Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
AJNR Am J Neuroradiol ; 41(10): 1943-1948, 2020 10.
Article in English | MEDLINE | ID: mdl-32855188

ABSTRACT

SGPL1 encodes sphingosine-1-phosphate lyase, the final enzyme of sphingolipid metabolism. In 2017, a condition featuring steroid-resistant nephrotic syndrome and/or adrenal insufficiency associated with pathogenic SGPL1 variants was reported. In addition to the main features of the disease, patients often exhibit a range of neurologic deficits. In a handful of cases, brain imaging results were described. However, high-quality imaging results and a systematic analysis of brain MR imaging findings associated with the condition are lacking. In this study, MR images from 4 new patients and additional published case reports were reviewed by a pediatric neuroradiologist. Analysis reveals recurring patterns of features in affected patients, including isolated callosal dysgenesis and prominent involvement of the globus pallidus, thalamus, and dentate nucleus, with progressive atrophy and worsening of brain lesions. MR imaging findings of abnormal deep gray nuclei, microcephaly, or callosal dysgenesis in an infant or young child exhibiting other typical clinical features of sphingosine-1-phosphate lyase insufficiency syndrome should trigger prompt genetic testing for SGPL1 mutations.


Subject(s)
Aldehyde-Lyases/deficiency , Brain/diagnostic imaging , Brain/pathology , Metabolism, Inborn Errors/diagnostic imaging , Metabolism, Inborn Errors/pathology , Aldehyde-Lyases/genetics , Child , Child, Preschool , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Metabolism, Inborn Errors/genetics , Mutation , Nephrotic Syndrome/enzymology , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology
2.
J Steroid Biochem Mol Biol ; 202: 105730, 2020 09.
Article in English | MEDLINE | ID: mdl-32682944

ABSTRACT

Deficiency in Sphingosine-1-phosphate lyase (S1P lyase) is associated with a multi-systemic disorder incorporating primary adrenal insufficiency (PAI), steroid resistant nephrotic syndrome and neurological dysfunction. Accumulation of sphingolipid intermediates, as seen with loss of function mutations in SGPL1, has been implicated in mitochondrial dysregulation, including alterations in mitochondrial membrane potentials and initiation of mitochondrial apoptosis. For the first time, we investigate the impact of S1P lyase deficiency on mitochondrial morphology and function using patient-derived human dermal fibroblasts and CRISPR engineered SGPL1-knockout HeLa cells. Reduced cortisol output in response to progesterone stimulation was observed in two patient dermal fibroblast cell lines. Mass spectrometric analysis of patient dermal fibroblasts revealed significantly elevated levels of sphingosine-1-phosphate, sphingosine, ceramide species and sphingomyelin when compared to control. Total mitochondrial volume was reduced in both S1P lyase deficient patient and HeLa cell lines. Mitochondrial dynamics and parameters of oxidative phosphorylation were altered when compared to matched controls, though differentially across the cell lines. Mitochondrial dysfunction may represent a major event in the pathogenesis of this disease, associated with severity of phenotype.


Subject(s)
Adrenal Insufficiency/metabolism , Aldehyde-Lyases/deficiency , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Adrenal Insufficiency/genetics , Aldehyde-Lyases/genetics , Cell Respiration , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hydrocortisone/metabolism , Mitochondrial Diseases/genetics , Phosphoproteins/genetics , Progesterone/pharmacology , Skin/cytology
3.
Article in English | MEDLINE | ID: mdl-32283310

ABSTRACT

Sphingosine-1-phosphate (S1P) is not only a catabolic intermediate of all sphingolipids but also an evolutionary conserved bioactive lipid with critical functions in cell survival, differentiation, and migration as well as in immunity and angiogenesis. S1P-lyase (SGPL1) irreversibly cleaves S1P in the final step of sphingolipid catabolism. As sphingoid bases and their 1-phosphates are not only metabolic intermediates but also highly bioactive lipids that modulate a wide range of physiological processes, it would be predicted that their elevation might induce adjustments in other facets of sphingolipid metabolism and/or alter cell behavior. We actually found in a previous study that in terminally differentiated neurons SGPL1 deficiency increases sphingolipid formation via recycling at the expense of de novo synthesis. We now investigated whether and how SGPL1 deficiency affects the metabolism of (glyco)sphingolipids in mouse embryonic fibroblasts (MEFs). According to our previous experiments in neurons, we found a strong accumulation of S1P in SGPL1-deficient MEFs. Surprisingly, a completely different situation arose as we analyzed sphingolipid metabolism in this non-differentiated cell type. The production of biosynthetic precursors of complex glycosphingolipids including ceramide, glucosylceramide and also ganglioside GM3 via de novo synthesis and recycling pathway was substantially increased whereas the amount of more complex gangliosides dropped significantly.


