Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.025
Filter
1.
FASEB J ; 38(14): e23827, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39012295

ABSTRACT

The COVID-19 pandemic, caused by SARS-CoV-2, has had a significant worldwide impact, affecting millions of people. COVID-19 is characterized by a heterogenous clinical phenotype, potentially involving hyperinflammation and prolonged tissue damage, although the exact underlying mechanisms are yet to be fully understood. Sphingolipid metabolites, which govern cell survival and proliferation, have emerged as key players in inflammatory signaling and cytokine responses. Given the complex metabolic pathway of sphingolipids, this study aimed to understand their potential role in the pathogenesis of COVID-19. We conducted a comprehensive examination of sphingolipid modulations across groups classified based on disease severity, incorporating a time-course in serum and urine samples. Several sphingolipids, including sphingosine, lactosylceramide, and hexosylceramide, emerged as promising indicators of COVID-19 severity, as validated by correlation analyses conducted on both serum and urine samples. Other sphingolipids, such as sphingosine 1-phosphate, ceramides, and deoxy-dihydroceramides, decreased in both COVID-19 patients and individuals with non-COVID infectious diseases. This suggests that these sphingolipids are not specifically associated with COVID-19 but rather with pathological conditions caused by infectious diseases. Our analysis of urine samples revealed elevated levels of various sphingolipids, with changes dependent on disease severity, potentially highlighting the acute kidney injury associated with COVID-19. This study illuminates the intricate relationship between disturbed sphingolipid metabolism, COVID-19 severity, and clinical factors. These findings provide valuable insights into the broader landscape of inflammatory diseases.


Subject(s)
COVID-19 , SARS-CoV-2 , Severity of Illness Index , Sphingolipids , COVID-19/metabolism , COVID-19/blood , COVID-19/virology , Humans , Sphingolipids/metabolism , Sphingolipids/blood , Male , Female , Middle Aged , Adult , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Aged , Biomarkers/blood , Biomarkers/metabolism
2.
Sci Rep ; 14(1): 15444, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38965272

ABSTRACT

Tobacco smoking is the main etiological factor of lung cancer (LC), which can also cause metabolome disruption. This study aimed to investigate whether the observed metabolic shift in LC patients was also associated with their smoking status. Untargeted metabolomics profiling was applied for the initial screening of changes in serum metabolic profile between LC and chronic obstructive pulmonary disease (COPD) patients, selected as a non-cancer group. Differences in metabolite profiles between current and former smokers were also tested. Then, targeted metabolomics methods were applied to verify and validate the proposed LC biomarkers. For untargeted metabolomics, a single extraction-dual separation workflow was applied. The samples were analyzed using a liquid chromatograph-high resolution quadrupole time-of-flight mass spectrometer. Next, the selected metabolites were quantified using liquid chromatography-triple-quadrupole mass spectrometry. The acquired data confirmed that patients' stratification based on smoking status impacted the discriminating ability of the identified LC marker candidates. Analyzing a validation set of samples enabled us to determine if the putative LC markers were truly robust. It demonstrated significant differences in the case of four metabolites: allantoin, glutamic acid, succinic acid, and sphingosine-1-phosphate. Our research showed that studying the influence of strong environmental factors, such as tobacco smoking, should be considered in cancer marker research since it reduces the risk of false positives and improves understanding of the metabolite shifts in cancer patients.


Subject(s)
Biomarkers, Tumor , Lung Neoplasms , Metabolomics , Smoking , Humans , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Metabolomics/methods , Biomarkers, Tumor/blood , Male , Female , Middle Aged , Smoking/blood , Smoking/adverse effects , Aged , Sphingosine/analogs & derivatives , Sphingosine/blood , Sphingosine/metabolism , Lysophospholipids/blood , Lysophospholipids/metabolism , Metabolome , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/blood , Chromatography, Liquid/methods , Succinic Acid/blood , Succinic Acid/metabolism , Glutamic Acid/blood , Glutamic Acid/metabolism
3.
Nat Commun ; 15(1): 6067, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025856

ABSTRACT

After recognizing its ligand lipopolysaccharide, Toll-like receptor 4 (TLR4) recruits adaptor proteins to the cell membrane, thereby initiating downstream signaling and triggering inflammation. Whether this recruitment of adaptor proteins is dependent solely on protein-protein interactions is unknown. Here, we report that the sphingolipid sphinganine physically interacts with the adaptor proteins MyD88 and TIRAP and promotes MyD88 recruitment in macrophages. Myeloid cell-specific deficiency in serine palmitoyltransferase long chain base subunit 2, which encodes the key enzyme catalyzing sphingolipid biosynthesis, decreases the membrane recruitment of MyD88 and inhibits inflammatory responses in in vitro bone marrow-derived macrophage and in vivo sepsis models. In a melanoma mouse model, serine palmitoyltransferase long chain base subunit 2 deficiency decreases anti-tumor myeloid cell responses and increases tumor growth. Therefore, sphinganine biosynthesis is required for the initiation of TLR4 signal transduction and serves as a checkpoint for macrophage pattern recognition in sepsis and melanoma mouse models.


