicular Glutamate Transport Protein 2 metabolism
- Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which there are very limited treatment options. Dysfunction of the excitatory neurotransmitter system is thought to play a major role in the pathogenesis of this condition. Vesicular glutamate transporters (VGLUTs) are key to controlling the quantal release of glutamate. Thus, expressional changes in disease can have implications for aberrant neuronal activity, raising the possibility of a therapeutic target. There is no information regarding the expression of VGLUTs in the human medial temporal lobe in AD, one of the earliest and most severely affected brain regions. This study aimed to quantify and compare the layer-specific expression of VGLUT1 and VGLUT2 between control and AD cases in the hippocampus, subiculum, entorhinal cortex, and superior temporal gyrus. Free-floating fluorescent immunohistochemistry was used to label VGLUT1 and VGLUT2 in the hippocampus, subiculum, entorhinal cortex, and superior temporal gyrus. Sections were imaged using laser-scanning confocal microscopy and transporter densitometric analysis was performed. VGLUT1 density was not significantly different in AD tissue, except lower staining density observed in the dentate gyrus stratum moleculare (p = 0.0051). VGLUT2 expression was not altered in the hippocampus and entorhinal cortex of AD cases but was significantly lower in the subiculum (p = 0.015) and superior temporal gyrus (p = 0.0023). This study indicates a regionally specific vulnerability of VGLUT1 and VGLUT2 expression in the medial temporal lobe and superior temporal gyrus in AD. However, the causes and functional consequences of these disturbances need to be further explored to assess VGLUT1 and VGLUT2 as viable therapeutic targets., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Microglial and Astrocytic Responses in the Human Midcingulate Cortex in Huntington's Disease.
- Author
-
Palpagama T, Mills AR, Ferguson MW, Vikas Ankeal P, Turner C, Tippett L, van der Werf B, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Humans, Microglia metabolism, Astrocytes metabolism, Huntingtin Protein genetics, Neuroinflammatory Diseases, Gyrus Cinguli, Huntington Disease pathology
- Abstract
Objective: Patients with Huntington's disease can present with variable difficulties of motor functioning, mood, and cognition. Neurodegeneration occurs in the anterior cingulate cortex of some patients with Huntington's disease and is linked to the presentation of mood symptomatology. Neuroinflammation, perpetrated by activated microglia and astrocytes, has been reported in Huntington's disease and may contribute to disease progression and presentation. This study sought to quantify the density of mutant huntingtin protein and neuroinflammatory glial changes in the midcingulate cortex of postmortem patients with Huntington's disease and determine if either correlates with the presentation of mood, motor, or mixed symptomatology., Methods: Free-floating immunohistochemistry quantified 1C2 immunolabeling density as an indicative marker of mutant huntingtin protein, and protein and morphological markers of astrocyte (EAAT2, Cx43, and GFAP), and microglial (Iba1 and HLA-DP/DQ/DR) activation. Relationships among the level of microglial activation, mutant huntingtin burden, and case characteristics were explored using correlative analysis., Results: We report alterations in activated microglia number and morphology in the midcingulate cortex of Huntington's disease cases with predominant mood symptomatology. An increased proportion of activated microglia was observed in the midcingulate of all Huntington's disease cases and positively correlated with 1C2 burden. Alterations in the astrocytic glutamate transporter EAAT2 were observed in the midcingulate cortex of patients associated with mood symptoms., Interpretation: This study presents pathological changes in microglia and astrocytes in the midcingulate cortex in Huntington's disease, which coincide with mood symptom presentation. These findings further the understanding of neuroinflammation in Huntington's disease, a necessary step for developing inflammation-targeted therapeutics. ANN NEUROL 2023;94:895-910., (© 2023 The Authors. Annals of Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.)
- Published
- 2023
- Full Text
- View/download PDF
9. DARPP-32 cells and neuropil define striosomal system and isolated matrix cells in human striatum.
- Author
-
Arasaratnam CJ, Song JJ, Yoshida T, Curtis MA, Graybiel AM, Faull RLM, and Waldvogel HJ
- Subjects
- Humans, Basal Ganglia, Neurons metabolism, Receptors, Dopamine D2 metabolism, Dopamine and cAMP-Regulated Phosphoprotein 32 metabolism, Neuropil metabolism, Corpus Striatum metabolism, Caudate Nucleus metabolism
- Abstract
The dorsal striatum forms a central node of the basal ganglia interconnecting the neocortex and thalamus with circuits modulating mood and movement. Striatal projection neurons (SPNs) include relatively intermixed populations expressing D1-type or D2-type dopamine receptors (dSPNs and iSPNs) that give rise to the direct (D1) and indirect (D2) output systems of the basal ganglia. Overlaid on this organization is a compartmental organization, in which a labyrinthine system of striosomes made up of sequestered SPNs is embedded within the larger striatal matrix. Striosomal SPNs also include D1-SPNs and D2-SPNs, but they can be distinguished from matrix SPNs by many neurochemical markers. In the rodent striatum the key signaling molecule, DARPP-32, is a exception to these compartmental expression patterns, thought to befit its functions through opposite actions in both D1- and D2-expressing SPNs. We demonstrate here, however, that in the dorsal human striatum, DARPP-32 is concentrated in the neuropil and SPNs of striosomes, especially in the caudate nucleus and dorsomedial putamen, relative to the matrix neuropil in these regions. The generally DARPP-32-poor matrix contains scattered DARPP-32-positive cells. DARPP-32 cell bodies in both compartments proved negative for conventional intraneuronal markers. These findings raise the potential for specialized DARPP-32 expression in the human striosomal system and in a set of DARPP-32-positive neurons in the matrix. If DARPP-32 immunohistochemical positivity predicts differential functional DARPP-32 activity, then the distributions demonstrated here could render striosomes and dispersed matrix cells susceptible to differential signaling through cAMP and other signaling systems in health and disease. DARPP-32 is highly concentrated in cells and neuropil of striosomes in post-mortem human brain tissue, particularly in the dorsal caudate nucleus. Scattered DARPP-32-positive cells are found in the human striatal matrix. Calbindin and DARPP-32 do not colocalize within every spiny projection neuron in the dorsal human caudate nucleus., (© 2023 The Authors. The Journal of Comparative Neurology published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
10. The regional and cellular distribution of GABA A receptor subunits in the human amygdala.
- Author
-
Song JJ, Curtis MA, Faull RLM, and Waldvogel HJ
- Subjects
- Humans, Parvalbumins metabolism, Interneurons metabolism, Brain metabolism, Receptors, GABA-A metabolism, Amygdala metabolism
- Abstract
GABAergic neurotransmission in the amygdala plays a crucial role in mediating emotional learning, fear, and memory. In this study, expression of five major GABA
A receptor subunits (α1, α2, α3, β2,3, and γ2) was investigated in the normal human amygdala using immunohistochemistry. At the regional level, the amygdala contains a highly heterogeneous distribution of all the subunits investigated. The most intense staining for α1, α2, β2,3, and γ2 subunits was present in the lateral nucleus (LA), and α3 in the intercalated nuclei (ICM). Six distinct cell populations that express GABAA receptor subunits were identified throughout the amygdala: type 1 aspiny cells in the basolateral nuclear group (BLNG) and superficial cortical-like nuclear region (SCLR) express α1, β2,3, and γ2; type 2 larger aspiny cells in the paralaminar nucleus (PL) express α1, β2,3, and γ2; type 3 aspiny cells in the BLNG express α1, β2,3, and γ2 as well as calcium-binding proteins including parvalbumin (PV), calbindin (CB), and calretinin (CR); type 4 pyramidal cells in the BLNG and SCLR express α2, α3, β2,3, and γ2 subunits at high levels on proximal specialised spines; type 5 cells in the central nucleus (CE) express α2, α3, and β2,3; type 6 cells are found closely packed in the intercalated cell masses (ICM) and express α3 and β2,3. The α1 subunit rarely co-labelled with α2 and α3 in the same cell population, while the α2 and α3 were often expressed within the same type 4 or 5 cell though not at always at the same puncta. The predominant GABAA receptor subunit combinations expressed in the human amygdala are the α1β2,3γ2 and α2β2,3γ2. Cells classified as interneuron types (types 1-3) contained GAD and principally expressed α1β2,3γ2. The major projection neurons of the BLNG (type 4) are non-GABAergic and mainly express α2β2,3γ2. The α3 subunit was found intracellularly in type 5 cells and decorating the surface of type 6 cells but rarely co-labelled with the subunits investigated. The results reveal a complex and heterogeneous distribution of GABAA receptor subtypes throughout the amygdala as well as on a variety of cell types through which inhibitory processing is carried out to maintain emotional responses, and control anxiety and fear responses in the brain., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
11. iGluR expression in the hippocampal formation, entorhinal cortex, and superior temporal gyrus in Alzheimer's disease.
- Author
-
Y Yeung JH, Waldvogel HJ, M Faull RL, and Kwakowsky A
- Published
- 2022
- Full Text
- View/download PDF
12. mGluR1α expression in the hippocampus, subiculum, entorhinal cortex and superior temporal gyrus in Alzheimer's disease.