Subject(s)
Aldehyde-Lyases/deficiency , Aldehyde-Lyases/metabolism , Carcinogenesis , Gangliosides/biosynthesis , Animals , Cells, Cultured , Gangliosides/analysis , Mice , Mice, Knockout
4.
Adv Biol Regul ; 71: 128-140, 2019 01.
Article in English | MEDLINE | ID: mdl-30274713

ABSTRACT

Sphingosine-1-phosphate lyase (SPL) is an intracellular enzyme that controls the final step in the sphingolipid degradative pathway, the only biochemical pathway for removal of sphingolipids. Specifically, SPL catalyzes the cleavage of sphingosine 1-phosphate (S1P) at the C2-3 carbon bond, resulting in its irreversible degradation to phosphoethanolamine (PE) and hexadecenal. The substrate of the reaction, S1P, is a bioactive sphingolipid metabolite that signals through a family of five G protein-coupled S1P receptors (S1PRs) to mediate biological activities including cell migration, cell survival/death/proliferation and cell extrusion, thereby contributing to development, physiological functions and - when improperly regulated - the pathophysiology of disease. In 2017, several groups including ours reported a novel childhood syndrome that featured a wide range of presentations including fetal hydrops, steroid-resistant nephrotic syndrome (SRNS), primary adrenal insufficiency (PAI), rapid or insidious neurological deterioration, immunodeficiency, acanthosis and endocrine abnormalities. In all cases, the disease was attributed to recessive mutations in the human SPL gene, SGPL1. We now refer to this condition as SPL Insufficiency Syndrome, or SPLIS. Some features of this new sphingolipidosis were predicted by the reported phenotypes of Sgpl1 homozygous null mice that serve as vertebrate SPLIS disease models. However, other SPLIS features reveal previously unrecognized roles for SPL in human physiology. In this review, we briefly summarize the biochemistry, functions and regulation of SPL, the main clinical and biochemical features of SPLIS and what is known about the pathophysiology of this condition from murine and cell models. Lastly, we consider potential therapeutic strategies for the treatment of SPLIS patients.


Subject(s)
Aldehyde-Lyases/deficiency , Cell Movement , Lipid Metabolism, Inborn Errors , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Disease Models, Animal , Humans , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Lysophospholipids/genetics , Mice , Mice, Mutant Strains , Sphingosine/genetics , Sphingosine/metabolism , Syndrome
5.
J Clin Endocrinol Metab ; 104(5): 1484-1490, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30517686

ABSTRACT

CONTEXT: Multiple autosomal recessive genes have been etiologically linked to primary adrenal insufficiency (PAI). Recently, sphingosine-1-phosphate lyase 1 (SGPL1) gene mutations were recognized as a cause of steroid-resistant nephrotic syndrome type 14 (NPHS14), a sphingolipidosis with multisystemic manifestations, including PAI. OBJECTIVE: To check if SGPL1 mutations are involved in the pathogenesis of PAI in patients who do not exhibit nephrotic syndrome. METHODS: Sequencing of the SGPL1 gene in 21 patients with familial glucocorticoid disease or triple A syndrome. RESULTS: We identified two missense SGPL1 variants in four patients, two of whom were first cousins. We describe in detail the proband, a boy born to Saudi Arabian consanguineous parents with a homozygous c.665G>A, p.R222Q SGPL1 variant. The patient presented with hypoglycemia and seizures at age 2 years and was ultimately diagnosed with PAI (isolated glucocorticoid deficiency). Brain MRI showed abnormalities in the basal ganglia consistent with a degenerative process albeit the patient had no neurologic symptoms. CONCLUSIONS: New genetic causes of PAI continue to be identified. We suggest that screening for SGPL1 mutations should not be reserved only for patients with nephrotic syndrome but may also include patients with PAI who lack other clinical manifestations of NPHS14 because, in certain cases, kidney disease and accompanying features might develop. Timely diagnosis of this specific sphingolipidosis while the kidneys still function normally can lead to prompt initiation of therapy and improve outcome.


Subject(s)
Addison Disease/etiology , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Biomarkers/analysis , Mutation , Addison Disease/pathology , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Pedigree , Prognosis , Risk Factors
6.
J Immunol ; 199(2): 677-687, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28600291