Subject(s)
Macrophages , Melanoma , Myeloid Differentiation Factor 88 , Sepsis , Serine C-Palmitoyltransferase , Sphingosine , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Sepsis/metabolism , Macrophages/metabolism , Myeloid Differentiation Factor 88/metabolism , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Melanoma/metabolism , Melanoma/pathology , Melanoma/genetics , Serine C-Palmitoyltransferase/metabolism , Serine C-Palmitoyltransferase/genetics , Humans , Signal Transduction , Disease Models, Animal , Inflammation/metabolism , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , HEK293 Cells , Lipopolysaccharides
4.
Neurobiol Dis ; 199: 106585, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38955289

ABSTRACT

Sphingosine-1 phosphate (S1P) is a lipid metabolite regulating diverse biological processes, including proliferation, differentiation, migration, and apoptosis, highlighting its physiological and therapeutic significance. Current S1P-based therapeutic approaches primarily focus on modulating the downstream signalling via targeting S1P receptors, however, this is challenged by incomplete receptor internalisation. Sphingosine-1-phosphate lyase (SPL) is a highly conserved enzyme that "gatekeeps" the final step of S1P degradation. Cognisant of the complex ligand and receptor interaction and dynamic metabolic networks, the selective modulation of SPL activity presents a new opportunity to regulate S1P biosynthesis and reveal its role in various systems. Over the past decade, an evolving effort has been made to identify new molecules that could block SPL activity in vitro or in vivo. This review focuses on summarising the current understanding of the reported SPL inhibitors identified through various screening approaches, discussing their efficacy in diverse model systems and the possible mechanism of action. Whilst effective modulation of S1P levels via inhibiting SPL is feasible, the specificity of those inhibitors remains inconclusive, presenting a clear challenge for future implications. Yet, none of the currently available SPL inhibitors is proven effective in elevating S1P levels within the central nervous system. This review article embraces future research focusing on investigating selective SPL inhibitors with high potency and possibly blood-brain-barrier permeability, which would aid the development of new S1P-based therapeutics for neurological disorders.


Subject(s)
Aldehyde-Lyases , Lysophospholipids , Sphingosine , Aldehyde-Lyases/metabolism , Aldehyde-Lyases/antagonists & inhibitors , Humans , Animals , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use
5.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-39030066

ABSTRACT

Sphingosine has been previously shown to kill many strains of pathogenic bacteria including Pseudomonas aeruginosa, Staphyloccus aureus, Acinetobacter, and atypical mycobacteria. However, these studies were performed on isolated or extracellular bacteria and it is unknown whether sphingosine also targets intracellular bacteria. Here, we demonstrate that exogenously-added sphingosine directly binds to extracellular P. aeruginosa and S. aureus, but also targets and binds to intracellular bacteria. Intracellular sphingosine and bacteria were identified by sequential immunostainings. We further show that exogenously-added sphingosine also kills intracellular P. aeruginosa and S. aureus using modified gentamycin assays. Intracellular killing of P. aeruginosa and S. aureus by sphingosine is not mediated by improved phagosomal-lysosomal fusion. In summary, our data indicate that sphingosine binds to and most likely also directly kills extra- and intracellular P. aeruginosa and S. aureus.


Subject(s)
Pseudomonas aeruginosa , Sphingosine , Staphylococcus aureus , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine/metabolism , Humans , Anti-Bacterial Agents/pharmacology , Microbial Viability/drug effects , Animals
6.
Methods Mol Biol ; 2816: 35-40, 2024.
Article in English | MEDLINE | ID: mdl-38977586

ABSTRACT

Sphingolipids, including sphingosine and sphinganine, are one of the major classes of lipids. They serve as constituents of cell membranes and lipid rafts and aid in the performance of cell-cell communication and adhesion. Abnormal levels of sphingolipids in the aqueous humor can indicate impaired sphingolipid metabolism and associated ocular pathologies. Sphingolipids can be extracted from the aqueous humor by the methyl-tert-butyl ether (MTBE) lipid extraction method and subsequently analyzed by liquid chromatography-mass spectrometry (LC-MS). This chapter describes a modified protocol for an MTBE lipid extraction from the aqueous humor, followed by analysis with ultrahigh-performance liquid chromatography-mass spectrometry (UHPLC-MS).


Subject(s)
Aqueous Humor , Mass Spectrometry , Sphingosine , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/analysis , Chromatography, High Pressure Liquid/methods , Aqueous Humor/metabolism , Aqueous Humor/chemistry , