Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 20(1): 276, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-37996924

ABSTRACT

Chronic activation and dysfunction of microglia have been implicated in the pathogenesis and progression of many neurodegenerative disorders, including Huntington's disease (HD). HD is a genetic condition caused by a mutation that affects the folding and function of huntingtin (HTT). Signs of microglia activation have been observed in HD patients even before the onset of symptoms. It is unclear, however, whether pro-inflammatory microglia activation in HD results from cell-autonomous expression of mutant HTT, is the response of microglia to a diseased brain environment, or both. In this study, we used primary microglia isolated from HD knock-in (Q140) and wild-type (Q7) mice to investigate their response to inflammatory conditions in vitro in the absence of confounding effects arising from brain pathology. We show that naïve Q140 microglia do not undergo spontaneous pro-inflammatory activation and respond to inflammatory triggers, including stimulation of TLR4 and TLR2 and exposure to necrotic cells, with similar kinetics of pro-inflammatory gene expression as wild-type microglia. Upon termination of the inflammatory insult, the transcription of pro-inflammatory cytokines is tapered off in Q140 and wild-type microglia with similar kinetics. However, the ability of Q140 microglia to develop tolerance in response to repeated inflammatory stimulations is partially impaired in vitro and in vivo, potentially contributing to the establishment of chronic neuroinflammation in HD. We further show that ganglioside GM1, a glycosphingolipid with anti-inflammatory effects on wild-type microglia, not only decreases the production of pro-inflammatory cytokines and nitric oxide in activated Q140 microglia, but also dramatically dampen microglia response to re-stimulation with LPS in an experimental model of tolerance. These effects are independent from the expression of interleukin 1 receptor associated kinase 3 (Irak-3), a strong modulator of LPS signaling involved in the development of innate immune tolerance and previously shown to be upregulated by immune cell treatment with gangliosides. Altogether, our data suggest that external triggers are required for HD microglia activation, but a cell-autonomous dysfunction that affects the ability of HD microglia to acquire tolerance might contribute to the establishment of neuroinflammation in HD. Administration of GM1 might be beneficial to attenuate chronic microglia activation and neuroinflammation.


Subject(s)
G(M1) Ganglioside , Huntington Disease , Humans , Mice , Animals , Huntington Disease/metabolism , Microglia/metabolism , Neuroinflammatory Diseases , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , Cytokines/metabolism , Disease Models, Animal
2.
Nat Commun ; 14(1): 2327, 2023 04 22.
Article in English | MEDLINE | ID: mdl-37087495

ABSTRACT

Immunomodulatory Siglecs are controlled by their glycoprotein and glycolipid ligands. Siglec-glycolipid interactions are often studied outside the context of a lipid bilayer, missing the complex behaviors of glycolipids in a membrane. Through optimizing a liposomal formulation to dissect Siglec-glycolipid interactions, it is shown that Siglec-6 can recognize glycolipids independent of its canonical binding pocket, suggesting that Siglec-6 possesses a secondary binding pocket tailored for recognizing glycolipids in a bilayer. A panel of synthetic neoglycolipids is used to probe the specificity of this glycolipid binding pocket on Siglec-6, leading to the development of a neoglycolipid with higher avidity for Siglec-6 compared to natural glycolipids. This neoglycolipid facilitates the delivery of liposomes to Siglec-6 on human mast cells, memory B-cells and placental syncytiotrophoblasts. A physiological relevance for glycolipid recognition by Siglec-6 is revealed for the binding and internalization of extracellular vesicles. These results demonstrate a unique and physiologically relevant ability of Siglec-6 to recognize glycolipids in a membrane.