ABSTRACT

Sphingosine 1-phosphate (S1P) lyase (SPL) is an intracellular enzyme that mediates the irreversible degradation of the bioactive lipid S1P. We have previously reported that overexpressed SPL displays anti-influenza viral activity; however, the underlying mechanism is incompletely understood. In this study, we demonstrate that SPL functions as a positive regulator of IKKε to propel type I IFN-mediated innate immune responses against viral infection. Exogenous SPL expression inhibited influenza A virus replication, which correlated with an increase in type I IFN production and IFN-stimulated gene accumulation upon infection. In contrast, the lack of SPL expression led to an elevated cellular susceptibility to influenza A virus infection. In support of this, SPL-deficient cells were defective in mounting an effective IFN response when stimulated by influenza viral RNAs. SPL augmented the activation status of IKKε and enhanced the kinase-induced phosphorylation of IRF3 and the synthesis of type I IFNs. However, the S1P degradation-incompetent form of SPL also enhanced IFN responses, suggesting that SPL's pro-IFN function is independent of S1P. Biochemical analyses revealed that SPL, as well as the mutant form of SPL, interacts with IKKε. Importantly, when endogenous IKKε was downregulated using a small interfering RNA approach, SPL's anti-influenza viral activity was markedly suppressed. This indicates that IKKε is crucial for SPL-mediated inhibition of influenza virus replication. Thus, the results illustrate the functional significance of the SPL-IKKε-IFN axis during host innate immunity against viral infection.


Subject(s)
Aldehyde-Lyases/metabolism , I-kappa B Kinase/metabolism , Immunity, Innate , Influenza A virus/immunology , Interferon Type I/immunology , A549 Cells , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Down-Regulation , Enzyme Activation , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Influenza A virus/physiology , Interferon Regulatory Factor-3/metabolism , Interferon Type I/biosynthesis , Lysophospholipids/metabolism , Phosphorylation , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Virus Replication
7.
Sci Rep ; 7: 43575, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262793

ABSTRACT

Sphingosine-1-phosphate (S1P) lyase irreversibly cleaves S1P, thereby catalysing the ultimate step of sphingolipid degradation. We show here that embryonic fibroblasts from S1P lyase-deficient mice (Sgpl1-/--MEFs), in which S1P and sphingosine accumulate, have features of Niemann-Pick disease type C (NPC) cells. In the presence of serum, overall cholesterol content was elevated in Sgpl1-/--MEFs, due to upregulation of the LDL receptor and enhanced cholesterol uptake. Despite this, activation of sterol regulatory element-binding protein-2 was increased in Sgpl1-/--MEFs, indicating a local lack of cholesterol at the ER. Indeed, free cholesterol was retained in NPC1-containing vesicles, which is a hallmark of NPC. Furthermore, upregulation of amyloid precursor protein in Sgpl1-/--MEFs was mimicked by an NPC1 inhibitor in Sgpl1+/+-MEFs and reduced by overexpression of NPC1. Lysosomal pH was not altered by S1P lyase deficiency, similar to NPC. Interestingly, lysosomal Ca2+ content and bafilomycin A1-induced [Ca2+]i increases were enhanced in Sgpl1-/--MEFs, contrary to NPC. These results show that both a primary defect in cholesterol trafficking and S1P lyase deficiency cause overlapping phenotypic alterations, and challenge the present view on the role of sphingosine in lysosomal Ca2+ homeostasis.


Subject(s)
Aldehyde-Lyases/deficiency , Calcium/metabolism , Cholesterol/metabolism , Fibroblasts/metabolism , Lysosomes/metabolism , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Aldehyde-Lyases/blood , Animals , Biomarkers , Disease Models, Animal , Histone Deacetylases , Homeostasis , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Mice , Mice, Knockout , Niemann-Pick Disease, Type C/diagnosis , Phenotype
8.
Hum Mutat ; 38(4): 365-372, 2017 04.
Article in English | MEDLINE | ID: mdl-28181337

ABSTRACT

We identified two unrelated consanguineous families with three children affected by the rare association of congenital nephrotic syndrome (CNS) diagnosed in the first days of life, of hypogonadism, and of prenatally detected adrenal calcifications, associated with congenital adrenal insufficiency in one case. Using exome sequencing and targeted Sanger sequencing, two homozygous truncating mutations, c.1513C>T (p.Arg505*) and c.934delC (p.Leu312Phefs*30), were identified in SGPL1-encoding sphingosine-1-phosphate (S1P) lyase 1. SGPL1 catalyzes the irreversible degradation of endogenous and dietary S1P, the final step of sphingolipid catabolism, and of other phosphorylated long-chain bases. S1P is an intracellular and extracellular signaling molecule involved in angiogenesis, vascular maturation, and immunity. The levels of SGPL1 substrates, S1P, and sphingosine were markedly increased in the patients' blood and fibroblasts, as determined by liquid chromatography-tandem mass spectrometry. Vascular alterations were present in a patient's renal biopsy, in line with changes seen in Sgpl1 knockout mice that are compatible with a developmental defect in vascular maturation. In conclusion, loss of SGPL1 function is associated with CNS, adrenal calcifications, and hypogonadism.


