Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 82
1.
Hum Mol Genet ; 32(8): 1361-1379, 2023 04 06.
Article En | MEDLINE | ID: mdl-36519759

Infantile Krabbe disease is a rapidly progressive and fatal disorder of myelin, caused by inherited deficiency of the lysosomal enzyme ß-galactocerebrosidase. Affected children lose their motor skills and other faculties; uncontrolled seizures are a frequent terminal event. Overexpression of the sphingolipid metabolite psychosine is a pathogenic factor, but does not fully account for the pleiotropic manifestations and there is a clear need to investigate additional pathological mechanisms. We examined innate immunity, caspase-11 and associated inflammatory pathways in twitcher mice, an authentic model of Krabbe disease. Combined use of molecular tools, RNAscope in situ hybridization and immunohistochemical staining established that the expression of pro-inflammatory non-canonical caspase-11, canonical caspase-1, gasdermin D and cognate genes is induced in nervous tissue. Early onset and progressive upregulation of these genes accompany demyelination and gliosis and although the molecules are scant in healthy tissue, abundance of the respective translation products is greatly increased in diseased animals. Caspase-11 is found in reactive microglia/macrophages as well as astrocytes but caspase-1 and gasdermin D are restricted to reactive microglia/macrophages. The inflammasome signature is not unique to Krabbe disease; to varying degrees, this signature is also prominent in other lysosomal diseases, Sandhoff and Niemann-Pick Type-C1, and the lysolecithin toxin model of focal demyelination. Given the potent inflammatory response here identified in Krabbe disease and the other neurodegenerative disorders studied, a broad induction of inflammasomes is likely to be a dominant factor in the pathogenesis, and thus represents a platform for therapeutic exploration.


Leukodystrophy, Globoid Cell , Mice , Animals , Leukodystrophy, Globoid Cell/genetics , Inflammasomes/metabolism , Up-Regulation , Gasdermins , Disease Models, Animal , Psychosine/metabolism , Psychosine/pharmacology , Caspases/metabolism
2.
ASN Neuro ; 14: 17590914221087817, 2022.
Article En | MEDLINE | ID: mdl-35300522

Psychosine exerts most of its toxic effects by altering membrane dynamics with increased shedding of extracellular vesicles (EVs). In this study, we discovered that a fraction of psychosine produced in the brain of the Twitcher mouse, a model for Krabbe disease, is associated with secreted EVs. We evaluated the effects of attenuating EV secretion in the Twitcher brain by depleting ceramide production with an inhibitor of neutral sphingomyelinase 2, GW4869. Twitcher mice treated with GW4869 had decreased overall EV levels, reduced EV-associated psychosine and unexpectedly, correlated with increased disease severity. Notably, characterization of well-established, neuroanatomic hallmarks of disease pathology, such as demyelination and inflammatory gliosis, remained essentially unaltered in the brains of GW4869-treated Twitcher mice compared to vehicle-treated Twitcher controls. Further analysis of Twitcher brain pathophysiology is required to understand the mechanism behind early-onset disease severity in GW4869-treated mice. The results herein demonstrate that some pathogenic lipids like psychosine may be secreted using EV pathways. Our results highlight the relevance of this secretory mechanism as a possible contributor to spreading pathogenic lipids in neurological lipidoses.


Extracellular Vesicles , Leukodystrophy, Globoid Cell , Animals , Brain/metabolism , Disease Models, Animal , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Leukodystrophy, Globoid Cell/metabolism , Leukodystrophy, Globoid Cell/pathology , Mice , Psychosine/analysis , Psychosine/metabolism , Psychosine/pharmacology , Sphingolipids/metabolism
3.
Acta Pharmacol Sin ; 43(3): 552-562, 2022 Mar.
Article En | MEDLINE | ID: mdl-33935286

We previously show that fatty acid-binding protein 3 (FABP3) triggers α-synuclein (Syn) accumulation and induces dopamine neuronal cell death in Parkinson disease mouse model. But the role of fatty acid-binding protein 7 (FABP7) in the brain remains unclear. In this study we investigated whether FABP7 was involved in synucleinopathies. We showed that FABP7 was co-localized and formed a complex with Syn in Syn-transfected U251 human glioblastoma cells, and treatment with arachidonic acid (100 M) significantly promoted FABP7-induced Syn aggregation, which was associated with cell death. We demonstrated that synthetic FABP7 ligand 6 displayed a high affinity against FABP7 with Kd value of 209 nM assessed in 8-anilinonaphthalene-1-sulfonic acid (ANS) assay; ligand 6 improved U251 cell survival via disrupting the FABP7-Syn interaction. We showed that activation of phospholipase A2 (PLA2) by psychosine (10 M) triggered oligomerization of endogenous Syn and FABP7, and induced cell death in both KG-1C human oligodendroglia cells and oligodendrocyte precursor cells (OPCs). FABP7 ligand 6 (1 M) significantly decreased Syn oligomerization and aggregation thereby prevented KG-1C and OPC cell death. This study demonstrates that FABP7 triggers α-synuclein oligomerization through oxidative stress, while FABP7 ligand 6 can inhibit FABP7-induced Syn oligomerization and aggregation, thereby rescuing glial cells and oligodendrocytes from cell death.