Subject(s)
Extracellular Vesicles , Sialic Acid Binding Immunoglobulin-like Lectins , Female , Humans , Pregnancy , Extracellular Vesicles/metabolism , Glycolipids/chemistry , Glycolipids/metabolism , Liposomes , Mast Cells/metabolism , Memory B Cells/metabolism , Placenta/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
3.
Stem Cell Reports ; 18(2): 519-533, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36608690

ABSTRACT

Demyelinating disorders of the central nervous system (CNS) occur when myelin and oligodendrocytes are damaged or lost. Remyelination and regeneration of oligodendrocytes can be achieved from endogenous oligodendrocyte precursor cells (OPCs) that reside in the adult CNS tissue. Using a cuprizone mouse model of demyelination, we show that infusion of fractalkine (CX3CL1) into the demyelinated murine brain increases de novo oligodendrocyte formation and enhances remyelination in the corpus callosum and cortical gray matter. This is achieved by increased OPC proliferation in the cortical gray matter as well as OPC differentiation and attenuation of microglia/macrophage activation both in corpus callosum and cortical gray matter. Finally, we show that activated OPCs and microglia/macrophages express fractalkine receptor CX3CR1 in vivo, and that in OPC-microglia co-cultures fractalkine increases in vitro oligodendrocyte differentiation by modulating both OPC and microglia biology. Our results demonstrate a novel pro-regenerative role of fractalkine in a demyelinating mouse model.


Subject(s)
Demyelinating Diseases , Remyelination , Mice , Animals , Chemokine CX3CL1 , Oligodendroglia/physiology , Myelin Sheath , Disease Models, Animal , Cell Differentiation/physiology , Mice, Inbred C57BL
4.
Cells ; 11(3)2022 01 20.
Article in English | MEDLINE | ID: mdl-35159161

ABSTRACT

Extracellular vesicles (EVs) are involved in a multitude of physiological functions and play important roles in health and disease. The largest proportion of studies on EVs is based on the analysis and characterization of EVs secreted in the cell culture medium. These studies remain challenging due to the small size of the EV particles, a lack of universal EV markers, and sample loss or technical artifacts that are often associated with EV labeling for single particle tracking and/or separation techniques. To address these problems, we characterized and validated a method for in-cell EV labeling with fluorescent lipids coupled with direct analysis of lipid-labeled EVs in the conditioned medium by imaging flow cytometry (IFC). This approach significantly reduces sample processing and loss compared to established methods for EV separation and labeling in vitro, resulting in improved detection of quantitative changes in EV secretion and subpopulations compared to protocols that rely on EV separation by size-exclusion chromatography and ultracentrifugation. Our optimized protocol for in-cell EV labeling and analysis of the conditioned medium reduces EV sample processing and loss, and is well-suited for cell biology studies that focus on modulation of EV secretion by cells in culture.


Subject(s)
Extracellular Vesicles , Culture Media, Conditioned/analysis , Culture Media, Conditioned/pharmacology , Extracellular Vesicles/chemistry , Flow Cytometry/methods , Specimen Handling , Ultracentrifugation/methods
5.
J Neuroinflammation ; 19(1): 9, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34991625