Subject(s)
Adrenal Gland Diseases/genetics , Aldehyde-Lyases/genetics , Calcinosis/genetics , Mutation , Nephrotic Syndrome/genetics , Adrenal Gland Diseases/congenital , Adrenal Gland Diseases/enzymology , Adult , Aldehyde-Lyases/deficiency , Animals , Base Sequence , Calcinosis/enzymology , Consanguinity , Female , Humans , Infant , Lysophospholipids/blood , Lysophospholipids/metabolism , Male , Mice, Knockout , Nephrotic Syndrome/congenital , Nephrotic Syndrome/enzymology , Pedigree , Sequence Analysis, DNA/methods , Sphingosine/analogs & derivatives , Sphingosine/blood , Sphingosine/metabolism
9.
Neurology ; 88(6): 533-542, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28077491

ABSTRACT

OBJECTIVE: To identify the unknown genetic cause in a nuclear family with an axonal form of peripheral neuropathy and atypical disease course. METHODS: Detailed neurologic, electrophysiologic, and neuropathologic examinations of the patients were performed. Whole exome sequencing of both affected individuals was done. The effect of the identified sequence variations was investigated at cDNA and protein level in patient-derived lymphoblasts. The plasma sphingoid base profile was analyzed. Functional consequences of neuron-specific downregulation of the gene were studied in Drosophila. RESULTS: Both patients present an atypical form of axonal peripheral neuropathy, characterized by acute or subacute onset and episodes of recurrent mononeuropathy. We identified compound heterozygous mutations cosegregating with disease and absent in controls in the SGPL1 gene, encoding sphingosine 1-phosphate lyase (SPL). The p.Ser361* mutation triggers nonsense-mediated mRNA decay. The missense p.Ile184Thr mutation causes partial protein degradation. The plasma levels of sphingosine 1-phosphate and sphingosine/sphinganine ratio were increased in the patients. Neuron-specific downregulation of the Drosophila orthologue impaired the morphology of the neuromuscular junction and caused progressive degeneration of the chemosensory neurons innervating the wing margin bristles. CONCLUSIONS: We suggest SPL deficiency as a cause of a distinct form of Charcot-Marie-Tooth disease in humans, thus extending the currently recognized clinical and genetic spectrum of inherited peripheral neuropathies. Our data emphasize the importance of sphingolipid metabolism for neuronal function.


Subject(s)
Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Charcot-Marie-Tooth Disease/genetics , Codon, Nonsense , Drosophila Proteins/metabolism , Mutation, Missense , Adult , Animals , Animals, Genetically Modified , Cells, Cultured , Charcot-Marie-Tooth Disease/physiopathology , Cohort Studies , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Lysophospholipids/blood , Male , Neurons/metabolism , Neurons/pathology , Siblings , Sphingosine/analogs & derivatives , Sphingosine/blood
10.
Chem Phys Lipids ; 194: 101-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26408264

ABSTRACT

A new technique for quantifying sphingosine phosphate lyase activity in biological samples is described. In this procedure, 2-hydrazinoquinoline is used to convert (2E)-hexadecenal into the corresponding hydrazone derivative to improve ionization efficiency and selectivity of detection. Combined utilization of liquid chromatographic separation and multiple reaction monitoring-mass spectrometry allows for simultaneous quantification of the substrate S1P and product (2E)-hexadecenal. Incorporation of (2E)- d5-hexadecenal as an internal standard improves detection accuracy and precision. A simple one-step derivatization procedure eliminates the need for further extractions. Limits of quantification for (2E)-hexadecenal and sphingosine-1-phosphate are 100 and 50fmol, respectively. The assay displays a wide dynamic detection range useful for detection of low basal sphingosine phosphate lyase activity in wild type cells, SPL-overexpressing cell lines, and wild type mouse tissues. Compared to current methods, the capacity for simultaneous detection of sphingosine-1-phosphate and (2E)-hexadecenal greatly improves the accuracy of results and shows excellent sensitivity and specificity for sphingosine phosphate lyase activity detection.


Subject(s)
Aldehyde-Lyases/metabolism , Enzyme Assays/methods , Aldehyde-Lyases/deficiency , Animals , Cell Line , Hydrazones/chemical synthesis , Hydrazones/chemistry , Indicator Dilution Techniques , Isotopes , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure
11.
J Lipid Res ; 56(1): 60-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25385827

ABSTRACT

Sphingosine 1-phosphate (S1P) is an extra- and intracellular mediator that regulates cell growth, survival, migration, and adhesion in many cell types. S1P lyase is the enzyme that irreversibly cleaves S1P and thereby constitutes the ultimate step in sphingolipid catabolism. It has been reported previously that embryonic fibroblasts from S1P lyase-deficient mice (Sgpl1(-/-)-MEFs) are resistant to chemotherapy-induced apoptosis through upregulation of B cell lymphoma 2 (Bcl-2) and Bcl-2-like 1 (Bcl-xL). Here, we demonstrate that the transporter proteins Abcc1/MRP1, Abcb1/MDR1, Abca1, and spinster-2 are upregulated in Sgpl1(-/-)-MEFs. Furthermore, the cells efficiently sequestered the substrates of Abcc1 and Abcb1, fluo-4 and doxorubicin, in subcellular compartments. In line with this, Abcb1 was localized mainly at intracellular vesicular structures. After 16 h of incubation, wild-type MEFs had small apoptotic nuclei containing doxorubicin, whereas the nuclei of Sgpl1(-/-)-MEFs appeared unchanged and free of doxorubicin. A combined treatment with the inhibitors of Abcb1 and Abcc1, zosuquidar and MK571, respectively, reversed the compartmentalization of doxorubicin and rendered the cells sensitive to doxorubicin-induced apoptosis. It is concluded that upregulation of multidrug resistance transporters contributes to the chemoresistance of S1P lyase-deficient MEFs.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Aldehyde-Lyases/deficiency , Drug Resistance, Neoplasm , Fibroblasts/drug effects , Up-Regulation/drug effects , Aniline Compounds/pharmacology , Animals , Doxorubicin/pharmacology , Fibroblasts/cytology , Fibroblasts/enzymology , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Protein Transport/drug effects , Xanthenes/pharmacology
12.
Cell Physiol Biochem ; 34(1): 148-57, 2014.
Article in English | MEDLINE | ID: mdl-24977488

ABSTRACT

Sphingosine-1-phosphate (S1P), an evolutionary conserved bioactive lipid, is essential for brain development, but might also exert detrimental effects in terminally differentiated post-mitotic neurons. Its concentration in the brain is tightly regulated by specific kinases and phosphatases, and mainly by the S1P degrading enzyme, S1P-lyase (S1PL). The role of S1P in neurons was initially studied in primary cultures by using structural analogues. During the last 3 years generation of a S1PL deficient mouse model substantially promoted our knowledge on the functional role of S1P metabolism in the brain, and its potential relation to neurodegenerative diseases. However, our understanding of the molecular mechanisms that underlie the physiological and pathophysiological actions of S1P in neurons remains rather scarce.


Subject(s)
Brain/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Lysophospholipids/chemistry , Sphingosine/chemistry , Sphingosine/metabolism , tau Proteins/metabolism
13.
Am J Pathol ; 183(4): 1169-1182, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23933064

ABSTRACT

Bronchopulmonary dysplasia of the premature newborn is characterized by lung injury, resulting in alveolar simplification and reduced pulmonary function. Exposure of neonatal mice to hyperoxia enhanced sphingosine-1-phosphate (S1P) levels in lung tissues; however, the role of increased S1P in the pathobiological characteristics of bronchopulmonary dysplasia has not been investigated. We hypothesized that an altered S1P signaling axis, in part, is responsible for neonatal lung injury leading to bronchopulmonary dysplasia. To validate this hypothesis, newborn wild-type, sphingosine kinase1(-/-) (Sphk1(-/-)), sphingosine kinase 2(-/-) (Sphk2(-/-)), and S1P lyase(+/-) (Sgpl1(+/-)) mice were exposed to hyperoxia (75%) from postnatal day 1 to 7. Sphk1(-/-), but not Sphk2(-/-) or Sgpl1(+/-), mice offered protection against hyperoxia-induced lung injury, with improved alveolarization and alveolar integrity compared with wild type. Furthermore, SphK1 deficiency attenuated hyperoxia-induced accumulation of IL-6 in bronchoalveolar lavage fluids and NADPH oxidase (NOX) 2 and NOX4 protein expression in lung tissue. In vitro experiments using human lung microvascular endothelial cells showed that exogenous S1P stimulated intracellular reactive oxygen species (ROS) generation, whereas SphK1 siRNA, or inhibitor against SphK1, attenuated hyperoxia-induced S1P generation. Knockdown of NOX2 and NOX4, using specific siRNA, reduced both basal and S1P-induced ROS formation. These results suggest an important role for SphK1-mediated S1P signaling-regulated ROS in the development of hyperoxia-induced lung injury in a murine neonatal model of bronchopulmonary dysplasia.


Subject(s)
Bronchopulmonary Dysplasia/enzymology , Bronchopulmonary Dysplasia/prevention & control , Hyperoxia/complications , Lysophospholipids/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Sphingosine/analogs & derivatives , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/metabolism , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Humans , Hyperoxia/enzymology , Hyperoxia/pathology , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidase 4 , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pneumonia/complications , Pneumonia/pathology , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/pathology , Reactive Oxygen Species/metabolism , Signal Transduction , Sphingosine/metabolism , rac1 GTP-Binding Protein/metabolism
14.
PLoS One ; 8(5): e63360, 2013.
Article in English | MEDLINE | ID: mdl-23700419

ABSTRACT

AIMS: Altered sphingosine 1-phosphate (S1P) homeostasis and signaling is implicated in various inflammatory diseases including atherosclerosis. As S1P levels are tightly controlled by S1P lyase, we investigated the impact of hematopoietic S1P lyase (Sgpl1(-/-)) deficiency on leukocyte subsets relevant to atherosclerosis. METHODS AND RESULTS: LDL receptor deficient mice that were transplanted with Sgpl1(-/-) bone marrow showed disrupted S1P gradients translating into lymphopenia and abrogated lymphocyte mitogenic and cytokine response as compared to controls. Remarkably however, Sgpl1(-/-) chimeras displayed mild monocytosis, due to impeded stromal retention and myelopoiesis, and plasma cytokine and macrophage expression patterns, that were largely compatible with classical macrophage activation. Collectively these two phenotypic features of Sgpl1 deficiency culminated in diminished atherogenic response. CONCLUSIONS: Here we not only firmly establish the critical role of hematopoietic S1P lyase in controlling S1P levels and T cell trafficking in blood and lymphoid tissue, but also identify leukocyte Sgpl1 as critical factor in monocyte macrophage differentiation and function. Its, partly counterbalancing, pro- and anti-inflammatory activity spectrum imply that intervention in S1P lyase function in inflammatory disorders such as atherosclerosis should be considered with caution.


Subject(s)
Aldehyde-Lyases/deficiency , Atherosclerosis/enzymology , Plaque, Atherosclerotic/enzymology , Receptors, LDL/deficiency , Aldehyde-Lyases/genetics , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Bone Marrow Cells/enzymology , Cell Differentiation , Female , Hematopoiesis , Lymphocyte Count , Lymphopenia/enzymology , Lymphopenia/immunology , Lysophospholipids/metabolism , Macrophages/enzymology , Macrophages/immunology , Macrophages/physiology , Mice , Mice, Knockout , Neutrophils/enzymology , Phenotype , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Receptors, LDL/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Spleen/metabolism
15.
PLoS One ; 8(3): e59630, 2013.
Article in English | MEDLINE | ID: mdl-23544080

ABSTRACT

BACKGROUND: Sphingosine-1-phosphate (S1P) regulates the egress of T cells from lymphoid organs; levels of S1P in the tissues are controlled by S1P lyase (Sgpl1). Hence, Sgpl1 offers a target to block T cell-dependent inflammatory processes. However, the involvement of Sgpl1 in models of disease has not been fully elucidated yet, since Sgpl1 KO mice have a short life-span. METHODOLOGY: We generated inducible Sgpl1 KO mice featuring partial reduction of Sgpl1 activity and analyzed them with respect to sphingolipid levels, T-cell distribution, and response in models of inflammation. PRINCIPAL FINDINGS: The partially Sgpl1 deficient mice are viable but feature profound reduction of peripheral T cells, similar to the constitutive KO mice. While thymic T cell development in these mice appears normal, mature T cells are retained in thymus and lymph nodes, leading to reduced T cell numbers in spleen and blood, with a skewing towards increased proportions of memory T cells and T regulatory cells. The therapeutic relevance of Sgpl1 is demonstrated by the fact that the inducible KO mice are protected in experimental autoimmune encephalomyelitis (EAE). T cell immigration into the CNS was found to be profoundly reduced. Since S1P levels in the brain of the animals are unchanged, we conclude that protection in EAE is due to the peripheral effect on T cells, leading to reduced CNS immigration, rather than on local effects in the CNS. SIGNIFICANCE: The data suggest Sgpl1 as a novel therapeutic target for the treatment of multiple sclerosis.


Subject(s)
Aldehyde-Lyases/deficiency , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Aldehyde-Lyases/metabolism , Animals , Brain/metabolism , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/complications , Forkhead Transcription Factors/metabolism , Hypersensitivity, Delayed/blood , Hypersensitivity, Delayed/complications , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Immunologic Memory/immunology , Integrases/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Count , Mice , Mice, Knockout , Sheep , Sphingolipids/metabolism , Spleen/immunology , Spleen/pathology , Survival Analysis , Thymus Gland/immunology , Thymus Gland/pathology
16.
J Biol Chem ; 287(12): 9128-36, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22291021

ABSTRACT

Sphingosine 1-phosphate lyase (S1P lyase) irreversibly cleaves sphingosine 1-phosphate (S1P) in the final step of sphingolipid catabolism. As sphingoid bases and their 1-phosphate are not only metabolic intermediates but also highly bioactive lipids that modulate a wide range of physiological processes, it would be predicted that their elevation might induce adjustments in other facets of sphingolipid metabolism and/or alter cell behavior. Indeed, we have previously reported that S1P lyase deficiency causes neurodegeneration and other adverse symptoms. We next asked the question whether and how S1P lyase deficiency affects the metabolism of (glyco)sphingolipids and cholesterol, two lipid classes that might be involved in the neurodegenerative processes observed in S1P lyase-deficient mice. As predicted, there was a considerable increase in free and phosphorylated sphingoid bases upon elimination of S1P lyase, but to our surprise, rather than increasing, the mass of (glyco)sphingolipids persisted at wild type levels. This was discovered to be due to reduced de novo sphingoid base biosynthesis and a corresponding increase in the recycling of the backbones via the salvage pathway. There was also a considerable increase in cholesterol esters, although free cholesterol persisted at wild type levels, which might be secondary to the shifts in sphingolipid metabolism. All in all, these findings show that accumulation of free and phosphorylated sphingoid bases by loss of S1P lyase causes an interesting readjustment of the balance between de novo biosynthesis and recycling to maintain (glyco)sphingolipid homeostasis. These changes, and their impact on the metabolism of other cellular lipids, should be explored as possible contributors to the neurodegeneration in S1P lyase deficiency.


Subject(s)
Aldehyde-Lyases/deficiency , Neurons/enzymology , Sphingolipids/biosynthesis , Aldehyde-Lyases/genetics , Animals , Cells, Cultured , Mice , Mice, Knockout , Neurons/metabolism
17.
J Biotechnol ; 161(2): 174-80, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22222309

ABSTRACT

A new 2-deoxy-D-ribose-5-phoshate aldolase (DERA) gene was cloned from Rhodococcus erythropolis strain DSM 311, recombinantly expressed in Escherichia coli, and purified via affinity chromatography which yielded a homo-dimeric enzyme of 44.3 kDa as apparent by size exclusion chromatography. To characterise the enzyme, investigations about pH and temperature tolerance, stability, as well as analyses on resistance to organic solvents and acetaldehyde were performed. In addition, kinetic constants of the new DERA(RE) were compared to respective values of the DERA from E. coli (DERA(EC)). Stability of DERA(RE) turned out to be a crucial factor: The pH for optimal DERA(RE) activity was determined to be 7.0, whereas the highest stability was achieved at pH 9.0 with a half-life of approximately 20 days. The optimal temperature for DERA(RE) activity was 65 °C, but coupled with a rather low stability (half-life of 2 min). The highest stability was achieved at 25 °C. The new enzyme exhibits high resistance to organic solvents and acetaldehyde with a half-life being 2.5× higher compared to DERA(EC) under the exposure of 300 mM acetaldehyde. Hence it has the potential as a new promising biocatalyst with applications in organic synthesis.


Subject(s)
Bacterial Proteins/chemistry , Recombinant Proteins/chemistry , Rhodococcus/enzymology , Acetaldehyde , Aldehyde-Lyases/chemistry , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cloning, Molecular , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrogen-Ion Concentration , Kinetics , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/metabolism , Molecular Sequence Data , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rhodococcus/genetics , Sequence Alignment , Temperature
18.
Appl Microbiol Biotechnol ; 91(3): 769-76, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21519932

ABSTRACT

The ß-proteobacterium Ralstonia eutropha H16 utilizes fructose and gluconate as carbon sources for heterotrophic growth exclusively via the Entner-Doudoroff pathway with its key enzyme 2-keto-3-deoxy-6-phosphogluconate (KDPG) aldolase. By deletion of the responsible gene eda, we constructed a KDPG aldolase-negative strain, which is disabled to supply pyruvate for energy metabolism from fructose or gluconate as sole carbon sources. To restore growth on fructose, an alternative pathway, similar to the fructose-6-phosphate shunt of heterofermentative bifidobacteria, was established. For this, the xfp gene from Bifidobacterium animalis, coding for a bifunctional xylulose-5-phosphate/fructose-6-phosphate phosphoketolase (Xfp; Meile et al. in J Bacteriol 183:2929-2936, 2001), was expressed in R. eutropha H16 PHB(-)4 Δeda. This Xfp catalyzes the phosphorolytic cleavage of fructose 6-phosphate to erythrose 4-phosphate and acetylphosphate as well as of xylulose 5-phosphate to glyceralaldehyde 3-phosphate and acetylphosphate. The recombinant strain showed phosphoketolase (PKT) activity on either substrate, and was able to use fructose as sole carbon source for growth, because PKT is the only enzyme that is missing in R. eutropha H16 to establish the artificial fructose-6-phosphate shunt. The Xfp-expressing strain R. eutropha H16 PHB(-)4 Δeda (pBBR1MCS-3::xfp) should be applicable for a novel variant of a plasmid addiction system to stably maintain episomally encoded genetic information during fermentative production processes. Plasmid addiction systems are often used to ensure plasmid stability in many biotechnology relevant microorganisms and processes without the need to apply external selection pressure, like the addition of antibiotics. By episomal expression of xfp in a R. eutropha H16 mutant lacking KDPG aldolase activity and cultivation in mineral salt medium with fructose as sole carbon source, the growth of this bacterium was addicted to the constructed xfp harboring plasmid. This novel selection principle extends the applicability of R. eutropha H16 as production platform in biotechnological processes.


Subject(s)
Aldehyde-Lyases/metabolism , Cupriavidus necator/metabolism , Fructose/metabolism , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Bifidobacterium/enzymology , Bifidobacterium/genetics , Carbohydrate Metabolism/genetics , Cupriavidus necator/genetics , Gene Knockout Techniques , Plasmids/genetics , Polymerase Chain Reaction
19.
Cell Signal ; 22(3): 476-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19913094

ABSTRACT

Sphingosine-1-phosphate (S1P) regulates cell growth and survival, migration and adhesion in many cell types. S1P is generated by sphingosine kinases (SphKs), and dephosphorylated by phosphatases or cleaved by S1P lyase. Extracellular S1P activates specific G protein-coupled receptors while intracellular S1P can mobilize Ca2+ from thapsigargin-sensitive stores. Here, we have studied Ca2+ signalling in mouse embryonic fibroblasts (MEFs) deficient in S1P lyase. In these cells, S1P and sphingosine concentrations were elevated about 6-fold and 2-fold, respectively, as measured by liquid chromatography/tandem mass spectrometry. Measurements with fura-2-loaded cells in suspension revealed that resting [Ca2+]i was elevated and agonist-induced [Ca2+]i increases were augmented in S1P lyase-deficient MEFs both in the presence and absence of extracellular Ca2+. Importantly, [Ca2+]i increases and Ca2+ mobilization induced by the SERCA inhibitor, thapsigargin, were augmented, indicating enhanced Ca2+ storage in S1P lyase-deficient MEFs. Measurements with single cells expressing the calmodulin-based Ca2+ sensor, cameleon, revealed that at least two cell types could be distinguished in both MEF cell populations, one with a rapid and transient [Ca2+]i increase and the other with a slower and prolonged [Ca2+]i elevation upon stimulation with thapsigargin. The area under the time course of thapsigargin-induced [Ca2+]i increases, reflecting overall Ca2+ release, was significantly increased by more than 50% in both rapidly and slowly responding S1P lyase-deficient cells. It is concluded that elevated concentrations of S1P and/or sphingosine lead to enhanced Ca2+ storage and elevated basal [Ca2+]i. S1P metabolism thus plays a role not only in acute Ca2+ mobilization but also in long-term regulation of Ca2+ homeostasis.


Subject(s)
Aldehyde-Lyases/metabolism , Calcium/metabolism , Fibroblasts/metabolism , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/genetics , Animals , Calcium Signaling , Calmodulin/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Lysophospholipids/metabolism , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Thapsigargin/pharmacology
20.
Cancer Res ; 69(24): 9346-53, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19934311

ABSTRACT

Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite involved in cancer development through stimulation of cell survival, proliferation, migration, and angiogenesis. Irreversible degradation of S1P is catalyzed by S1P lyase (SPL). The human SGPL1 gene that encodes SPL maps to a region often mutated in cancers. To investigate the effect of SPL deficiency on cell survival and transformation, the susceptibility to anticancer drugs of fibroblasts generated from SPL-deficient mouse embryos (Sgpl1(-/-)) was compared with that of cells from heterozygous (Sgpl1(+/-)) or wild-type (Sgpl1(+/+)) embryos. First, loss of SPL caused resistance to the toxic effects of etoposide and doxorubicin. Interestingly, heterozygosity for the Sgpl1 gene resulted in partial resistance to apoptosis. Secondly, doxorubicin-induced apoptotic signaling was strongly inhibited in Sgpl1(-/-) cells (phosphatidylserine externalization, caspase activation, and cytochrome c release). This was accompanied by a strong increase in Bcl-2 and Bcl-xL protein content. Whereas correction of SPL deficiency in Sgpl1(-/-) cells led to downregulation of antiapoptotic proteins, Bcl-2 and Bcl-xL small interfering RNA-mediated knockdown in SPL-deficient cells resulted in increased sensitivity to doxorubicin, suggesting that Bcl-2 upregulation mediates SPL protective effects. Moreover, SPL deficiency led to increased cell proliferation, anchorage-independent cell growth, and formation of tumors in nude mice. Finally, transcriptomic studies showed that SPL expression is downregulated in human melanoma cell lines. Thus, by affecting S1P metabolism and the expression of Bcl-2 members, the loss of SPL enhances cell resistance to anticancer regimens and results in an increased ability of cells to acquire a transformed phenotype and become malignant.


Subject(s)
Aldehyde-Lyases/deficiency , Proto-Oncogene Proteins c-bcl-2/biosynthesis , bcl-X Protein/biosynthesis , Aldehyde-Lyases/biosynthesis , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Down-Regulation , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Gene Dosage , Humans , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Up-Regulation , bcl-X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...