- Author
-
Yeung JHY, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Abstract
Glutamate is the main excitatory neurotransmitter in the central nervous system, responsible for a plethora of cellular processes including memory formation and higher cerebral function and has been implicated in various neurological disease states. Alzheimer's disease (AD) is the leading neurodegenerative disorder worldwide and is characterized by significant cell loss and glutamatergic dysfunction. While there has been a focus on ionotropic glutamatergic receptors few studies have attempted to elucidate the pathological changes of metabotropic glutamate receptors (mGluRs) in AD. mGluRs are G-protein coupled receptors which have a wide-ranging functionality, including the regulation of neuronal injury and survival. In particular, the group I mGluRs (mGluR1 and mGluR5) are associated with ionotropic receptor activation and upregulation with resultant glutamate release in normal neuronal functioning. The mGluR subtype 1 splice variant a (mGluR1α) is the longest variant of the mGluR1 receptor, is localized to dendritic processes and is mainly plasma membrane-bound. Activation of mGluR1a has been shown to result in increased constitutive activity of ionotropic receptors, although its role in neurodegenerative and other neurological diseases is controversial, with some animal studies demonstrating potential neuroprotective properties in excito- and neurotoxic environments. In this study, the expression of mGluR1a within normal and AD human hippocampal tissue was quantified using immunohistochemistry. We found a significantly reduced expression of mGluR1α within the stratum pyramidale and radiatum of the CA1subregion, subiculum, and entorhinal cortex. This downregulation could result in potential dysregulation of the glutamatergic system with consequences on AD progression by promoting excitotoxicity, but alternatively may also be a neuroprotective mechanism to prevent mGluR1α associated excitotoxic effects. In summary, more research is required to understand the role and possible consequences of mGluR1α downregulation in the human AD hippocampus, subiculum and entorhinal cortex and its potential as a therapeutic target., Competing Interests: The authors declare no competing financial interests., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
13. Neuroprotective Effect of Caffeine in Alzheimer's Disease.
- Author
-
M Yelanchezian YM, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Animals, Caffeine pharmacology, Caffeine therapeutic use, Coffee metabolism, Humans, Alzheimer Disease etiology, Cognition Disorders etiology, Neuroprotective Agents pharmacology, Neuroprotective Agents therapeutic use
- Abstract
Alzheimer's disease (AD) is the leading cause of dementia, predicted to be the most significant health burden of the 21st century, with an estimated 131.5 million dementia patients by the year 2050. This review aims to provide an overview of the effect of caffeine on AD and cognition by summarizing relevant research conducted on this topic. We searched the Web of Science core collection and PubMed for studies related to the effect of caffeine on AD and cognition using title search terms: caffeine; coffee; Alzheimer's; cognition. There is suggestive evidence from clinical studies that caffeine is neuroprotective against dementia and possibly AD (20 out of 30 studies support this), but further studies, such as the "ideal" study proposed in this review, are required to prove this link. Clinical studies also indicate that caffeine is a cognitive normalizer and not a cognitive enhancer. Furthermore, clinical studies suggest the neuroprotective effect of caffeine might be confounded by gender. There is robust evidence based on in vivo and in vitro studies that caffeine has neuroprotective properties in AD animal models (21 out of 22 studies support this), but further studies are needed to identify the mechanistic pathways mediating these effects.
- Published
- 2022
- Full Text
- View/download PDF
14. Current and Possible Future Therapeutic Options for Huntington's Disease.
- Author
-
Ferguson MW, Kennedy CJ, Palpagama TH, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Abstract
Huntington's disease (HD) is an autosomal neurodegenerative disease that is characterized by an excessive number of CAG trinucleotide repeats within the huntingtin gene ( HTT). HD patients can present with a variety of symptoms including chorea, behavioural and psychiatric abnormalities and cognitive decline. Each patient has a unique combination of symptoms, and although these can be managed using a range of medications and non-drug treatments there is currently no cure for the disease. Current therapies prescribed for HD can be categorized by the symptom they treat. These categories include chorea medication, antipsychotic medication, antidepressants, mood stabilizing medication as well as non-drug therapies. Fortunately, there are also many new HD therapeutics currently undergoing clinical trials that target the disease at its origin; lowering the levels of mutant huntingtin protein (mHTT). Currently, much attention is being directed to antisense oligonucleotide (ASO) therapies, which bind to pre-RNA or mRNA and can alter protein expression via RNA degradation, blocking translation or splice modulation. Other potential therapies in clinical development include RNA interference (RNAi) therapies, RNA targeting small molecule therapies, stem cell therapies, antibody therapies, non-RNA targeting small molecule therapies and neuroinflammation targeted therapies. Potential therapies in pre-clinical development include Zinc-Finger Protein (ZFP) therapies, transcription activator-like effector nuclease (TALEN) therapies and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated system (Cas) therapies. This comprehensive review aims to discuss the efficacy of current HD treatments and explore the clinical trial progress of emerging potential HD therapeutics., Competing Interests: Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article., (© The Author(s) 2022.)
- Published
- 2022
- Full Text
- View/download PDF
15. Beta-Amyloid (Aβ 1-42 ) Increases the Expression of NKCC1 in the Mouse Hippocampus.
- Author
-
Lam P, Vinnakota C, Guzmán BC, Newland J, Peppercorn K, Tate WP, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Amyloid beta-Peptides, Animals, Chlorides metabolism, Male, Mice, Mice, Inbred C57BL, Peptide Fragments, Bumetanide metabolism, Bumetanide pharmacology, Hippocampus metabolism, Solute Carrier Family 12, Member 2 genetics, Solute Carrier Family 12, Member 2 metabolism, Symporters metabolism
- Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with an increasing need for developing disease-modifying treatments as current therapies only provide marginal symptomatic relief. Recent evidence suggests the γ-aminobutyric acid (GABA) neurotransmitter system undergoes remodeling in AD, disrupting the excitatory/inhibitory (E/I) balance in the brain. Altered expression levels of K-Cl-2 (KCC2) and N-K-Cl-1 (NKCC1), which are cation-chloride cotransporters (CCCs), have been implicated in disrupting GABAergic activity by regulating GABA
A receptor signaling polarity in several neurological disorders, but these have not yet been explored in AD. NKCC1 and KCC2 regulate intracellular chloride [Cl- ]i by accumulating and extruding Cl- , respectively. Increased NKCC1 expression in mature neurons has been reported in these disease conditions, and bumetanide, an NKCC1 inhibitor, is suggested to show potential therapeutic benefits. This study used primary mouse hippocampal neurons to explore if KCC2 and NKCC1 expression levels are altered following beta-amyloid (Aβ1-42 ) treatment and the potential neuroprotective effects of bumetanide. KCC2 and NKCC1 expression levels were also examined in 18-months-old male C57BL/6 mice following bilateral hippocampal Aβ1-42 stereotaxic injection. No change in KCC2 and NKCC1 expression levels were observed in mouse hippocampal neurons treated with 1 nM Aβ1-42 , but NKCC1 expression increased 30-days post-Aβ1-42 -injection in the CA1 region of the mouse hippocampus. Primary mouse hippocampal cultures were treated with 1 nM Aβ1-42 alone or with various concentrations of bumetanide (1 µM, 10 µM, 100 µM, 1 mM) to investigate the effect of the drug on cell viability. Aβ1-42 produced 53.1 ± 1.4% cell death after 5 days, and the addition of bumetanide did not reduce this. However, the drug at all concentrations significantly reduced cell viability, suggesting bumetanide is highly neurotoxic. In summary, these results suggest that chronic exposure to Aβ1-42 alters the balance of KCC2 and NKCC1 expression in a region-and layer-specific manner in mouse hippocampal tissue; therefore, this process most likely contributes to altered hippocampal E/I balance in this model. Furthermore, bumetanide induces hippocampal neurotoxicity, thus questioning its suitability for AD therapy. Further investigations are required to examine the effects of Aβ1-42 on KCC2 and NKCC1 expression and whether targeting CCCs might offer a therapeutic approach for AD.- Published
- 2022
- Full Text
- View/download PDF
16. Identifying Neural Progenitor Cells in the Adult Human Brain.
- Author
-
Park TIH, Waldvogel HJ, Montgomery JM, Mee EW, Bergin PS, Faull RLM, Dragunow M, and Curtis MA
- Subjects
- Adult, Adult Stem Cells, Brain, Cell Culture Techniques, Cell Differentiation, Humans, Immunohistochemistry, Neural Stem Cells
- Abstract
The discovery, in 1998, that the adult human brain contains at least two populations of progenitor cells and that progenitor cells are upregulated in response to a range of degenerative brain diseases has raised hopes for their use in replacing dying brain cells. Since these early findings, the race has been on to understand the biology of progenitor cells in the human brain, and they have now been isolated and studied in many major neurodegenerative diseases. Before these cells can be exploited for cell replacement purposes, it is important to understand how to (1) locate them, (2) label them, (3) determine what receptors they express, (4) isolate them, and (5) examine their electrophysiological properties when differentiated. In this chapter we have described the methods we use for studying progenitor cells in the adult human brain and in particular the tissue processing, immunohistochemistry, autoradiography, progenitor cell culture, and electrophysiology on brain cells. The Neurological Foundation of New Zealand Human Brain Bank has been receiving human tissue for approximately 25 years during which time we have developed a number of unique ways to examine and isolate progenitor cells from resected surgical specimens as well as from postmortem brain tissue. There are ethical and technical considerations that are unique to working with human brain tissue, and these, as well as the processing of this tissue and the culturing of it for the purpose of studying progenitor cells, are the topic of this chapter., (© 2022. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
17. The effect of age and sex on the expression of GABA signaling components in the human hippocampus and entorhinal cortex.