ABSTRACT

BACKGROUND: Gangliosides are glycosphingolipids highly enriched in the brain, with important roles in cell signaling, cell-to-cell communication, and immunomodulation. Genetic defects in the ganglioside biosynthetic pathway result in severe neurodegenerative diseases, while a partial decrease in the levels of specific gangliosides was reported in Parkinson's disease and Huntington's disease. In models of both diseases and other conditions, administration of GM1-one of the most abundant gangliosides in the brain-provides neuroprotection. Most studies have focused on the direct neuroprotective effects of gangliosides on neurons, but their role in other brain cells, in particular microglia, is not known. In this study we investigated the effects of exogenous ganglioside administration and modulation of endogenous ganglioside levels on the response of microglia to inflammatory stimuli, which often contributes to initiation or exacerbation of neurodegeneration. METHODS: In vitro studies were performed using BV2 cells, mouse, rat, and human primary microglia cultures. Modulation of microglial ganglioside levels was achieved by administration of exogenous gangliosides, or by treatment with GENZ-123346 and L-t-PDMP, an inhibitor and an activator of glycolipid biosynthesis, respectively. Response of microglia to inflammatory stimuli (LPS, IL-1ß, phagocytosis of latex beads) was measured by analysis of gene expression and/or secretion of pro-inflammatory cytokines. The effects of GM1 administration on microglia activation were also assessed in vivo in C57Bl/6 mice, following intraperitoneal injection of LPS. RESULTS: GM1 decreased inflammatory microglia responses in vitro and in vivo, even when administered after microglia activation. These anti-inflammatory effects depended on the presence of the sialic acid residue in the GM1 glycan headgroup and the presence of a lipid tail. Other gangliosides shared similar anti-inflammatory effects in in vitro models, including GD3, GD1a, GD1b, and GT1b. Conversely, GM3 and GQ1b displayed pro-inflammatory activity. The anti-inflammatory effects of GM1 and other gangliosides were partially reproduced by increasing endogenous ganglioside levels with L-t-PDMP, whereas inhibition of glycolipid biosynthesis exacerbated microglial activation in response to LPS stimulation. CONCLUSIONS: Our data suggest that gangliosides are important modulators of microglia inflammatory responses and reveal that administration of GM1 and other complex gangliosides exerts anti-inflammatory effects on microglia that could be exploited therapeutically.


Subject(s)
Anti-Inflammatory Agents/pharmacology , G(M1) Ganglioside/pharmacology , Inflammation/pathology , Microglia/drug effects , Animals , Cells, Cultured , Dioxanes/pharmacology , Humans , Inflammation/metabolism , Interleukin-1beta/pharmacology , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , Microglia/pathology , Phagocytosis/drug effects , Pyrrolidines/pharmacology , Rats
6.
Stem Cell Reports ; 16(8): 1968-1984, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34270934

ABSTRACT

Neural and oligodendrocyte precursor cells (NPCs and OPCs) in the subventricular zone (SVZ) of the brain contribute to oligodendrogenesis throughout life, in part due to direct regulation by chemokines. The role of the chemokine fractalkine is well established in microglia; however, the effect of fractalkine on SVZ precursor cells is unknown. We show that murine SVZ NPCs and OPCs express the fractalkine receptor (CX3CR1) and bind fractalkine. Exogenous fractalkine directly enhances OPC and oligodendrocyte genesis from SVZ NPCs in vitro. Infusion of fractalkine into the lateral ventricle of adult NPC lineage-tracing mice leads to increased newborn OPC and oligodendrocyte formation in vivo. We also show that OPCs secrete fractalkine and that inhibition of endogenous fractalkine signaling reduces oligodendrocyte formation in vitro. Finally, we show that fractalkine signaling regulates oligodendrogenesis in cerebellar slices ex vivo. In summary, we demonstrate a novel role for fractalkine signaling in regulating oligodendrocyte genesis from postnatal CNS precursor cells.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/metabolism , Lateral Ventricles/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Signal Transduction , Animals , CX3C Chemokine Receptor 1/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Chemokine CX3CL1/pharmacology , Gene Expression/drug effects , Lateral Ventricles/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Transcription Factor 2/genetics , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/cytology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
7.
Front Neurosci ; 14: 572965, 2020.
Article in English | MEDLINE | ID: mdl-33117120

ABSTRACT

Gangliosides are glycosphingolipids highly abundant in the nervous system, and carry most of the sialic acid residues in the brain. Gangliosides are enriched in cell membrane microdomains ("lipid rafts") and play important roles in the modulation of membrane proteins and ion channels, in cell signaling and in the communication among cells. The importance of gangliosides in the brain is highlighted by the fact that loss of function mutations in ganglioside biosynthetic enzymes result in severe neurodegenerative disorders, often characterized by very early or childhood onset. In addition, changes in the ganglioside profile (i.e., in the relative abundance of specific gangliosides) were reported in healthy aging and in common neurological conditions, including Huntington's disease (HD), Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), stroke, multiple sclerosis and epilepsy. At least in HD, PD and in some forms of epilepsy, experimental evidence strongly suggests a potential role of gangliosides in disease pathogenesis and potential treatment. In this review, we will summarize ganglioside functions that are crucial to maintain brain health, we will review changes in ganglioside levels that occur in major neurological conditions and we will discuss their contribution to cellular dysfunctions and disease pathogenesis. Finally, we will review evidence of the beneficial roles exerted by gangliosides, GM1 in particular, in disease models and in clinical trials.