Fatty Acid-Binding Protein 7/metabolism , Neuroglia/metabolism , Oligodendroglia/metabolism , Oxidative Stress/physiology , alpha-Synuclein/metabolism , Animals , Arachidonic Acid/pharmacology , Cell Death/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Oligodendrocyte Precursor Cells/drug effects , Phospholipases A2/drug effects , Protein Binding/physiology , Psychosine/pharmacology
4.
Br J Pharmacol ; 179(10): 2193-2207, 2022 05.
Article En | MEDLINE | ID: mdl-34766332

BACKGROUND AND PURPOSE: Glucosylsphingosine (GS), an endogenous sphingolipid, is highly accumulated in the epidermis of patients with atopic dermatitis (AD) due to abnormal ceramide metabolism. More importantly, GS can evoke scratching behaviours. However, the precise molecular mechanism by which GS induces pruritus has been elusive. Thus, the present study aimed to elucidate the molecular signalling pathway of GS, especially at the peripheral sensory neuronal levels. EXPERIMENTAL APPROACH: Calcium imaging was used to investigate the responses of HEK293T cells or mouse dorsal root ganglion (DRG) neurons to application of GS. Scratching behaviour tests were also performed with wild-type and Trpv4 knockout mice. KEY RESULTS: GS activated DRG neurons in a manner involving both the 5-HT2A receptor and TRPV4. Furthermore, GS-induced responses were significantly suppressed by various inhibitors, including ketanserin (5-HT2A receptor antagonist), YM254890 (Gαq/11 inhibitor), gallein (Gßγ complex inhibitor), U73122 (phospholipase C inhibitor), bisindolylmaleimide I (PKC inhibitor) and HC067047 (TRPV4 antagonist). Moreover, DRG neurons from Trpv4 knockout mice exhibited significantly reduced responses to GS. Additionally, GS-evoked scratching behaviours were greatly decreased by pretreatment with inhibitors of either 5-HT2A receptor or TRPV4. As expected, GS-evoked scratching behaviour was also significantly decreased in Trpv4 knockout mice. CONCLUSION AND IMPLICATIONS: Overall, the present study provides evidence for a novel molecular signalling pathway for GS-evoked pruritus, which utilizes both 5-HT2A receptor and TRPV4 in mouse sensory neurons. Considering the high accumulation of GS in the epidermis of patients with AD, GS could be another pruritogen in patients with AD.


Pruritus , Psychosine , Receptor, Serotonin, 5-HT2A , Sensory Receptor Cells , TRPV Cation Channels , Animals , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Mice , Pruritus/chemically induced , Pruritus/metabolism , Psychosine/analogs & derivatives , Psychosine/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Sensory Receptor Cells/drug effects , TRPV Cation Channels/metabolism
5.
Neurotherapeutics ; 18(4): 2608-2622, 2021 10.
Article En | MEDLINE | ID: mdl-34480290

Polymeric nanoparticles are being extensively investigated as an approach for brain delivery of drugs, especially for their controlled release and targeting capacity. Nose-to-brain administration of nanoparticles, bypassing the blood brain barrier, offers a promising strategy to deliver drugs to the central nervous system. Here, we investigated the potential of hybrid nanoparticles as a therapeutic approach for demyelinating diseases, more specifically for Krabbe's disease. This rare leukodystrophy is characterized by the lack of enzyme galactosylceramidase, leading to the accumulation of toxic psychosine in glial cells causing neuroinflammation, extensive demyelination and death. We present evidence that lecithin/chitosan nanoparticles prevent damage associated with psychosine by sequestering the neurotoxic sphingolipid via physicochemical hydrophobic interactions. We showed how nanoparticles prevented the cytotoxicity caused by psychosine in cultured human astrocytes in vitro, and how the nanoparticle size and PDI augmented while the electrostatic charges of the surface decreased, suggesting a direct interaction between psychosine and the nanoparticles. Moreover, we studied the effects of nanoparticles ex vivo using mouse cerebellar organotypic cultures, observing that nanoparticles prevented the demyelination and axonal damage caused by psychosine, as well as a moderate prevention of the astrocytic death. Taken together, these results suggest that lecithin-chitosan nanoparticles are a potential novel delivery system for drugs for certain demyelinating conditions such as Krabbe's disease, due to their dual effect: not only are they an efficient platform for drug delivery, but they exert a protective effect themselves in tampering the levels of psychosine accumulation.