- Author
-
Ethiraj J, Palpagama TH, Turner C, van der Werf B, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Adult, Age Factors, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Sex Factors, Signal Transduction, Brain metabolism, Entorhinal Cortex metabolism, Hippocampus metabolism, Receptors, GABA-A metabolism, gamma-Aminobutyric Acid metabolism
- Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the nervous system. The GABA signaling system in the brain is comprised of GABA synthesizing enzymes, transporters, GABAA and GABAB receptors (GABA
A R and GABAB R). Alterations in the expression of these signaling components have been observed in several brain regions throughout aging and between sexes in various animal models. The hippocampus is the memory centre of the brain and is impaired in several age-related disorders. It is composed of two main regions: the Cornu Ammonis (CA1-4) and the Dentate Gyrus (DG), which are interconnected with the Entorhinal Cortex (ECx). The age- and sex-specific changes of GABA signaling components in these regions of the human brain have not been examined. This study is the first to determine the effect of age and sex on the expression of GABA signaling components-GABAA R α1,2,3,5, β1-3, γ2, GABAB R R1 and R2 subunits and the GABA synthesizing enzymes GAD 65/67-in the ECx, and the CA1 and DG regions of the human hippocampus using Western blotting. No significant differences were found in GABAA R α1,2,3,5, β1-3, γ2, GABAB R R1 and R2 subunit and GAD65/76 expression levels in the ECx, CA1 and DG regions between the younger and older age groups for both sexes. However, we observed a significant negative correlation between age and GABAA R α1subunit level in the CA1 region for females; significant negative correlation between age and GABAA R β1, β3 and γ2 subunit expression in the DG region for males. In females a significant positive correlation was found between age and GABAA R γ2 subunit expression in the ECx and GABAB R R2 subunit expression in the CA1 region. The results indicate that age and sex do not affect the expression of GAD 65/67. In conclusion, our results show age- and sex-related GABAA/B R subunit alterations in the ECx and hippocampus that might significantly influence GABAergic neurotransmission and underlie disease susceptibility and progression., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
18. Glutamatergic receptor expression changes in the Alzheimer's disease hippocampus and entorhinal cortex.
- Author
-
Yeung JHY, Walby JL, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease pathology, Entorhinal Cortex pathology, Female, Hippocampus pathology, Humans, Male, Middle Aged, Neurons metabolism, Neurons pathology, Alzheimer Disease metabolism, Entorhinal Cortex metabolism, Hippocampus metabolism, Receptors, Glutamate metabolism
- Abstract
Alzheimer's Disease (AD) is the leading form of dementia worldwide. Currently, the pathological mechanisms underlying AD are not well understood. Although the glutamatergic system is extensively implicated in its pathophysiology, there is a gap in knowledge regarding the expression of glutamate receptors in the AD brain. This study aimed to characterize the expression of specific glutamate receptor subunits in post-mortem human brain tissue using immunohistochemistry and confocal microscopy. Free-floating immunohistochemistry and confocal laser scanning microscopy were used to quantify the density of glutamate receptor subunits GluA2, GluN1, and GluN2A in specific cell layers of the hippocampal sub-regions, subiculum, entorhinal cortex, and superior temporal gyrus. Quantification of GluA2 expression in human post-mortem hippocampus revealed a significant increase in the stratum (str.) moleculare of the dentate gyrus (DG) in AD compared with control. Increased GluN1 receptor expression was found in the str. moleculare and hilus of the DG, str. oriens of the CA2 and CA3, str. pyramidale of the CA2, and str. radiatum of the CA1, CA2, and CA3 subregions and the entorhinal cortex. GluN2A expression was significantly increased in AD compared with control in the str. oriens, str. pyramidale, and str. radiatum of the CA1 subregion. These findings indicate that the expression of glutamatergic receptor subunits shows brain region-specific changes in AD, suggesting possible pathological receptor functioning. These results provide evidence of specific glutamatergic receptor subunit changes in the AD hippocampus and entorhinal cortex, indicating the requirement for further research to elucidate the pathophysiological mechanisms it entails, and further highlight the potential of glutamatergic receptor subunits as therapeutic targets., (© 2021 The Authors. Brain Pathology published by John Wiley & Sons Ltd on behalf of International Society of Neuropathology.)
- Published
- 2021
- Full Text
- View/download PDF
19. EAAT2 Expression in the Hippocampus, Subiculum, Entorhinal Cortex and Superior Temporal Gyrus in Alzheimer's Disease.
- Author
-
Yeung JHY, Palpagama TH, Wood OWG, Turner C, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Abstract
Alzheimer's disease (AD) is a neuropathological disorder characterized by the presence and accumulation of amyloid-beta plaques and neurofibrillary tangles. Glutamate dysregulation and the concept of glutamatergic excitotoxicity have been frequently described in the pathogenesis of a variety of neurodegenerative disorders and are postulated to play a major role in the progression of AD. In particular, alterations in homeostatic mechanisms, such as glutamate uptake, have been implicated in AD. An association with excitatory amino acid transporter 2 (EAAT2), the main glutamate uptake transporter, dysfunction has also been described. Several animal and few human studies examined EAAT2 expression in multiple brain regions in AD but studies of the hippocampus, the most severely affected brain region, are scarce. Therefore, this study aims to assess alterations in the expression of EAAT2 qualitatively and quantitatively through DAB immunohistochemistry (IHC) and immunofluorescence within the hippocampus, subiculum, entorhinal cortex, and superior temporal gyrus (STG) regions, between human AD and control cases. Although no significant EAAT2 density changes were observed between control and AD cases, there appeared to be increased transporter expression most likely localized to fine astrocytic branches in the neuropil as seen on both DAB IHC and immunofluorescence. Therefore, individual astrocytes are not outlined by EAAT2 staining and are not easily recognizable in the CA1-3 and dentate gyrus regions of AD cases, but the altered expression patterns observed between AD and control hippocampal cases could indicate alterations in glutamate recycling and potentially disturbed glutamatergic homeostasis. In conclusion, no significant EAAT2 density changes were found between control and AD cases, but the observed spatial differences in transporter expression and their functional significance will have to be further explored., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Yeung, Palpagama, Wood, Turner, Waldvogel, Faull and Kwakowsky.)
- Published
- 2021
- Full Text
- View/download PDF
20. Blood-spinal cord barrier leakage is independent of motor neuron pathology in ALS.
- Author
-
Waters S, Swanson MEV, Dieriks BV, Zhang YB, Grimsey NL, Murray HC, Turner C, Waldvogel HJ, Faull RLM, An J, Bowser R, Curtis MA, Dragunow M, and Scotter E
- Subjects
- Adult, Aged, Aged, 80 and over, Blood-Brain Barrier metabolism, Cerebrospinal Fluid Leak metabolism, Female, Hemoglobins cerebrospinal fluid, Humans, Male, Middle Aged, Motor Neurons metabolism, Spinal Cord metabolism, Amyotrophic Lateral Sclerosis cerebrospinal fluid, Amyotrophic Lateral Sclerosis pathology, Blood-Brain Barrier pathology, Cerebrospinal Fluid Leak pathology, Motor Neurons pathology, Spinal Cord pathology
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease involving progressive degeneration of upper and lower motor neurons. The pattern of lower motor neuron loss along the spinal cord follows the pattern of deposition of phosphorylated TDP-43 aggregates. The blood-spinal cord barrier (BSCB) restricts entry into the spinal cord parenchyma of blood components that can promote motor neuron degeneration, but in ALS there is evidence for barrier breakdown. Here we sought to quantify BSCB breakdown along the spinal cord axis, to determine whether BSCB breakdown displays the same patterning as motor neuron loss and TDP-43 proteinopathy. Cerebrospinal fluid hemoglobin was measured in living ALS patients (n = 87 control, n = 236 ALS) as a potential biomarker of BSCB and blood-brain barrier leakage. Cervical, thoracic, and lumbar post-mortem spinal cord tissue (n = 5 control, n = 13 ALS) were then immunolabelled and semi-automated imaging and analysis performed to quantify hemoglobin leakage, lower motor neuron loss, and phosphorylated TDP-43 inclusion load. Hemoglobin leakage was observed along the whole ALS spinal cord axis and was most severe in the dorsal gray and white matter in the thoracic spinal cord. In contrast, motor neuron loss and TDP-43 proteinopathy were seen at all three levels of the ALS spinal cord, with most abundant TDP-43 deposition in the anterior gray matter of the cervical and lumbar cord. Our data show that leakage of the BSCB occurs during life, but at end-stage disease the regions with most severe BSCB damage are not those where TDP-43 accumulation is most abundant. This suggests BSCB leakage and TDP-43 pathology are independent pathologies in ALS., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
21. Therapeutic potential of alpha 5 subunit containing GABA A receptors in Alzheimer's disease.
- Author
-
Kwakowsky A, Waldvogel HJ, and Faull RLM
- Abstract
Competing Interests: None
- Published
- 2021
- Full Text
- View/download PDF
22. The effects of amyloid-beta on hippocampal glutamatergic receptor and transporter expression.
- Author
-
Kwakowsky A, Waldvogel HJ, and Faull RL
- Abstract
Competing Interests: None
- Published
- 2021
- Full Text
- View/download PDF
23. The autocrine regulation of insulin-like growth factor-1 in human brain of Alzheimer's disease.
- Author
-
Kang D, Waldvogel HJ, Wang A, Fan D, Faull RLM, Curtis MA, Shorten PR, and Guan J
- Subjects
- Humans, Alzheimer Disease physiopathology, Brain metabolism, Insulin-Like Growth Factor I physiology
- Abstract
Background: Insulin-like growth factor (IGF) binding protein (IGFBP)-3 and cyclic Glycine-Proline (cGP) regulate circulating IGF-1 function that is associated with cognition. The association between IGF-1 function and Alzheimer's disease (AD) remains inconclusive. This study evaluated the changes of IGFBPs and cGP, and their effects on the bioavailability and function of IGF-1 in human brain of AD cases., Methods: Using biological and mathematic analysis we measured the concentrations of total, bound and unbound forms of IGF-1, IGFBPs and cGP in the inferior-frontal gyrus and middle-frontal gyrus of human AD (n = 15) and control cases (n = 15). The association between the changes of total concentration of these peptides and total protein concentration in brain tissues were also analyzed., Results: The unbound bioavailable IGF-1 was lower whereas the bound cGP and IGFBP-3 were higher in AD than the control cases. Total protein that was lower in AD than control cases, was negatively associated with cGP concentration of control cases and with IGFBP-3 concentration of AD cases., Conclusions: The results provide direct evidence for IGF-1 deficiency in AD brain due to lower bioavailable IGF-1. The increase of bound IGFBP-3 impaired autocrine regulation. The increase of bound cGP is an autocrine response to improve the bioavailability and function of IGF-1 in AD brain., Availability of Data and Material: All data generated or analysed during this study are included in this published article. Additional datasets analysed during the current study available from the corresponding author on reasonable request., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Promise and challenges of dystonia brain banking: establishing a human tissue repository for studies of X-Linked Dystonia-Parkinsonism.
- Author
-
Fernandez-Cerado C, Legarda GP, Velasco-Andrada MS, Aguil A, Ganza-Bautista NG, Lagarde JBB, Soria J, Jamora RDG, Acuña PJ, Vanderburg C, Sapp E, DiFiglia M, Murcar MG, Campion L, Ozelius LJ, Alessi AK, Singh-Bains MK, Waldvogel HJ, Faull RLM, Macalintal-Canlas R, Muñoz EL, Penney EB, Ang MA, Diesta CCE, Bragg DC, and Acuña-Sunshine G
- Subjects
- Brain diagnostic imaging, Genetic Diseases, X-Linked, Humans, Dystonia, Dystonic Disorders genetics, Neurodegenerative Diseases
- Abstract
X-Linked Dystonia-Parkinsonism (XDP) is a neurodegenerative disease affecting individuals with ancestry to the island of Panay in the Philippines. In recent years there has been considerable progress at elucidating the genetic basis of XDP and candidate disease mechanisms in patient-derived cellular models, but the neural substrates that give rise to XDP in vivo are still poorly understood. Previous studies of limited XDP postmortem brain samples have reported a selective dropout of medium spiny neurons within the striatum, although neuroimaging of XDP patients has detected additional abnormalities in multiple brain regions beyond the basal ganglia. Given the need to fully define the CNS structures that are affected in this disease, we created a brain bank in Panay to serve as a tissue resource for detailed studies of XDP-related neuropathology. Here we describe this platform, from donor recruitment and consent to tissue collection, processing, and storage, that was assembled within a predominantly rural region of the Philippines with limited access to medical and laboratory facilities. Thirty-six brains from XDP individuals have been collected over an initial 4 years period. Tissue quality was assessed based on histologic staining of cortex, RNA integrity scores, detection of neuronal transcripts in situ by fluorescent hybridization chain reaction, and western blotting of neuronal and glial proteins. The results indicate that this pipeline preserves tissue integrity to an extent compatible with a range of morphologic, molecular, and biochemical analyses. Thus the algorithms that we developed for working in rural communities may serve as a guide for establishing similar brain banks for other rare diseases in indigenous populations.
- Published
- 2021
- Full Text
- View/download PDF
25. A Multi-Omic Huntington's Disease Transgenic Sheep-Model Database for Investigating Disease Pathogenesis.
- Author
-
Mears ER, Handley RR, Grant MJ, Reid SJ, Day BT, Rudiger SR, McLaughlan CJ, Verma PJ, Bawden SC, Patassini S, Unwin RD, Cooper GJS, Gusella JF, MacDonald ME, Brauning R, Maclean P, Pearson JF, Waldvogel HJ, Faull RLM, and Snell RG
- Subjects
- Animals, Brain, Humans, Proteomics, Sheep, Huntington Disease genetics
- Abstract
Background: The pathological mechanism of cellular dysfunction and death in Huntington's disease (HD) is not well defined. Our transgenic HD sheep model (OVT73) was generated to investigate these mechanisms and for therapeutic testing. One particular cohort of animals has undergone focused investigation resulting in a large interrelated multi-omic dataset, with statistically significant changes observed comparing OVT73 and control 'omic' profiles and reported in literature., Objective: Here we make this dataset publicly available for the advancement of HD pathogenic mechanism discovery., Methods: To enable investigation in a user-friendly format, we integrated seven multi-omic datasets from a cohort of 5-year-old OVT73 (n = 6) and control (n = 6) sheep into a single database utilising the programming language R. It includes high-throughput transcriptomic, metabolomic and proteomic data from blood, brain, and other tissues., Results: We present the 'multi-omic' HD sheep database as a queryable web-based platform that can be used by the wider HD research community (https://hdsheep.cer.auckland.ac.nz/). The database is supported with a suite of simple automated statistical analysis functions for rapid exploratory analyses. We present examples of its use that validates the integrity relative to results previously reported. The data may also be downloaded for user determined analysis., Conclusion: We propose the use of this online database as a hypothesis generator and method to confirm/refute findings made from patient samples and alternate model systems, to expand our understanding of HD pathogenesis. Importantly, additional tissue samples are available for further investigation of this cohort.
- Published
- 2021
- Full Text
- View/download PDF
26. Neuroimaging and neuropathology studies of X-linked dystonia parkinsonism.
- Author
-
Arasaratnam CJ, Singh-Bains MK, Waldvogel HJ, and Faull RLM
- Subjects
- Basal Ganglia diagnostic imaging, Basal Ganglia pathology, Brain pathology, Diffusion Magnetic Resonance Imaging, Dystonic Disorders pathology, Genetic Diseases, X-Linked pathology, Humans, Magnetic Resonance Imaging, Neostriatum diagnostic imaging, Neostriatum pathology, Brain diagnostic imaging, Dystonic Disorders diagnostic imaging, Genetic Diseases, X-Linked diagnostic imaging
- Abstract
X-linked Dystonia Parkinsonism (XDP) is a recessive, genetically inherited neurodegenerative disorder endemic to Panay Island in the Philippines. Clinical symptoms include the initial appearance of dystonia, followed by parkinsonian traits after 10-15 years. The basal ganglia, particularly the striatum, is an area of focus in XDP neuropathology research, as the striatum shows marked atrophy that correlates with disease progression. Thus, XDP shares features of Parkinson's disease symptomatology, in addition to the genetic predisposition and presence of striatal atrophy resembling Huntington's disease. However, further research is required to reveal the detailed pathology and indicators of disease in the XDP brain. First, there are limited neuropathological studies that have investigated neuronal changes and neuroinflammation in the XDP brain. However, multiple neuroimaging studies on XDP patients provide clues to other affected brain regions. Furthermore, molecular pathological studies have elucidated that the main genetic cause of XDP is in the TAF-1 gene, but how this mutation relates to XDP neuropathology still remains to be fully investigated. Hence, we aim to provide an extensive overview of the current literature describing neuropathological changes within the XDP brain, and discuss future research avenues, which will provide a better understanding of XDP neuropathogenesis., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
27. Impaired Expression of GABA Signaling Components in the Alzheimer's Disease Middle Temporal Gyrus.
- Author
-
Govindpani K, Turner C, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease genetics, Autopsy, Female, GABA Plasma Membrane Transport Proteins genetics, GABA Plasma Membrane Transport Proteins metabolism, Gene Expression, Glutamate Decarboxylase genetics, Glutamate Decarboxylase metabolism, Humans, Male, Protein Isoforms genetics, Protein Isoforms metabolism, Receptors, GABA genetics, Receptors, GABA-A genetics, Alzheimer Disease metabolism, Receptors, GABA metabolism, Receptors, GABA-A metabolism, Signal Transduction, Temporal Lobe metabolism, gamma-Aminobutyric Acid metabolism
- Abstract
γ-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter, playing a central role in the regulation of cortical excitability and the maintenance of the excitatory/inhibitory (E/I) balance. Several lines of evidence point to a remodeling of the cerebral GABAergic system in Alzheimer's disease (AD), with past studies demonstrating alterations in GABA receptor and transporter expression, GABA synthesizing enzyme activity and focal GABA concentrations in post-mortem tissue. AD is a chronic neurodegenerative disorder with a poorly understood etiology and the temporal cortex is one of the earliest regions in the brain to be affected by AD neurodegeneration. Utilizing NanoString nCounter analysis, we demonstrate here the transcriptional downregulation of several GABA signaling components in the post-mortem human middle temporal gyrus (MTG) in AD, including the GABA
A receptor α1 , α2 , α3 , α5 , β1 , β2 , β3 , δ, γ2 , γ3 , and θ subunits and the GABAB receptor 2 (GABAB R2) subunit. In addition to this, we note the transcriptional upregulation of the betaine-GABA transporter (BGT1) and GABA transporter 2 (GAT2), and the downregulation of the 67 kDa isoform of glutamate decarboxylase (GAD67 ), the primary GABA synthesizing enzyme. The functional consequences of these changes require further investigation, but such alterations may underlie disruptions to the E/I balance that are believed to contribute to cognitive decline in AD.- Published
- 2020
- Full Text
- View/download PDF
28. The Interplay Between Beta-Amyloid 1-42 (Aβ 1-42 )-Induced Hippocampal Inflammatory Response, p-tau, Vascular Pathology, and Their Synergistic Contributions to Neuronal Death and Behavioral Deficits.
- Author
-
Calvo-Flores Guzmán B, Elizabeth Chaffey T, Hansika Palpagama T, Waters S, Boix J, Tate WP, Peppercorn K, Dragunow M, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Abstract
Alzheimer's disease (AD), the most common chronic neurodegenerative disorder, has complex neuropathology. The principal neuropathological hallmarks of the disease are the deposition of extracellular β-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs) comprised of hyperphosphorylated tau (p-tau) protein. These changes occur with neuroinflammation, a compromised blood-brain barrier (BBB) integrity, and neuronal synaptic dysfunction, all of which ultimately lead to neuronal cell loss and cognitive deficits in AD. Aβ
1-42 was stereotaxically administered bilaterally into the CA1 region of the hippocampi of 18-month-old male C57BL/6 mice. This study aimed to characterize, utilizing immunohistochemistry and behavioral testing, the spatial and temporal effects of Aβ1-42 on a broad set of parameters characteristic of AD: p-tau, neuroinflammation, vascular pathology, pyramidal cell survival, and behavior. Three days after Aβ1-42 injection and before significant neuronal cell loss was detected, acute neuroinflammatory and vascular responses were observed. These responses included the up-regulation of glial fibrillary acidic protein (GFAP), cell adhesion molecule-1 (PECAM-1, also known as CD31), fibrinogen labeling, and an increased number of activated astrocytes and microglia in the CA1 region of the hippocampus. From day 7, there was significant pyramidal cell loss in the CA1 region of the hippocampus, and by 30 days, significant localized up-regulation of p-tau, GFAP, Iba-1, CD31, and alpha-smooth muscle actin (α-SMA) in the Aβ1-42 -injected mice compared with controls. These molecular changes in Aβ1-42 -injected mice were accompanied by cognitive deterioration, as demonstrated by long-term spatial memory impairment. This study is reporting a comprehensive examination of a complex set of parameters associated with intrahippocampal administration of Aβ1-42 in mice, their spatiotemporal interactions and combined contribution to the disease progression. We show that a single Aβ injection can reproduce aspects of the inflammatory, vascular, and p-tau induced pathology occurring in the AD human brain that lead to cognitive deficits., (Copyright © 2020 Calvo-Flores Guzmán, Chaffey, Palpagama, Waters, Boix, Tate, Peppercorn, Dragunow, Waldvogel, Faull and Kwakowsky.)- Published
- 2020
- Full Text
- View/download PDF
29. Amyloid-beta 1-42 induced glutamatergic receptor and transporter expression changes in the mouse hippocampus.
- Author
-
Yeung JHY, Calvo-Flores Guzmán B, Palpagama TH, Ethiraj J, Zhai Y, Tate WP, Peppercorn K, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Amyloid beta-Peptides pharmacology, Animals, CA1 Region, Hippocampal drug effects, CA1 Region, Hippocampal metabolism, CA3 Region, Hippocampal drug effects, CA3 Region, Hippocampal metabolism, Dentate Gyrus drug effects, Dentate Gyrus metabolism, Hippocampus drug effects, Immunohistochemistry, Male, Mice, Mice, Inbred C57BL, Peptide Fragments pharmacology, Receptors, AMPA genetics, Vesicular Glutamate Transport Protein 1 genetics, Amyloid beta-Peptides toxicity, Hippocampus metabolism, Peptide Fragments toxicity, Receptors, AMPA biosynthesis, Vesicular Glutamate Transport Protein 1 biosynthesis
- Abstract
Alzheimer's disease (AD) is the leading type of dementia worldwide. With an increasing burden of an aging population coupled with the lack of any foreseeable cure, AD warrants the current intense research effort on the toxic effects of an increased concentration of beta-amyloid (Aβ) in the brain. Glutamate is the main excitatory brain neurotransmitter and it plays an essential role in the function and health of neurons and neuronal excitability. While previous studies have shown alterations in expression of glutamatergic signaling components in AD, the underlying mechanisms of these changes are not well understood. This is the first comprehensive anatomical study to characterize the subregion- and cell layer-specific long-term effect of Aβ
1-42 on the expression of specific glutamate receptors and transporters in the mouse hippocampus, using immunohistochemistry with confocal microscopy. Outcomes are examined 30 days after Aβ1-42 stereotactic injection in aged male C57BL/6 mice. We report significant decreases in density of the glutamate receptor subunit GluA1 and the vesicular glutamate transporter (VGluT) 1 in the conus ammonis 1 region of the hippocampus in the Aβ1-42 injected mice compared with artificial cerebrospinal fluid injected and naïve controls, notably in the stratum oriens and stratum radiatum. GluA1 subunit density also decreased within the dentate gyrus dorsal stratum moleculare in Aβ1-42 injected mice compared with artificial cerebrospinal fluid injected controls. These changes are consistent with findings previously reported in the human AD hippocampus. By contrast, glutamate receptor subunits GluA2, GluN1, GluN2A, and VGluT2 showed no changes in expression. These findings indicate that Aβ1-42 induces brain region and layer specific expression changes of the glutamatergic receptors and transporters, suggesting complex and spatial vulnerability of this pathway during development of AD neuropathology. Read the Editorial Highlight for this article on page 7. Cover Image for this issue: https://doi.org/10.1111/jnc.14763., (© 2020 International Society for Neurochemistry.)- Published
- 2020
- Full Text
- View/download PDF
30. Cerebral deficiency of vitamin B5 (d-pantothenic acid; pantothenate) as a potentially-reversible cause of neurodegeneration and dementia in sporadic Alzheimer's disease.
- Author
-
Xu J, Patassini S, Begley P, Church S, Waldvogel HJ, Faull RLM, Unwin RD, and Cooper GJS
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease etiology, Alzheimer Disease pathology, Brain pathology, Brain Chemistry, Case-Control Studies, Female, Humans, Male, Middle Aged, Pantothenic Acid analysis, Pantothenic Acid metabolism, Alzheimer Disease metabolism, Brain metabolism, Pantothenic Acid deficiency
- Abstract
Alzheimer's disease (AD) is the most common cause of age-related neurodegeneration and dementia, and there are no available treatments with proven disease-modifying actions. It is therefore appropriate to study hitherto-unknown aspects of brain structure/function in AD to seek alternative disease-related mechanisms that might be targeted by new therapeutic interventions with disease-modifying actions. During hypothesis-generating metabolomic studies of brain, we identified apparent differences in levels of vitamin B5 between AD cases and controls. We therefore developed a method based on gas chromatography-mass spectrometry by which we quantitated vitamin B5 concentrations in seven brain regions from nine AD cases and nine controls. We found that widespread, severe cerebral deficiency of vitamin B5 occurs in AD. This deficiency was worse in those regions known to undergo severe damage, including the hippocampus, entorhinal cortex, and middle temporal gyrus. Vitamin B5 is the obligate precursor of CoA/acetyl-CoA (acetyl-coenzyme A), which plays myriad key roles in the metabolism of all organs, including the brain. In brain, acetyl-CoA is the obligate precursor of the neurotransmitter acetylcholine, and the complex fatty-acyl groups that mediate the essential insulator role of myelin, both processes being defective in AD; moreover, the large cerebral vitamin B5 concentrations co-localize almost entirely to white matter. Vitamin B5 is well tolerated when administered orally to humans and other mammals. We conclude that cerebral vitamin B5 deficiency may well cause neurodegeneration and dementia in AD, which might be preventable or even reversible in its early stages, by treatment with suitable oral doses of vitamin B5., Competing Interests: Declaration of competing interest The authors state that they have no competing interests with respect to this work. Sponsors had no role in the study design; the collection, analysis, and interpretation of data; the writing of the manuscript, or the decision to submit the article for publication. This work was generated during previous employment of JX and SP, and not during their current employment., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
31. No symphony without bassoon and piccolo: changes in synaptic active zone proteins in Huntington's disease.
- Author
-
Huang TT, Smith R, Bacos K, Song DY, Faull RM, Waldvogel HJ, and Li JY
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Dendritic Spines pathology, Gene Expression, HEK293 Cells, Hippocampus metabolism, Hippocampus pathology, Humans, Huntingtin Protein genetics, Huntington Disease genetics, Male, Mice, Transgenic, Middle Aged, Mutation, RNA, Messenger metabolism, Cytoskeletal Proteins metabolism, Huntington Disease metabolism, Huntington Disease pathology, Nerve Tissue Proteins metabolism, Neuropeptides metabolism, Synapses metabolism, Synapses pathology
- Abstract
Prominent features of HD neuropathology are the intranuclear and cytoplasmic inclusions of huntingtin and striatal and cortical neuronal cell death. Recently, synaptic defects have been reported on HD-related studies, including impairment of neurotransmitter release and alterations of synaptic components. However, the definite characteristics of synapse dysfunction and the underlying mechanisms remain largely unknown. We studied the gene expression levels and patterns of a number of proteins forming the cytoskeletal matrix of the presynaptic active zones in HD transgenic mice (R6/1), in hippocampal neuronal cultures overexpressing mutant huntingtin and in postmortem brain tissues of HD patients. To investigate the interactions between huntingtin and active proteins, we performed confocal microscopic imaging and immunoprecipitation in mouse and HEK 293 cell line models. The mRNA and protein levels of Bassoon were reduced in mouse and cell culture models of HD and in brain tissues of patients with HD. Moreover, a striking re-distribution of a complex of proteins including Bassoon, Piccolo and Munc 13-1 from the cytoplasm and synapses into intranuclear huntingtin aggregates with loss of active zone proteins and dendritic spines. This re-localization was age-dependent and coincided with the formation of huntingtin aggregates. Using co-immunoprecipitation, we demonstrated that huntingtin interacts with Bassoon, and that this interaction is likely mediated by a third linking protein. Three structural proteins involved in neurotransmitter release in the presynaptic active zones of neurons are altered in expression and that the proteins are redistributed from their normal functional site into mutant huntingtin aggregates.
- Published
- 2020
- Full Text
- View/download PDF
32. Vascular dysfunction in Alzheimer's disease: a biomarker of disease progression and a potential therapeutic target.
- Author
-
Govindpani K, Vinnakota C, Waldvogel HJ, Faull RL, and Kwakowsky A
- Abstract
Competing Interests: None
- Published
- 2020
- Full Text
- View/download PDF
33. An 5 GABAA Receptor Inverse Agonist, 5IA, Attenuates Amyloid Beta-Induced Neuronal Death in Mouse Hippocampal Cultures.