9.
Hum Mol Genet ; 29(3): 418-431, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31875875

ABSTRACT

Brain cholesterol homeostasis is altered in Huntington's disease (HD), a neurodegenerative disorder caused by the expansion of a CAG nucleotide repeat in the HTT gene. Genes involved in the synthesis of cholesterol and fatty acids were shown to be downregulated shortly after the expression of mutant huntingtin (mHTT) in inducible HD cells. Nuclear levels of the transcription factors that regulate lipid biogenesis, the sterol regulatory element-binding proteins (SREBP1 and SREBP2), were found to be decreased in HD models compared to wild-type, but the underlying causes were not known. SREBPs are synthesized as inactive endoplasmic reticulum-localized precursors. Their mature forms (mSREBPs) are generated upon transport of the SREBP precursors to the Golgi and proteolytic cleavage, and are rapidly imported into the nucleus by binding to importin ß. We show that, although SREBP2 processing into mSREBP2 is not affected in YAC128 HD mice, mSREBP2 is mislocalized to the cytoplasm. Chimeric mSREBP2-and mSREBP1-EGFP proteins are also mislocalized to the cytoplasm in immortalized striatal cells expressing mHTT, in YAC128 neurons and in fibroblasts from HD patients. We further show that mHTT binds to the SREBP2/importin ß complex required for nuclear import and sequesters it in the cytoplasm. As a result, HD cells fail to upregulate cholesterogenic genes under sterol-depleted conditions. These findings provide mechanistic insight into the downregulation of genes involved in the synthesis of cholesterol and fatty acids in HD models, and have potential implications for other pathways modulated by SREBPs, including autophagy and excitotoxicity.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus/pathology , Cholesterol/metabolism , Green Fluorescent Proteins/metabolism , Huntingtin Protein/metabolism , Mutant Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Cell Nucleus/metabolism , Green Fluorescent Proteins/genetics , Homeostasis , Humans , Huntingtin Protein/genetics , Mice , Mutant Proteins/genetics , Mutation , Neurons/metabolism , Neurons/pathology , Sterol Regulatory Element Binding Protein 1/genetics
10.
Commun Biol ; 2: 450, 2019.
Article in English | MEDLINE | ID: mdl-31815204

ABSTRACT

CD33 is an immunomodulatory receptor linked to Alzheimer's disease (AD) susceptibility via regulation of phagocytosis in microglia. Divergent features between human CD33 (hCD33) and murine CD33 (mCD33) include a unique transmembrane lysine in mCD33 and cytoplasmic tyrosine in hCD33. The functional consequences of these differences in restraining phagocytosis remains poorly understood. Using a new αmCD33 monoclonal antibody, we show that mCD33 is expressed at high levels on neutrophils and low levels on microglia. Notably, cell surface expression of mCD33 is entirely dependent on Dap12 due to an interaction with the transmembrane lysine in mCD33. In RAW264.7 cultured macrophages, BV-2 cultured microglia, primary neonatal and adult microglia, uptake of cargo - including aggregated Aß1-42 - is not altered upon genetic ablation of mCD33. Alternatively, deletion of hCD33 in monocytic cell lines increased cargo uptake. Moreover, transgenic mice expressing hCD33 in the microglial cell lineage showed repressed cargo uptake in primary microglia. Therefore, mCD33 and hCD33 have divergent roles in regulating phagocytosis, highlighting the importance of studying hCD33 in AD susceptibility.

11.
Front Neurol ; 10: 859, 2019.
Article in English | MEDLINE | ID: mdl-31447771

ABSTRACT

Gangliosides are cell membrane components, most abundantly in the central nervous system (CNS) where they exert among others neuro-protective and -restorative functions. Clinical development of ganglioside replacement therapy for several neurodegenerative diseases was impeded by the BSE crisis in Europe during the 1990s. Nowadays, gangliosides are produced bovine-free and new pre-clinical and clinical data justify a reevaluation of their therapeutic potential in neurodegenerative diseases. Clinical experience is greatest with monosialo-tetrahexosyl-ganglioside (GM1) in the treatment of stroke. Fourteen randomized controlled trials (RCTs) in overall >2,000 patients revealed no difference in survival, but consistently superior neurological outcomes vs. placebo. GM1 was shown to attenuate ischemic neuronal injuries in diabetes patients by suppression of ERK1/2 phosphorylation and reduction of stress to the endoplasmic reticulum. There is level-I evidence from 5 RCTs of a significantly faster recovery with GM1 vs. placebo in patients with acute and chronic spinal cord injury (SCI), disturbance of consciousness after subarachnoid hemorrhage, or craniocerebral injuries due to closed head trauma. In Parkinson's disease (PD), two RCTs provided evidence of GM1 to be superior to placebo in improving motor symptoms and long-term to result in a slower than expected symptom progression, suggesting disease-modifying potential. In Alzheimer's disease (AD), the role of gangliosides has been controversial, with some studies suggesting a "seeding" role for GM1 in amyloid ß polymerization into toxic forms, and others more recently suggesting a rather protective role in vivo. In Huntington's disease (HD), no clinical trials have been conducted yet. However, low GM1 levels observed in HD cells were shown to increase cell susceptibility to apoptosis. Accordingly, treatment with GM1 increased survival of HD cells in vitro and consistently ameliorated pathological phenotypes in several murine HD models, with effects seen at molecular, cellular, and behavioral level. Given that in none of the clinical trials using GM1 any clinically relevant safety issues have occurred to date, current data supports expanding GM1 clinical research, particularly to conditions with high, unmet medical need.

12.
FASEB J ; 33(4): 5045-5057, 2019 04.
Article in English | MEDLINE | ID: mdl-30615497

ABSTRACT

Phosphatidylethanolamine N-methyltransferase (PEMT) is an important enzyme in hepatic phosphatidylcholine (PC) biosynthesis. Pemt-/- mice fed a high-fat diet are protected from obesity and whole-body insulin resistance. However, Pemt-/- mice develop severe nonalcoholic steatohepatitis (NASH). Because NASH is often associated with hepatic insulin resistance, we investigated whether the increased insulin sensitivity in Pemt-/- mice was restricted to nonhepatic tissues or whether the liver was also insulin sensitive. Strikingly, the livers of Pemt-/- mice compared with those of Pemt+/+ mice were not insulin resistant, despite elevated levels of hepatic triacylglycerols and diacylglycerols, as well as increased hepatic inflammation and fibrosis. Endogenous glucose production was lower in Pemt-/- mice under both basal and hyperinsulinemic conditions. Experiments in primary hepatocytes and hepatoma cells revealed improved insulin signaling in the absence of PEMT, which was not due to changes in diacylglycerols, ceramides, or gangliosides. On the other hand, the phospholipid composition in hepatocytes seems critically important for insulin signaling such that lowering the PC:phosphatidylethanolamine (PE) ratio improves insulin signaling. Thus, treatments to reduce the PC:PE ratio in liver may protect against the development of hepatic insulin resistance.-Van der Veen, J. N., Lingrell, S., McCloskey, N., LeBlond, N. D., Galleguillos, D., Zhao, Y. Y., Curtis, J. M., Sipione, S., Fullerton, M. D., Vance, D. E., Jacobs, R. L. A role for phosphatidylcholine and phosphatidylethanolamine in hepatic insulin signaling.


Subject(s)
Insulin/metabolism , Liver/metabolism , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Animals , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Phosphatidylethanolamine N-Methyltransferase/metabolism , Signal Transduction/physiology
13.
Nat Commun ; 9(1): 4417, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356053

ABSTRACT

Kv1.2 is a prominent voltage-gated potassium channel that influences action potential generation and propagation in the central nervous system. We explored multi-protein complexes containing Kv1.2 using mass spectrometry followed by screening for effects on Kv1.2. We report that Slc7a5, a neutral amino acid transporter, has a profound impact on Kv1.2. Co-expression with Slc7a5 reduces total Kv1.2 protein, and dramatically hyperpolarizes the voltage-dependence of activation by -47 mV. These effects are attenuated by expression of Slc3a2, a known binding partner of Slc7a5. The profound Slc7a5-mediated current suppression is partly explained by a combination of gating effects including accelerated inactivation and a hyperpolarizing shift of channel activation, causing channels to accumulate in a non-conducting state. Two recently reported Slc7a5 mutations linked to neurodevelopmental delay exhibit a localization defect and have attenuated effects on Kv1.2. In addition, epilepsy-linked gain-of-function Kv1.2 mutants exhibit enhanced sensitivity to Slc7a5.


Subject(s)
Epilepsy/metabolism , Kv1.2 Potassium Channel/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Animals , Blotting, Western , Electrophysiology , Epilepsy/genetics , Flow Cytometry , HEK293 Cells , Humans , Immunoprecipitation , Kv1.2 Potassium Channel/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Mass Spectrometry , Mice , Rats, Sprague-Dawley
14.
Proc Natl Acad Sci U S A ; 115(30): E7081-E7090, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29987005

ABSTRACT

The huntingtin N17 domain is a modulator of mutant huntingtin toxicity and is hypophosphorylated in Huntington's disease (HD). We conducted high-content analysis to find compounds that could restore N17 phosphorylation. One lead compound from this screen was N6-furfuryladenine (N6FFA). N6FFA was protective in HD model neurons, and N6FFA treatment of an HD mouse model corrects HD phenotypes and eliminates cortical mutant huntingtin inclusions. We show that N6FFA restores N17 phosphorylation levels by being salvaged to a triphosphate form by adenine phosphoribosyltransferase (APRT) and used as a phosphate donor by casein kinase 2 (CK2). N6FFA is a naturally occurring product of oxidative DNA damage. Phosphorylated huntingtin functionally redistributes and colocalizes with CK2, APRT, and N6FFA DNA adducts at sites of induced DNA damage. We present a model in which this natural product compound is salvaged to provide a triphosphate substrate to signal huntingtin phosphorylation via CK2 during low-ATP stress under conditions of DNA damage, with protective effects in HD model systems.


Subject(s)
Adenine , DNA Adducts/metabolism , DNA Damage , Huntington Disease/drug therapy , Neurons/metabolism , Signal Transduction/drug effects , Adenine/analogs & derivatives , Adenine/pharmacokinetics , Adenine/pharmacology , Adenine Phosphoribosyltransferase/genetics , Adenine Phosphoribosyltransferase/metabolism , Animals , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line, Transformed , DNA Adducts/genetics , Disease Models, Animal , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Transgenic , Neurons/pathology , Phosphorylation/drug effects , Phosphorylation/genetics , Signal Transduction/genetics
15.
EMBO Mol Med ; 9(11): 1537-1557, 2017 11.
Article in English | MEDLINE | ID: mdl-28993428

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disorder characterized by motor, cognitive and psychiatric problems. Previous studies indicated that levels of brain gangliosides are lower than normal in HD models and that administration of exogenous ganglioside GM1 corrects motor dysfunction in the YAC128 mouse model of HD In this study, we provide evidence that intraventricular administration of GM1 has profound disease-modifying effects across HD mouse models with different genetic background. GM1 administration results in decreased levels of mutant huntingtin, the protein that causes HD, and in a wide array of beneficial effects that include changes in levels of DARPP32, ferritin, Iba1 and GFAP, modulation of dopamine and serotonin metabolism, and restoration of normal levels of glutamate, GABA, L-Ser and D-Ser. Treatment with GM1 slows down neurodegeneration, white matter atrophy and body weight loss in R6/2 mice. Motor functions are significantly improved in R6/2 mice and restored to normal in Q140 mice, including gait abnormalities that are often resistant to treatments. Psychiatric-like and cognitive dysfunctions are also ameliorated by GM1 administration in Q140 and YAC128 mice. The widespread benefits of GM1 administration, at molecular, cellular and behavioural levels, indicate that this ganglioside has strong therapeutic and disease-modifying potential in HD.


Subject(s)
G(M1) Ganglioside/therapeutic use , Huntington Disease/drug therapy , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Ferritins/metabolism , G(M1) Ganglioside/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Huntingtin Protein/metabolism , Huntington Disease/mortality , Huntington Disease/pathology , Infusions, Intraventricular , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Serotonin/metabolism , Survival Rate , gamma-Aminobutyric Acid/metabolism
16.
Anal Chem ; 89(17): 9330-9338, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28768095

ABSTRACT

This work describes a versatile analytical approach, which combines the proxy ligand electrospray ionization mass spectrometry (ESI-MS) assay and model membranes of defined composition, to quantify the influence of lipid bilayer composition on protein-glycolipid binding in vitro. To illustrate the implementation of the assay (experimental design and data analysis), affinities of the monosialoganglioside ligand GM1, incorporated into nanodiscs (NDs), for cholera toxin B subunit homopentamer (CTB5) were measured. A series of NDs containing GM1 and cholesterol were prepared using three different phospholipids (1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC)), and the average GM1 and cholesterol content of each ND were determined. The intrinsic affinities of GM1-containing NDs prepared with the three phospholipids are found to be similar in magnitude, indicating that small differences in the fatty acid chain length and the number of unsaturated bonds do not significantly affect the CTB5-GM1 interaction. Moreover, the measured affinities are similar to the value measured for GM1 pentasaccharide, indicating that neither the ceramide moiety nor the surface of the phospholipid membrane plays a significant role in CTB5 binding. The intrinsic (per binding site) affinity of the CTB5-GM1 interaction was found to decrease with increasing GM1 content of the ND, consistent with the occurrence of GM1 clustering in the membrane, which sterically hinders binding to CTB5. Notably, the addition of cholesterol to GM1-containing NDs did not have a significant effect on the strength of the CTB5-GM1 interaction. This result, which is at odds with the findings of a previous study of CTB5 binding to GM1 in vesicles, suggests that cholesterol does not "mask" GM1, at least not in NDs. These data, in addition to providing new insights into the influence of membrane composition on CTB5-GM1 binding, demonstrate the potential of the proxy ligand ESI-MS approach for comprehensive and quantitative studies of lectin interactions with glycolipids in native-like, membrane environments.


Subject(s)
Glycolipids/chemistry , Membranes, Artificial , Nanostructures/chemistry , Proteins/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Ligands , Models, Molecular , Protein Conformation
17.
Exp Neurol ; 283(Pt A): 365-74, 2016 09.
Article in English | MEDLINE | ID: mdl-27401133

ABSTRACT

Elevated levels of the second messenger molecule cyclic adenosine monophosphate (cAMP) are often associated with neuron sprouting and neurite extension (i.e., neuroplasticity). Phosphokinase A (PKA) is a prominent downstream target of cAMP that has been associated with neurite outgrowth. We hypothesized that rehabilitative motor training following spinal cord injuries promotes neuroplasticity via PKA activation. However, in two independent experiments, inhibition of cortical PKA using Rp-cAMPS throughout rehabilitative training robustly increased functional recovery and collateral sprouting of injured corticospinal tract axons, an indicator of neuroplasticity. Consistent with these in vivo findings, using cultured STHdh neurons, we found that Rp-cAMPS had no effect on the phosphorylation of CREB (cAMP response element-binding protein), a prominent downstream target of PKA, even with the concomitant application of the adenylate cyclase agonist forskolin to increase cAMP levels. Conversely, when cAMP levels were increased using the phosphodiesterase inhibitor IBMX, Rp-cAMPS potently inhibited CREB phosphorylation. Taken together, our results suggest that an alternate cAMP dependent pathway was involved in increasing CREB phosphorylation and neuroplasticity. This idea was supported by an in vitro neurite outgrowth assay, where inhibiting PKA did enhance neurite outgrowth. However, when PKA inhibition was combined with inhibition of EPAC2 (exchange protein directly activated by cAMP), another downstream target of cAMP in neurons, neurite outgrowth was significantly reduced. In conclusion, blocking PKA in cortical neurons of spinal cord injured rats increases neurite outgrowth of the lesioned corticospinal tract fibres and the efficacy of rehabilitative training, likely via EPAC.


Subject(s)
Cerebral Cortex/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/rehabilitation , 1-Methyl-3-isobutylxanthine/pharmacology , Analysis of Variance , Animals , CREB-Binding Protein/metabolism , Cell Line, Transformed/metabolism , Cell Line, Transformed/pathology , Cells, Cultured , Cerebral Cortex/metabolism , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Microglia/metabolism , Microglia/pathology , Neurites/drug effects , Neurites/physiology , Neurons/drug effects , Neurons/metabolism , Phosphodiesterase Inhibitors/pharmacology , Pyramidal Tracts/metabolism , Rats , Rats, Inbred Lew , Recovery of Function/physiology , Thionucleotides/metabolism
18.
Lipids ; 51(5): 561-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26846325

ABSTRACT

Huntington's Disease (HD) is an autosomal dominant disease that occurs as a result of expansion of the trinucleotide repeat CAG (glutamine) on the HTT gene. HD patients exhibit various forms of mitochondrial dysfunction within neurons and peripheral tissues. Cardiolipin (Ptd2Gro) is a polyglycerophospholipid found exclusively in mitochondria and is important for maintaining mitochondrial function. We examined if altered Ptd2Gro metabolism was involved in the mitochondrial dysfunction associated with HD. Mitochondrial basal respiration, spare respiratory capacity, ATP coupling efficiency and rate of glycolysis were markedly diminished in Epstein-Barr virus transformed HD lymphoblasts compared to controls (CTRL). Mitochondrial supercomplex formation and Complex I activity within these supercomplexes did not vary between HD patients with different length of CAG repeats and appeared unaltered compared to CTRL. In contrast, in vitro Complex I enzyme activity in mitochondrial enriched samples was reduced in HD lymphoblasts compared to CTRL. The total cellular pool size of Ptd2Gro and its synthesis/remodeling from [(3)H]acetate/[(14)C]oleate were unaltered in HD lymphoblasts compared to CTRL. In addition, the molecular species of Ptd2Gro were essentially unaltered in HD lymphoblasts compared to CTRL. We conclude that compared to CTRL lymphoblasts, HD lymphoblasts display impaired mitochondrial basal respiration, spare respiratory capacity, ATP coupling efficiency and rate of glycolysis with any pathological CAG repeat length, but this is not due to alterations in Ptd2Gro metabolism. We suggest that HD patient lymphoblasts may be a useful model to study defective energy metabolism that does not involve alterations in Ptd2Gro metabolism.


Subject(s)
Cardiolipins/metabolism , Huntington Disease/metabolism , Lymphocytes/pathology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Adenosine Triphosphate/metabolism , Epstein-Barr Virus Infections/complications , Glycolysis , Herpesvirus 4, Human/isolation & purification , Humans , Huntington Disease/pathology , Huntington Disease/virology , Lymphocytes/metabolism , Mitochondria/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...