Demyelinating Diseases , Leukodystrophy, Globoid Cell , Nanoparticles , Animals , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Leukodystrophy, Globoid Cell/drug therapy , Mice , Neuroinflammatory Diseases , Psychosine/pharmacology , Psychosine/therapeutic use
6.
Cells ; 9(5)2020 05 01.
Article En | MEDLINE | ID: mdl-32370022

Impairment of the ubiquitin-proteasome-system (UPS) and autophagy causing cytoplasmic aggregation of ubiquitin andp62 have been implicated in the pathogenesis of most neurodegenerative disorders, yet, they have not been fully elucidated in leukodystrophies. The relationship among impairment of UPS, autophagy, and globoid cell leukodystrophy (GLD), one of the most common demyelinating leukodystrophies, is clarified in this study. We examined the ubiquitin and autophagy markers in the brains of twitcher mice, a murine model of infantile GLD, and in human oligodendrocytes incubated with psychosine. Immunohistochemical examinations showed spatiotemporal accumulation of ubiquitin- and p62-aggregates mainly in the white matter of brain and spinal cord at disease progression. Western blot analysis demonstrated a significant accumulation of ubiquitin, p62, and LC3-II in insoluble fraction in parallel with progressive demyelination and neuroinflammation in twitcher brains. In vitro study validated a dose- and time-dependent cytotoxicity of psychosine upon autophagy and UPS machinery. Inhibition of autophagy and UPS exacerbated the accumulation of insoluble ubiquitin, p62, and LC3-II proteins mediated by psychosine cytotoxicity as well as increased cytoplasmic deposition of ubiquitin- and p62-aggregates, and accumulation of autophagosomes and autolysosomes. Further, the subsequent accumulation of reactive oxygen species and reduction of mitochondrial respiration led to cell death. Our studies validate the impairment of proteasome and autophagy underlying the pathogenesis of GLD. These findings provide a novel insight into pathogenesis of GLD and suggest a specific pathomechanism as an ideal target for therapeutic approaches.


Autophagy , Leukodystrophy, Globoid Cell/pathology , Proteasome Endopeptidase Complex/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Brain/metabolism , Brain/pathology , Cell Line , Cell Respiration/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Degeneration/pathology , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Protein Aggregates/drug effects , Psychosine/pharmacology , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Time Factors , Ubiquitin/metabolism , White Matter/pathology
7.
Dis Model Mech ; 12(5)2019 05 23.
Article En | MEDLINE | ID: mdl-31036560

Glycosphingolipid (GSL) accumulation is implicated in the neuropathology of several lysosomal conditions, such as Krabbe disease, and may also contribute to neuronal and glial dysfunction in adult-onset conditions such as Parkinson's disease, Alzheimer's disease and multiple sclerosis. GSLs accumulate in cellular membranes and disrupt their structure; however, how membrane disruption leads to cellular dysfunction remains unknown. Using authentic cellular and animal models for Krabbe disease, we provide a mechanism explaining the inactivation of lipid raft (LR)-associated IGF-1-PI3K-Akt-mTORC2, a pathway of crucial importance for neuronal function and survival. We show that psychosine, the GSL that accumulates in Krabbe disease, leads to a dose-dependent LR-mediated inhibition of this pathway by uncoupling IGF-1 receptor phosphorylation from downstream Akt activation. This occurs by interfering with the recruitment of PI3K and mTORC2 to LRs. Akt inhibition can be reversed by sustained IGF-1 stimulation, but only during a time window before psychosine accumulation reaches a threshold level. Our study shows a previously unknown connection between LR-dependent regulation of mTORC2 activity at the cell surface and a genetic neurodegenerative disease. Our results show that LR disruption by psychosine desensitizes cells to extracellular growth factors by inhibiting signal transmission from the plasma membrane to intracellular compartments. This mechanism serves also as a mechanistic model to understand how alterations of the membrane architecture by the progressive accumulation of lipids undermines cell function, with potential implications in other genetic sphingolipidoses and adult neurodegenerative conditions. This article has an associated First Person interview with the first author of the paper.