- Author
-
Vinnakota C, Govindpani K, Tate WP, Peppercorn K, Anekal PV, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Animals, Cell Death, Cells, Cultured, GABAergic Neurons metabolism, GABAergic Neurons physiology, Hippocampus cytology, Male, Mice, Mice, Inbred C57BL, Receptors, GABA-A genetics, Receptors, GABA-A metabolism, Synaptic Transmission, Amyloid beta-Peptides toxicity, GABA-A Receptor Agonists pharmacology, GABAergic Neurons drug effects, Neuroprotective Agents pharmacology, Peptide Fragments toxicity, Phthalazines pharmacology, Triazoles pharmacology
- Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which no cognition-restoring therapies exist. Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the brain. Increasing evidence suggests a remodeling of the GABAergic system in AD, which might represent an important therapeutic target. An inverse agonist of 5 subunit-containing GABAA receptors (α5GABAARs), 3-(5-Methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-yl)methyloxy]-1,2,4-triazolo[3- a ]phthalazine (5IA) has cognition-enhancing properties. This study aimed to characterize the effects of 5IA on amyloid beta (A
1 - 42 )-induced molecular and cellular changes. Mouse primary hippocampal cultures were exposed to either A1-42 alone, or 5IA alone, 5IA with A1 - 42 or vehicle alone, and changes in cell viability and mRNA expression of several GABAergic signaling components were assessed. Treatment with 100 nM of 5IA reduced A1 - 42 -induced cell loss by 23.8% ( p < 0.0001) after 6 h and by 17.3% after 5 days of treatment ( p < 0.0001). Furthermore, we observed an A1-42 -induced increase in ambient GABA levels, as well as upregulated mRNA expression of the GABAAR α2,α5,2/3 subunits and the GABABR R1 and R2 subunits. Such changes in GABARs expression could potentially disrupt inhibitory neurotransmission and normal network activity. Treatment with 5IA restored A1-42 -induced changes in the expression of α5GABAARs. In summary, this compound might hold neuroprotective potential and represent a new therapeutic avenue for AD., Competing Interests: The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.- Published
- 2020
- Full Text
- View/download PDF
34. Amyloid-Beta 1-42 -Induced Increase in GABAergic Tonic Conductance in Mouse Hippocampal CA1 Pyramidal Cells.
- Author
-
Calvo-Flores Guzmán B, Kim S, Chawdhary B, Peppercorn K, Tate WP, Waldvogel HJ, Faull RL, Montgomery J, and Kwakowsky A
- Subjects
- Alzheimer Disease metabolism, Animals, Male, Memory physiology, Mice, Mice, Inbred C57BL, Receptors, GABA-A metabolism, Synapses metabolism, Synaptic Transmission physiology, Amyloid beta-Peptides metabolism, CA1 Region, Hippocampal metabolism, Hippocampus metabolism, Pyramidal Cells metabolism, gamma-Aminobutyric Acid metabolism
- Abstract
Alzheimer's disease (AD) is a complex and chronic neurodegenerative disorder that involves a progressive and severe decline in cognition and memory. During the last few decades a considerable amount of research has been done in order to better understand tau-pathology, inflammatory activity and neuronal synapse loss in AD, all of them contributing to cognitive decline. Early hippocampal network dysfunction is one of the main factors associated with cognitive decline in AD. Much has been published about amyloid-beta
1-42 (Aβ1-42 )-mediated excitotoxicity in AD. However, increasing evidence demonstrates that the remodeling of the inhibitory gamma-aminobutyric acid (GABAergic) system contributes to the excitatory/inhibitory (E/I) disruption in the AD hippocampus, but the underlying mechanisms are not well understood. In the present study, we show that hippocampal injection of Aβ1-42 is sufficient to induce cognitive deficits 7 days post-injection. We demonstrate using in vitro whole-cell patch-clamping an increased inhibitory GABAergic tonic conductance mediated by extrasynaptic type A GABA receptors (GABAA Rs), recorded in the CA1 region of the mouse hippocampus following Aβ1-42 micro injection. Such alterations in GABA neurotransmission and/or inhibitory GABAA Rs could have a significant impact on both hippocampal structure and function, causing E/I balance disruption and potentially contributing to cognitive deficits in AD.- Published
- 2020
- Full Text
- View/download PDF
35. The Acute Effects of Amyloid-Beta 1-42 on Glutamatergic Receptor and Transporter Expression in the Mouse Hippocampus.
- Author
-
Yeung JHY, Palpagama TH, Tate WP, Peppercorn K, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Abstract
Alzheimer's disease (AD) is the leading type of dementia worldwide. Despite an increasing burden of disease due to a rapidly aging population, there is still a lack of complete understanding of the precise pathological mechanisms which drive its progression. Glutamate is the main excitatory neurotransmitter in the brain and plays an essential role in the normal function and excitability of neuronal networks. While previous studies have shown alterations in the function of the glutamatergic system in AD, the underlying etiology of beta amyloid (Aβ
1-42 ) induced changes has not been explored. Here we have investigated the acute effects of stereotaxic hippocampal Aβ1-42 injection on specific glutamatergic receptors and transporters in the mouse hippocampus, using immunohistochemistry and confocal microscopy 3 days after Aβ1-42 injection in aged male C57BL/6 mice, before the onset of neuronal cell death. We show that acute injection of Aβ1-42 is sufficient to induce cognitive deficits 3 days post-injection. We also report no significant changes in glutamate receptor subunits GluA1, GluA2, VGluT1, and VGluT2 in response to acute injection of Aβ1-42 when compared with the ACSF-vehicle injected mice. However, we observed increased expression in the DG hilus and ventral stratum (str.) granulosum, CA3 str. radiatum and str. oriens, and CA1 str. radiatum of the GluN1 subunit, and increased expression within the CA3 str. radiatum and decreased expression within the DG str. granulosum of the GluN2A subunit in Aβ1-42 injected mice compared to NC, and a similar trend observed when compared to ACSF-injected mice. We also observed alterations in expression patterns of glutamatergic receptor subunits and transporters within specific layers of hippocampal subregions in response to a microinjection stimulus. These findings indicate that the pathological alterations in the glutamatergic system observed in AD are likely to be partially a result of both acute and chronic exposure to Aβ1-42 and implies a much more complex circuit mechanism associated with glutamatergic dysfunction than simply glutamate-mediated excitotoxic neuronal death., (Copyright © 2020 Yeung, Palpagama, Tate, Peppercorn, Waldvogel, Faull and Kwakowsky.)- Published
- 2020
- Full Text
- View/download PDF
36. The Role of Microglia and Astrocytes in Huntington's Disease.
- Author
-
Palpagama TH, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease. HD patients present with movement disorders, behavioral and psychiatric symptoms and cognitive decline. This review summarizes the contribution of microglia and astrocytes to HD pathophysiology. Neuroinflammation in the HD brain is characterized by a reactive morphology in these glial cells. Microglia and astrocytes are critical in regulating neuronal activity and maintaining an optimal milieu for neuronal function. Previous studies provide evidence that activated microglia and reactive astrocytes contribute to HD pathology through transcriptional activation of pro-inflammatory genes to perpetuate a chronic inflammatory state. Reactive astrocytes also display functional changes in glutamate and ion homeostasis and energy metabolism. Astrocytic and microglial changes may further contribute to the neuronal death observed with the progression of HD. Importantly, the degree to which these neuroinflammatory changes are detrimental to neurons and contribute to the progression of HD pathology is not well understood. Furthermore, recent observations provide compelling evidence that activated microglia and astrocytes exert a variety of beneficial functions that are essential for limiting tissue damage and preserving neuronal function in the HD brain. Therefore, a better understanding of the neuroinflammatory environment in the brain in HD may lead to the development of targeted and innovative therapeutic opportunities., (Copyright © 2019 Palpagama, Waldvogel, Faull and Kwakowsky.)
- Published
- 2019
- Full Text
- View/download PDF
37. GABA A Receptors Are Well Preserved in the Hippocampus of Aged Mice.
- Author
-
Palpagama TH, Sagniez M, Kim S, Waldvogel HJ, Faull RL, and Kwakowsky A
- Subjects
- Animals, Axon Initial Segment metabolism, Male, Mice, Inbred C57BL, Aging metabolism, Hippocampus metabolism, Protein Subunits metabolism, Pyramidal Cells metabolism, Receptors, GABA-A metabolism
- Abstract
GABA is the primary inhibitory neurotransmitter in the nervous system. GABA
A receptors (GABAA Rs) are pentameric ionotropic channels. Subunit composition of the receptors is associated with the affinity of GABA binding and its downstream inhibitory actions. Fluctuations in subunit expression levels with increasing age have been demonstrated in animal and human studies. However, our knowledge regarding the age-related hippocampal GABAA R expression changes is limited and based on rat studies. This study is the first analysis of the aging-related changes of the GABAA R subunit expression in the CA1, CA2/3, and dentate gyrus regions of the mouse hippocampus. Using Western blotting and immunohistochemistry we found that the GABAergic system is robust, with no significant age-related differences in GABAA R α1, α2, α3, α5, β3, and γ2 subunit expression level differences found between the young (6 months) and old (21 months) age groups in any of the hippocampal regions examined. However, we detected a localized decrease of α2 subunit expression around the soma, proximal dendrites, and in the axon initial segment of pyramidal cells in the CA1 and CA3 regions that is accompanied by a pronounced upregulation of the α2 subunit immunoreactivity in the neuropil of aged mice. In summary, GABAA Rs are well preserved in the mouse hippocampus during normal aging although GABAA Rs in the hippocampus are severely affected in age-related neurological disorders, including Alzheimer's disease., (Copyright © 2019 Palpagama et al.)- Published
- 2019
- Full Text
- View/download PDF
38. Cerebral Vitamin B5 (D-Pantothenic Acid) Deficiency as a Potential Cause of Metabolic Perturbation and Neurodegeneration in Huntington's Disease.
- Author
-
Patassini S, Begley P, Xu J, Church SJ, Kureishy N, Reid SJ, Waldvogel HJ, Faull RLM, Snell RG, Unwin RD, and Cooper GJS
- Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded CAG repeat in exon 1 of the HTT gene. HD usually manifests in mid-life with loss of GABAergic projection neurons from the striatum accompanied by progressive atrophy of the putamen followed by other brain regions, but linkages between the genetics and neurodegeneration are not understood. We measured metabolic perturbations in HD-human brain in a case-control study, identifying pervasive lowering of vitamin B5, the obligatory precursor of coenzyme A (CoA) that is essential for normal intermediary metabolism. Cerebral pantothenate deficiency is a newly-identified metabolic defect in human HD that could potentially: (i) impair neuronal CoA biosynthesis; (ii) stimulate polyol-pathway activity; (iii) impair glycolysis and tricarboxylic acid cycle activity; and (iv) modify brain-urea metabolism. Pantothenate deficiency could lead to neurodegeneration/dementia in HD that might be preventable by treatment with vitamin B5.
- Published
- 2019
- Full Text
- View/download PDF
39. Vascular Dysfunction in Alzheimer's Disease: A Prelude to the Pathological Process or a Consequence of It?
- Author
-
Govindpani K, McNamara LG, Smith NR, Vinnakota C, Waldvogel HJ, Faull RL, and Kwakowsky A
- Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Despite decades of research following several theoretical and clinical lines, all existing treatments for the disorder are purely symptomatic. AD research has traditionally been focused on neuronal and glial dysfunction. Although there is a wealth of evidence pointing to a significant vascular component in the disease, this angle has been relatively poorly explored. In this review, we consider the various aspects of vascular dysfunction in AD, which has a significant impact on brain metabolism and homeostasis and the clearance of β-amyloid and other toxic metabolites. This may potentially precede the onset of the hallmark pathophysiological and cognitive symptoms of the disease. Pathological changes in vessel haemodynamics, angiogenesis, vascular cell function, vascular coverage, blood-brain barrier permeability and immune cell migration may be related to amyloid toxicity, oxidative stress and apolipoprotein E (APOE) genotype. These vascular deficits may in turn contribute to parenchymal amyloid deposition, neurotoxicity, glial activation and metabolic dysfunction in multiple cell types. A vicious feedback cycle ensues, with progressively worsening neuronal and vascular pathology through the course of the disease. Thus, a better appreciation for the importance of vascular dysfunction in AD may open new avenues for research and therapy., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
40. Chemical neuroanatomy of the substantia nigra in the ovine brain.
- Author
-
Murray SJ, Black BL, Reid SJ, Rudiger SR, Simon Bawden C, Snell RG, Waldvogel HJ, and Faull RLM
- Subjects
- Animals, Sheep, Substantia Nigra anatomy & histology, Substantia Nigra metabolism
- Abstract
The substantia nigra is an integral component of the basal ganglia circuitry for limbic and motor functions. Dysfunction and degeneration of the basal ganglia are fundamental aspects of neurodegenerative diseases such as Parkinson's disease and Huntington's disease. With the increasing use of sheep to model neurological diseases, it is crucial to understand the anatomy and neurochemistry of these key basal ganglia nuclei in the normal sheep brain and how they compare to the human brain. Therefore, studies of the gross anatomy, cellular morphology, and neurochemical expression patterns within the sheep substantia nigra were performed. We show that the sheep substantia nigra reflects all important aspects of the anatomy and neurochemistry of the human substantia nigra, with only minor inter-species differences evident. Many neurochemicals that are central to the functioning of the SN, and wider basal ganglia circuitry, are present throughout the sheep SN. In a wider context, the results of this study provide evidence that the sheep substantia nigra accurately reflects the anatomy of the human substantia nigra, which validates the use of sheep models of basal ganglia neurological disorders., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
41. Variable colocalisation of GABA A receptor subunits and glycine receptors on neurons in the human hypoglossal nucleus.
- Author
-
Waldvogel HJ, Biggins FM, Singh A, Arasaratnam CJ, and Faull RLM
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Hypoglossal Nerve metabolism, Male, Middle Aged, Motor Neurons metabolism, Medulla Oblongata metabolism, Neurons metabolism, Receptors, GABA-A metabolism, Receptors, Glycine metabolism
- Abstract
The hypoglossal nucleus, the nucleus of the twelfth cranial nerve, is located dorsally in the midline of the medulla oblongata. The hypoglossal nucleus contains lower motor neurons which innervate the tongue muscles that control tongue movements involved in speech production, swallowing, mastication and associated respiratory movements. GABA
A and glycine receptors are heteropentameric ionotropic receptors that facilitate fast-response, inhibitory neurotransmission in the mammalian brain and spinal cord. We investigated the immunohistochemical distribution of the GABAA receptor α1 , α2 , β2,3 subunits and glycine receptors as well as their relationship to the vesicular GABA transporter (VGAT) in the human hypoglossal nucleus at the light and confocal laser scanning microscope levels. The results showed that all of the GABAA receptor subunits as well as glycine receptor display punctate labelling indicative of synapses on the soma and dendritic membranes of large neurons within the hypoglossal nucleus. On average, approximately 50% of glycine receptors were co localised with GABAA receptor α1 subunits. Also on average GABAA α2 and β2,3 subunits were colocalised with approximately 30% of glycine receptor subunits. VGAT positive terminals were associated with both GABAA and glycine receptor types. Both glycinergic and GABAergic positive puncta were found adjacent to VGAT terminal-like staining. These results suggest that inhibition of human hypoglossal motor neurons occurs not only through complex interaction of separated GABAA R and glycine receptor regions, but also through synapses containing both inhibitory receptor types co-existing at the same synaptic sites., (Copyright © 2019 Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
42. Cerebellar degeneration correlates with motor symptoms in Huntington disease.
- Author
-
Singh-Bains MK, Mehrabi NF, Sehji T, Austria MDR, Tan AYS, Tippett LJ, Dragunow M, Waldvogel HJ, and Faull RLM
- Subjects
- Adult, Aged, Autopsy, Brain pathology, Case-Control Studies, Cell Count, Corpus Striatum pathology, Female, Humans, Male, Middle Aged, Neurodegenerative Diseases pathology, Phenotype, Cerebellum pathology, Huntington Disease pathology, Huntington Disease physiopathology, Huntington Disease psychology, Purkinje Cells pathology
- Abstract
Objective: Huntington disease (HD) is an autosomal dominant neurodegenerative disorder characterized by variable motor and behavioral symptoms attributed to major neuropathology of mainly the basal ganglia and cerebral cortex. The role of the cerebellum, a brain region involved in the coordination of movements, in HD neuropathology has been controversial. This study utilizes postmortem human brain tissue to investigate whether Purkinje cell degeneration in the neocerebellum is present in HD, and how this relates to disease symptom profiles., Methods: Unbiased stereological counting methods were used to quantify the total number of Purkinje cells in 15 HD cases and 8 neurologically normal control cases. Based on their predominant symptoms, the HD cases were categorized into 2 groups: "motor" or "mood.", Results: The results demonstrated a significant 43% loss of Purkinje cells in HD cases with predominantly motor symptoms, and no cell loss in cases showing a major mood phenotype. There was no significant correlation between Purkinje cell loss and striatal neuropathological grade, postmortem delay, CAG repeat in the IT15 gene, or age at death., Interpretation: This study shows a compelling relationship between Purkinje cell loss in the HD neocerebellum and the HD motor symptom phenotype, which, together with our previous human brain studies on the same HD cases, provides novel perspectives interrelating and correlating the variable cerebellar, basal ganglia, and neocortical neuropathology with the variability of motor/mood symptom profiles in the human HD brain. ANN NEUROL 2019;85:396-405., (© 2019 The Authors. Annals of Neurology published by Wiley Periodicals, Inc. on behalf of American Neurological Association.)
- Published
- 2019
- Full Text
- View/download PDF
43. Sex- and age-related changes in GABA signaling components in the human cortex.
- Author
-
Pandya M, Palpagama TH, Turner C, Waldvogel HJ, Faull RL, and Kwakowsky A
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Signal Transduction, Aging metabolism, Cerebral Cortex metabolism, Sex Characteristics, gamma-Aminobutyric Acid metabolism
- Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the nervous system. Previous studies have shown fluctuations in expression levels of GABA signaling components-glutamic acid decarboxylase (GAD), GABA receptor (GABAR) subunit, and GABA transporter (GAT)-with increasing age and between sexes; however, this limited knowledge is highly based on animal models that produce inconsistent findings. This study is the first analysis of the age- and sex-specific changes of the GAD, GABA
A/B R subunits, and GAT expression in the human primary sensory and motor cortices; superior (STG), middle (MTG), and inferior temporal gyrus (ITG); and cerebellum. Utilizing Western blotting, we found that the GABAergic system is relatively robust against sex and age-related differences in all brain regions examined. However, we observed several sex-dependent differences in GABAA R subunit expression in STG along with age-dependent GABAA R subunit and GAD level alteration. No significant age-related differences were found in α1, α2, α5, β3, and γ2 subunit expression in the STG. However, we found significantly higher GABAA R α3 subunit expression in the STG in young males compared to old males. We observed a significant sex-dependent difference in α1 subunit expression: males presenting significantly higher levels compared to women across all stages of life in STG. Older females showed significantly lower α2, α5, and β3 subunit expression compared to old males in the STG. These changes found in the STG might significantly influence GABAergic neurotransmission and lead to sex- and age-specific disease susceptibility and progression.- Published
- 2019
- Full Text
- View/download PDF
44. The GABAergic system as a therapeutic target for Alzheimer's disease.
- Author
-
Calvo-Flores Guzmán B, Vinnakota C, Govindpani K, Waldvogel HJ, Faull RLM, and Kwakowsky A
- Subjects
- Animals, Humans, Signal Transduction physiology, Alzheimer Disease metabolism, Alzheimer Disease therapy, Signal Transduction drug effects, gamma-Aminobutyric Acid metabolism
- Abstract
Glutamatergic and cholinergic dysfunction are well-attested features of Alzheimer's disease (AD), progressing with other pathological indices of the disorder and exacerbating neuronal and network dysfunction. However, relatively little attention has been paid to the inhibitory component of the excitatory/inhibitory (E/I) network, particularly dysfunction in the gamma-aminobutyric acid (GABA) signaling system. There is growing evidence in support of GABAergic remodeling in the AD brain, potentially beginning in early stages of disease pathogenesis, and this could thus be a valid molecular target for drug development and pharmacological therapies. Several GABAergic drugs have been tested for efficacy in attenuating or reversing various features and symptoms of AD, and this could represent a novel path by which we might address the growing need for more effective and benign therapies., (© 2018 International Society for Neurochemistry.)
- Published
- 2018
- Full Text
- View/download PDF
45. Gamma-aminobutyric acid A receptors in Alzheimer's disease: highly localized remodeling of a complex and diverse signaling pathway.
- Author
-
Kwakowsky A, Calvo-Flores Guzmán B, Govindpani K, Waldvogel HJ, and Faull RL
- Abstract
Competing Interests: None declared
- Published
- 2018
- Full Text
- View/download PDF
46. GABA A receptor subunit expression changes in the human Alzheimer's disease hippocampus, subiculum, entorhinal cortex and superior temporal gyrus.
- Author
-
Kwakowsky A, Calvo-Flores Guzmán B, Pandya M, Turner C, Waldvogel HJ, and Faull RL
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease pathology, Brain pathology, Female, Humans, Male, Alzheimer Disease metabolism, Brain metabolism, Receptors, GABA-A biosynthesis
- Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. GABA type A receptors (GABA
A Rs) are severely affected in Alzheimer's disease (AD). However, the distribution and subunit composition of GABAA Rs in the AD brain are not well understood. This is the first comprehensive study to show brain region- and cell layer-specific alterations in the expression of the GABAA R subunits α1-3, α5, β1-3 and γ2 in the human AD hippocampus, entorhinal cortex and superior temporal gyrus. In late-stage AD tissue samples using immunohistochemistry we found significant alteration of all investigated GABAA Rs subunits except for α3 and β1 that were well preserved. The most prominent changes include an increase in GABAA R α1 expression associated with AD in all layers of the CA3 region, in the stratum (str.) granulare and hilus of the dentate gyrus. We found a significant increase in GABAA R α2 expression in the str. oriens of the CA1-3, str. radiatum of the CA2,3 and decrease in the str. pyramidale of the CA1 region in AD cases. In AD there was a significant increase in GABAA R α5 subunit expression in str. pyramidale, str. oriens of the CA1 region and decrease in the superior temporal gyrus. We also found a significant decrease in the GABAA R β3 subunit immunoreactivity in the str. oriens of the CA2, str. granulare and str. moleculare of the dentate gyrus. In conclusion, these findings indicate that the expression of the GABAA R subunits shows brain region- and layer-specific alterations in AD, and these changes could significantly influence and alter GABAA R function in the disease. Cover Image for this issue: doi: 10.1111/jnc.14179., (© 2018 International Society for Neurochemistry.)- Published
- 2018
- Full Text
- View/download PDF
47. GABA A and GABA B receptor subunit localization on neurochemically identified neurons of the human subthalamic nucleus.
- Author
-
Wu XH, Song JJ, Faull RLM, and Waldvogel HJ
- Subjects
- Adult, Aged, Aged, 80 and over, Calbindin 2 metabolism, Female, Glutamate Decarboxylase metabolism, Humans, Male, Middle Aged, Neurofilament Proteins metabolism, Parvalbumins metabolism, Protein Subunits metabolism, Neurons metabolism, Receptors, GABA-A metabolism, Receptors, GABA-B metabolism, Subthalamic Nucleus cytology, gamma-Aminobutyric Acid metabolism
- Abstract
The subthalamic nucleus (STN) is a critical excitatory signaling center within the basal ganglia circuitry. The activity of subthalamic neurons is tightly controlled by upstream inhibitory signaling centers in the basal ganglia. In this study, we used immunohistochemical techniques to firstly, visualize and quantify the STN neurochemical organization based on neuronal markers including parvalbumin (PV), calretinin (CR), SMI-32, and GAD
65/67 . Secondly, we characterized the detailed regional, cellular and subcellular expression of GABAA (α1 , α2 , α3 , β2/3 , and γ2 ) and GABAB (R1 and R2) receptor subunits within the normal human STN. Overall, we found seven neurochemically distinct populations of principal neurons in the human STN. The three main populations detected were: (a) triple-labeled PV+ /CR+ /SMI32+ ; (b) double-labeled PV+ /CR+ ; and (c) single-labeled CR+ neurons. Subthalamic principal neurons were found to express GABAA receptor subunits α1 , α3 , β2/3 , γ2 , and GABAB receptor subunits R1 and R2. However, no expression of GABAA receptor α2 subunit was detected. We also found a trend of increasing regional staining intensity for all positive GABAA receptor subunits from the dorsolateral pole to ventromedial extremities. The GAD+ interneurons showed relatively low expression of GABAA receptor subunits. These results provide the morphological basis of GABAergic transmission within the normal human subthalamic nucleus and evidence of GABA innervation through both GABAA and GABAB receptors on subthalamic principal neurons., (© 2017 Wiley Periodicals, Inc.)- Published
- 2018
- Full Text
- View/download PDF
48. Stereological Methods to Quantify Cell Loss in the Huntington's Disease Human Brain.
- Author
-
Mehrabi NF, Singh-Bains MK, Waldvogel HJ, and Faull RLM
- Subjects
- Brain pathology, Cell Count instrumentation, Humans, Huntington Disease diagnosis, Imaging, Three-Dimensional instrumentation, Immunohistochemistry instrumentation, Immunohistochemistry methods, Microscopy instrumentation, Microscopy methods, Software, Brain cytology, Cell Count methods, Huntington Disease pathology, Imaging, Three-Dimensional methods
- Abstract
Design-based stereology is a quantification method to obtain a precise and unbiased estimate of the total number of cells (or any other objects) in a well-defined region of interest. There are two comparable stereological counting methods, (a) the Optical Fractionator and (b) the Nv:Vref method. Due to the adherence to strict stereological protocol, the Optical Fractionator is the most unbiased and preferable stereological method. However, the Nv:Vref method can be an alternative when tissue availability is limited. Both methods use systematic random sampling (SRS) techniques to account for the inhomogeneous nature of biological tissue. Here we describe the criteria for a successful and accurate stereological study, using human brain tissue.
- Published
- 2018
- Full Text
- View/download PDF
49. Differential Fatty Acid-Binding Protein Expression in Persistent Radial Glia in the Human and Sheep Subventricular Zone.
- Author
-
Dieriks BV, Dean JM, Aronica E, Waldvogel HJ, Faull RLM, and Curtis MA
- Subjects
- Adult, Aged, Animals, Female, Fetus, Humans, Male, Middle Aged, Sheep, Species Specificity, Ependymoglial Cells metabolism, Fatty Acid-Binding Proteins biosynthesis, Lateral Ventricles metabolism, Neurogenesis physiology
- Abstract
Fatty acid-binding proteins (FABPs) are a family of transport proteins that facilitate intracellular transport of fatty acids. Despite abundant expression in the brain, the role that FABPs play in the process of cell proliferation and migration in the subventricular zone (SVZ) remains unclear. Our results provide a detailed characterisation of FABP3, 5, and 7 expression in adult and fetal human and sheep SVZ. High FABP5 expression was specifically observed in the adult human SVZ and co-labelled with polysialylated neural cell adhesion molecule (PSA-NCAM), glial fibrillary acidic protein (GFAP), GFAPδ, and proliferating cell nuclear antigen (PCNA), indicating a role for FABP5 throughout the full maturation process of astrocytes and neuroblasts. Some FABP5+ cells had a radial glial-like appearance and co-labelled with the radial glia markers vimentin (40E-C) and GFAP. In the fetal human brain, FABP5 was expressed by radial glia cells throughout the ventricular zone. In contrast, radial glia-like cells in sheep highly expressed FABP3. Taken together, these differences highlight the species-specific expression profile of FABPs in the SVZ. In this study, we demonstrate the distribution of FABP in the adult human SVZ and fetal ventricular zone and reveal its expression on persistent radial glia that may be involved in adult neurogenesis., (© 2018 S. Karger AG, Basel.)
- Published
- 2018
- Full Text
- View/download PDF
50. Brain urea increase is an early Huntington's disease pathogenic event observed in a prodromal transgenic sheep model and HD cases.
- Author
-
Handley RR, Reid SJ, Brauning R, Maclean P, Mears ER, Fourie I, Patassini S, Cooper GJS, Rudiger SR, McLaughlan CJ, Verma PJ, Gusella JF, MacDonald ME, Waldvogel HJ, Bawden CS, Faull RLM, and Snell RG
- Subjects
- Adult, Animals, Animals, Genetically Modified, Corpus Striatum pathology, Disease Models, Animal, Female, Humans, Huntingtin Protein genetics, Huntingtin Protein metabolism, Huntington Disease genetics, Huntington Disease pathology, Male, Sheep, Trinucleotide Repeat Expansion genetics, Corpus Striatum metabolism, Huntington Disease metabolism, Urea metabolism
- Abstract
The neurodegenerative disorder Huntington's disease (HD) is typically characterized by extensive loss of striatal neurons and the midlife onset of debilitating and progressive chorea, dementia, and psychological disturbance. HD is caused by a CAG repeat expansion in the Huntingtin ( HTT ) gene, translating to an elongated glutamine tract in the huntingtin protein. The pathogenic mechanism resulting in cell dysfunction and death beyond the causative mutation is not well defined. To further delineate the early molecular events in HD, we performed RNA-sequencing (RNA-seq) on striatal tissue from a cohort of 5-y-old OVT73 -line sheep expressing a human CAG-expansion HTT cDNA transgene. Our HD OVT73 sheep are a prodromal model and exhibit minimal pathology and no detectable neuronal loss. We identified significantly increased levels of the urea transporter SLC14A1 in the OVT73 striatum, along with other important osmotic regulators. Further investigation revealed elevated levels of the metabolite urea in the OVT73 striatum and cerebellum, consistent with our recently published observation of increased urea in postmortem human brain from HD cases. Extending that finding, we demonstrate that postmortem human brain urea levels are elevated in a larger cohort of HD cases, including those with low-level neuropathology (Vonsattel grade 0/1). This elevation indicates increased protein catabolism, possibly as an alternate energy source given the generalized metabolic defect in HD. Increased urea and ammonia levels due to dysregulation of the urea cycle are known to cause neurologic impairment. Taken together, our findings indicate that aberrant urea metabolism could be the primary biochemical disruption initiating neuropathogenesis in HD., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.