Insulin-Like Growth Factor I/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Membrane Microdomains/metabolism , Neurons/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sphingolipidoses/genetics , Animals , Brain/metabolism , Cell Survival/drug effects , Cytosol/drug effects , Cytosol/metabolism , Down-Regulation/drug effects , Enzyme Activation/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Lysosomes/drug effects , Membrane Microdomains/drug effects , Mice, Inbred C57BL , Models, Biological , Neurons/drug effects , Phosphorylation/drug effects , Psychosine/pharmacology , Receptor, IGF Type 1/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Sphingolipidoses/metabolism
8.
J Neurosci Res ; 94(11): 1246-60, 2016 11.
Article En | MEDLINE | ID: mdl-27638607

Globoid cell leukodystrophy (GLD) is a rare, rapidly progressing childhood leukodystrophy triggered by deficit of the lysosomal enzyme galactosylceramidase (GALC) and characterized by the accumulation of galactosylsphingosine (psychosine; PSY) in the nervous system. PSY is a cytotoxic sphingolipid, which leads to widespread degeneration of oligodendrocytes and Schwann cells, causing demyelination. Here we report on autophagy in the human oligodendrocyte cell line MO3.13 treated with PSY and exploitation of Li as an autophagy modulator to rescue cell viability. We demonstrate that PSY causes upregulation of the autophagic flux at the level of autophagosome and autolysosome formation and LC3-II expression. We show that pretreatment with Li, a drug clinically used to treat bipolar disorders, can further stimulate autophagy, improving cell tolerance to PSY. This Li protective effect is found not to be linked to reduction of PSY-induced oxidative stress and might not stem from a reduction of PSY accumulation. These data provide novel information on the intracellular pathways activated during PSY-induced toxicity and suggest the autophagy pathway as a promising novel therapeutic target for ameliorating the GLD phenotype. © 2016 Wiley Periodicals, Inc.


Autophagy/drug effects , Lithium/pharmacology , Oligodendroglia/drug effects , Psychosine/pharmacology , Analysis of Variance , Annexin A5/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Dose-Response Relationship, Drug , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lactosylceramides/genetics , Lactosylceramides/metabolism , Psychosine/metabolism , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction/drug effects , Transfection , Tubulin/genetics , Tubulin/metabolism
9.
Mol Biol Cell ; 27(13): 2037-50, 2016 07 01.
Article En | MEDLINE | ID: mdl-27170180

Endomitosis is a special type of mitosis in which only cytokinesis-the final step of the cell division cycle-is defective, resulting in polyploid cells. Although endomitosis is biologically important, its regulatory aspects remain elusive. Psychosine, a lysogalactosylceramide, prevents proper cytokinesis when supplemented to proliferating cells. Cytokinetic inhibition by psychosine does not inhibit genome duplication. Consequently cells undergo multiple rounds of endomitotic cell cycles, resulting in the formation of giant multiploid cells. Here we successfully quantified psychosine-triggered multiploid cell formation, showing that membrane sphingolipids ratios modulate psychosine-triggered polyploidy in Namalwa cells. Among enzymes that experimentally remodel cellular sphingolipids, overexpression of glucosylceramide synthase to biosynthesize glycosylsphingolipids (GSLs) and neutral sphingomyelinase 2 to hydrolyze sphingomyelin (SM) additively enhanced psychosine-triggered multiploidy; almost all of the cells became polyploid. In the presence of psychosine, Namalwa cells showed attenuated cell surface SM clustering and suppression of phosphatidylinositol 4,5-bisphosphate production at the cleavage furrow, both important processes for cytokinesis. Depending on the sphingolipid balance between GSLs and SM, Namalwa cells could be effectively converted to viable multiploid cells with psychosine.


Phosphatidylinositols/metabolism , Psychosine/metabolism , Animals , Cell Cycle/physiology , Cell Membrane/metabolism , Cleavage Stage, Ovum , Cytokinesis/physiology , Glucosyltransferases , Humans , Membranes , Mitosis/drug effects , Mitosis/physiology , Polyploidy , Psychosine/pharmacology , Sphingolipids/metabolism
10.
Apoptosis ; 21(1): 25-35, 2016 Jan.
Article En | MEDLINE | ID: mdl-26459425

Krabbe disease is a genetic demyelinating syndrome characterized by deficiency of the enzyme ß-galactosylceramidase, lysosomal psychosine accumulation, and loss of myelin-forming cells. In this study, some apoptotic markers such as apoptotic index (AI), DNA fragmentation, caspase-3, PTEN, Bad, and PI3K were determined in oligodendrocyte precursors from wild type or twitcher mice untreated or treated with psychosine. Twitcher is a natural mouse model of Krabbe disease containing a premature stop codon (W339X) in the ß-galactosylceramidase gene. Moreover, a possible involvement of connexin (Cx)43 in cell death of oligodendrocyte precursors induced by psychosine was investigated with the final aim to provide a contribution to the knowledge of the molecular mechanisms and pathophysiological events that occur in Krabbe disease. Connexins are a multigene family of structurally related trans-membrane proteins able to modulate essential cellular processes such as proliferation, differentiation and migration. Among these, Cx43 is the predominant isoform in many cell types, including neural progenitor cells. Our results showed an increase of AI, DNA fragmentation, caspase-3, PTEN, Bad, and Cx43 associated to a decrease of PI3K, pAKT and pBad. Taken together, these findings suggest an involvement of Cx43 in the psychosine-mediated apoptosis of primary oligodendrocyte progenitors from wild type or twitcher mice, used for the first time as cell models in comparison. It could open unexplored perspective also for other demyelinating diseases.


Brain/drug effects , Connexin 43/genetics , Galactosylceramidase/deficiency , Leukodystrophy, Globoid Cell/genetics , Oligodendroglia/drug effects , Psychosine/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Brain/enzymology , Brain/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Differentiation/drug effects , Connexin 43/metabolism , DNA Fragmentation/drug effects , Disease Models, Animal , Galactosylceramidase/genetics , Gene Expression Regulation , Humans , Leukodystrophy, Globoid Cell/enzymology , Leukodystrophy, Globoid Cell/pathology , Lysosomes/drug effects , Lysosomes/enzymology , Lysosomes/pathology , Mice , Mice, Knockout , Oligodendroglia/enzymology , Oligodendroglia/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Psychosine/metabolism , Signal Transduction , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
11.
J Cell Sci ; 128(21): 3878-87, 2015 Nov 01.
Article En | MEDLINE | ID: mdl-26359302

Globoid cell leukodystrophy (Krabbe disease) is a rare infantile neurodegenerative disorder. Krabbe disease is caused by deficiency in the lysosomal enzyme galactocerebrosidase (GALC) resulting in accumulation, in the micromolar range, of the toxic metabolite galactosylsphingosine (psychosine) in the brain. Here we find that psychosine induces human astrocyte cell death probably via an apoptotic process in a concentration- and time-dependent manner (EC50 ∼ 15 µM at 4 h). We show these effects of psychosine are attenuated by pre-treatment with the sphingosine 1-phosphate receptor agonist pFTY720 (fingolimod) (IC50 ∼ 100 nM). Psychosine (1 µM, 10 µM) also enhances LPS-induced (EC50 ∼ 100 ng/ml) production of pro-inflammatory cytokines in mouse astrocytes, which is also attenuated by pFTY720 (1 µM). Most notably, for the first time, we show that psychosine, at a concentration found in the brains of patients with Krabbe disease (EC50 ∼ 100 nM), directly induces demyelination in mouse organotypic cerebellar slices in a manner that is independent of pro-inflammatory cytokine response and that pFTY720 (0.1 nM) significantly inhibits. These results support the idea that psychosine is a pathogenic agent in Krabbe disease and suggest that sphingosine 1-phosphate signalling could be a potential drug target for this disorder.


Demyelinating Diseases/chemically induced , Psychosine/pharmacology , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Humans , Leukodystrophy, Globoid Cell/drug therapy , Leukodystrophy, Globoid Cell/metabolism , Lipopolysaccharides/pharmacology , Lysophospholipids , Mice , Microscopy, Fluorescence , Psychosine/therapeutic use , Rats , Signal Transduction/drug effects , Sphingosine/analogs & derivatives
12.
PLoS One ; 10(8): e0135025, 2015.
Article En | MEDLINE | ID: mdl-26263376

Blood coagulation reactions are strongly influenced by phospholipids, but little is known about the influence of sphingolipids on coagulation mechanisms. Lysosulfatide (lyso-SF) (sulfogalactosyl sphingosine) prolonged factor Xa (fXa) 1-stage plasma clotting assays, showing it had robust anticoagulant activity. In studies using purified clotting factors, lyso-SF inhibited >90% of prothrombin (II) activation for reaction mixtures containing fXa/factor Va (fVa)/II, and also inhibited II activation generation by fXa/ phospholipids and by Gla-domainless-fXa/fVa/phospholipids. When lyso-SF analogs were tested, results showed that N-acetyl-sulfatide was not anticoagulant, implying that the free amine group was essential for the anticoagulant effects of lyso-SF. Lyso-SF did not inhibit fXa enzymatic hydrolysis of small peptide substrates, showing it did not directly inhibit the fXa activity. In surface plasmon resonance studies, lyso-SF bound to immobilized inactivated fXa as well as inactivated Gla-domainless-fXa. Confirming this lyso-SF:fXa interaction, fluorescence studies showed that fluorescently-labeled-fXa in solution bound to lyso-SF. Thus, lyso-SF is an anticoagulant lipid that inhibits fXa when this enzyme is bound to either phospholipids or to fVa. Mechanisms for inhibition of procoagulant activity are likely to involve lyso-SF binding to fXa domain(s) that are distinct from the fXa Gla domain. This suggests that certain sphingolipids, including lyso-SF and some of its analogs, may down-regulate fXa activity without inhibiting the enzyme's active site or binding to the fXa Gla domain.


Factor V/metabolism , Factor Xa/metabolism , Psychosine/analogs & derivatives , Thrombin/biosynthesis , Blood Coagulation/drug effects , Factor Va/metabolism , Factor Xa/chemistry , Factor Xa/pharmacology , Humans , Multiprotein Complexes/metabolism , Phospholipids/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Prothrombin/antagonists & inhibitors , Psychosine/chemistry , Psychosine/metabolism , Psychosine/pharmacology , Thrombin/antagonists & inhibitors
13.
J Endocrinol Invest ; 38(8): 891-9, 2015 Aug.
Article En | MEDLINE | ID: mdl-25841894

BACKGROUND: It was recently reported that G protein-coupled receptor 65 (GPR65) suppresses ovariectomy-induced bone loss. AIM: The present study investigated the role of the lysosphingolipid psychosine, a GPR65 ligand, on osteoclastic differentiation and bone resorption. METHODS: Osteoclasts were differentiated from mouse bone marrow macrophages. Tartrate-resistant acid phosphatase-positive multinucleated cells were considered to be osteoclasts, and the resorption area was measured by incubating the cells on dentine discs. The expression levels of osteoclast differentiation markers were assessed by qRT-PCR. GPR65 siRNA and its scrambled siRNA were transfected with lipofectamine. Intracellular cyclic adenosine monophosphate (cAMP) levels were assessed using a direct enzyme immunoassay. RESULTS: Psychosine inhibited osteoclastogenesis and in vitro bone resorption without any significant effect on the viability of pre-osteoclasts, decreased the expression of osteoclast differentiation markers significantly, and increased intracellular cAMP levels. The knockdown of GPR65 by its siRNA restored osteoclastogenesis and decreased cAMP levels in the presence of psychosine. CONCLUSION: Psychosine inhibits osteoclastogenesis by increasing intracellular cAMP levels via GPR65.


Bone Resorption/metabolism , Bone Resorption/prevention & control , Osteoclasts/drug effects , Psychosine/pharmacology , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Osteoclasts/physiology , Psychosine/therapeutic use , Receptors, G-Protein-Coupled/metabolism
14.
Cell Death Dis ; 5: e1529, 2014 Nov 20.
Article En | MEDLINE | ID: mdl-25412308

Globoid cell leukodystrophy (GLD) is a metabolic disease caused by mutations in the galactocerebrosidase (GALC) gene. GALC is a lysosomal enzyme whose function is to degrade galacto-lipids, including galactosyl-ceramide and galactosyl-sphingosine (psychosine, PSY). GALC loss of function causes progressive intracellular accumulation of PSY. It is widely held that PSY is the main trigger for the degeneration of myelinating cells and progressive white-matter loss. However, still little is known about the molecular mechanisms by which PSY imparts toxicity. Here, we address the role of calcium dynamics during PSY-induced cell death. Using the human oligodendrocyte cell line MO3.13, we report that cell death by PSY is accompanied by robust cytosolic and mitochondrial calcium (Ca(2+)) elevations, and by mitochondrial reactive oxygen species (ROS) production. Importantly, we demonstrate that the reduction of extracellular calcium content by the chelating agent ethylenediaminetetraacetic acid can decrease intra-mitochondrial ROS production and enhance cell viability. Antioxidant administration also reduces mitochondrial ROS production and cell loss, but this treatment does not synergize with Ca(2+) chelation. Our results disclose novel intracellular pathways involved in PSY-induced death that may be exploited for therapeutic purposes to delay GLD onset and/or slow down its progression.


Calcium/metabolism , Mitochondria/drug effects , Oligodendroglia/drug effects , Psychosine/pharmacology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Calcium Chelating Agents/pharmacology , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Culture Media/chemistry , Cytosol/drug effects , Cytosol/metabolism , Edetic Acid/pharmacology , Humans , Mitochondria/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Psychosine/antagonists & inhibitors
15.
J Neuropathol Exp Neurol ; 73(10): 964-74, 2014 Oct.
Article En | MEDLINE | ID: mdl-25192051

Globoid cell leukodystrophy (GLD), or Krabbe disease, is a rare and often fatal demyelinating disease caused by mutations in the galactocerebrosidase (galc) gene that result in accumulation of galactosylsphingosine (psychosine). We recently reported that the extracellular matrix (ECM) protease, matrix metalloproteinase-3, is elevated in GLD and that it regulates psychosine-induced microglial activation. Here, we examined central nervous system ECM component expression in human GLD patients and in the twitcher mouse model of GLD using immunohistochemistry. The influence of ECM proteins on primary murine microglial responses to psychosine was evaluated using ECM proteins as substrates and analyzed by quantitative real-time polymerase chain reaction, immunocytochemistry, and ELISA. Functional analysis of microglial cytotoxicity was performed on oligodendrocytes in coculture, and cell death was measured by lactose dehydrogenase assay. Tenascin-C (TnC) was expressed at higher levels in human GLD and in twitcher mice versus controls. Microglial responses to psychosine were enhanced by TnC, as determined by an increase in globoid-like cell formation, matrix metalloproteinase-3 mRNA expression, and higher toxicity toward oligodendrocytes in culture. These findings were consistent with a shift toward the M1 microglial phenotype in TnC-grown microglia. Thus, elevated TnC expression in GLD modified microglial responses to psychosine. These data offer a novel perspective and enhance understanding of the microglial contribution to GLD pathogenesis.


Leukodystrophy, Globoid Cell/metabolism , Microglia/physiology , Psychosine/pharmacology , Tenascin/biosynthesis , Animals , Animals, Newborn , Cells, Cultured , Child, Preschool , Coculture Techniques , Humans , Infant , Leukodystrophy, Globoid Cell/pathology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology
16.
Cancer Lett ; 348(1-2): 29-37, 2014 Jun 28.
Article En | MEDLINE | ID: mdl-24631520

To identify novel anti-cancer agents, we created and screened a unique nutraceutical library for activity against acute myeloid leukemia (AML) cells. From this screen, we determined that glucopsychosine was selectively toxic toward AML cell lines and primary AML patient samples with no effect toward normal hematopoietic cells. It delayed tumor growth and reduced tumor weights in mouse xenograft models without imparting toxicity. Glucopsychosine increased cytosolic calcium and induced apoptosis through calpain enzymes. Extracellular calcium was functionally important for glucopsychosine-induced AML cell death and surface calcium channel expression is altered in AML cells highlighting a unique mechanism of glucopsychosine's selectivity.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Calpain/metabolism , Leukemia, Myeloid, Acute/drug therapy , Psychosine/analogs & derivatives , Animals , Calcium Channels/drug effects , Calcium Channels/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Psychosine/pharmacology , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Am J Pathol ; 184(2): 382-96, 2014 Feb.
Article En | MEDLINE | ID: mdl-24316110

Globoid cell leukodystrophy is a lysosomal storage disease characterized by the loss of galactocerebrosidase. Galactocerebrosidase loss leads to the accumulation of psychosine and subsequent oligodendrocyte cell death, demyelination, macrophage recruitment, and astroglial activation and proliferation. To date, no studies have elucidated the mechanism of glial cell activation and cytokine and chemokine up-regulation and release. We explored a novel explanation for the development of the pathological changes in the early stages of globoid cell leukodystrophy associated with toll-like receptor (TLR) 2 up-regulation in the hindbrain and cerebellum as a response to dying oligodendrocytes. TLR2 up-regulation on microglia/macrophages coincided with morphological changes consistent with activation at 2 and 3 weeks of age. TLR2 up-regulation on activated microglia/macrophages resulted in astrocyte activation and marked up-regulation of cytokines/chemokines. Because oligodendrocyte cell death is an important feature of globoid cell leukodystrophy, we tested the ability of TLR2 reporter cells to respond to oligodendrocyte cell death. These reporter cells responded in vitro to medium conditioned by psychosine-treated oligodendrocytes, indicating the likelihood that oligodendrocytes release a TLR2 ligand during apoptosis. TLRs are a member of the innate immune system and initiate immune and inflammatory events; therefore, the identification of TLR2 as a potential driver in the activation of central nervous system glial activity in globoid cell leukodystrophy may provide important insight into its pathogenesis.


Immunity, Innate , Leukodystrophy, Globoid Cell/etiology , Leukodystrophy, Globoid Cell/immunology , Aging/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Calcium-Binding Proteins/metabolism , Cell Aggregation/drug effects , Cell Line , Cell Shape/drug effects , Chemokines/metabolism , Disease Models, Animal , Fluorescence , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunity, Innate/drug effects , Leukodystrophy, Globoid Cell/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Myelin Sheath/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Psychosine/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rhombencephalon/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Up-Regulation/drug effects
18.
Brain ; 136(Pt 9): 2859-75, 2013 Sep.
Article En | MEDLINE | ID: mdl-23983033

Globoid cell leukodystrophy (Krabbe disease) is a neurological disorder of infants caused by genetic deficiency of the lysosomal enzyme ß-galactosylceramidase leading to accumulation of the neurotoxic metabolite 1-ß-d-galactosylsphingosine (psychosine) in the central nervous system. Angiogenesis plays a pivotal role in the physiology and pathology of the brain. Here, we demonstrate that psychosine has anti-angiogenic properties by causing the disassembling of endothelial cell actin structures at micromolar concentrations as found in the brain of patients with globoid cell leukodystrophy. Accordingly, significant alterations of microvascular endothelium were observed in the post-natal brain of twitcher mice, an authentic model of globoid cell leukodystrophy. Also, twitcher endothelium showed a progressively reduced capacity to respond to pro-angiogenic factors, defect that was corrected after transduction with a lentiviral vector harbouring the murine ß-galactosylceramidase complementary DNA. Finally, RNA interference-mediated ß-galactosylceramidase gene silencing causes psychosine accumulation in human endothelial cells and hampers their mitogenic and motogenic response to vascular endothelial growth factor. Accordingly, significant alterations were observed in human microvasculature from brain biopsy of a globoid cell leukodystrophy case. Together these data demonstrate that ß-galactosylceramidase deficiency induces significant alterations in endothelial neovascular responses that may contribute to central nervous system and systemic damages that occur in globoid cell leukodystrophy.


Leukodystrophy, Globoid Cell/complications , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/pathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aorta/pathology , Aorta/ultrastructure , Biocompatible Materials , Brain/drug effects , Brain/pathology , Brain/ultrastructure , Cattle , Cell Movement/drug effects , Cell Movement/genetics , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/metabolism , Collagen/toxicity , Disease Models, Animal , Drug Combinations , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Green Fluorescent Proteins/metabolism , Humans , Laminin/toxicity , Leukodystrophy, Globoid Cell/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Neovascularization, Pathologic/prevention & control , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proteoglycans/toxicity , Psychosine/metabolism , Psychosine/pharmacology , RNA, Small Interfering/administration & dosage , Time Factors , Transfection , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zonula Occludens-1 Protein
19.
J Neurosci ; 33(24): 10048-56, 2013 Jun 12.
Article En | MEDLINE | ID: mdl-23761900

Loss of function of galactosylceramidase lysosomal activity causes demyelination and vulnerability of various neuronal populations in Krabbe disease. Psychosine, a lipid-raft-associated sphingolipid that accumulates in this disease, is thought to trigger these abnormalities. Myelin-free in vitro analyses showed that psychosine inhibited fast axonal transport through the activation of axonal PP1 and GSK3ß in the axon. Abnormal levels of activated GSK3ß and abnormally phosphorylated kinesin light chains were found in nerve samples from a mouse model of Krabbe disease. Administration of GSK3ß inhibitors significantly ameliorated transport defects in vitro and in vivo in peripheral axons of the mutant mouse. This study identifies psychosine as a pathogenic sphingolipid able to block fast axonal transport and is the first to provide a molecular mechanism underlying dying-back degeneration in this genetic leukodystrophy.


Axonal Transport/drug effects , Glycogen Synthase Kinase 3/metabolism , Leukodystrophy, Globoid Cell/pathology , Molecular Motor Proteins/metabolism , Neurons/pathology , Psychosine/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/pathology , Disease Models, Animal , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Glycogen Synthase Kinase 3 beta , Leukodystrophy, Globoid Cell/drug therapy , Leukodystrophy, Globoid Cell/genetics , Membrane Microdomains/drug effects , Membrane Microdomains/enzymology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/physiology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/ultrastructure , Sciatic Nerve/pathology , Time Factors
20.
Neurobiol Dis ; 46(2): 325-35, 2012 May.
Article En | MEDLINE | ID: mdl-22326830

Patients with Krabbe disease, a genetic demyelinating syndrome caused by deficiency of galactosyl-ceramidase and the resulting accumulation of galactosyl-sphingolipids, develop signs of a dying-back axonopathy compounded by a deficiency of large-caliber axons. Here, we show that axonal caliber in Twitcher mice, an animal model for Krabbe disease, is impaired in peripheral axons and is accompanied by a progressive reduction in the abundance and phosphorylation of the three neurofilament (NF) subunits. These changes correlate with an increase in the density of NFs per cross-sectional area in numerous mutant peripheral axons and abnormal increases in the activity of two serine/threonine phosphatases (PP1 and PP2A) in mutant tissue. Similarly, acutely isolated mutant cortical neurons show abnormal phosphorylation of NFs. Psychosine, the neurotoxin accumulated in Krabbe disease, was sufficient to induce abnormal dephosphorylation of NF subunits in a normal motor neuron cell line as well as in acutely isolated normal cortical neurons. This in vitro effect was mediated by PP1 and PP2A, which specifically dephosphorylated NFs. These results demonstrate that the reduced caliber observed in some axons in Krabbe disease involves abnormal dephosphorylation of NFs. We propose that a psychosine-driven pathogenic mechanism through deregulated phosphotransferase activities may be involved in this process.


Neurofilament Proteins/metabolism , Protein Phosphatase 1/physiology , Protein Phosphatase 2/physiology , Psychosine/pharmacology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/pathology , Myelin Sheath/physiology , Neural Inhibition/genetics , Neurofilament Proteins/antagonists & inhibitors , Neurotoxins/pharmacology , Phosphorylation/physiology , Up-Regulation/genetics
...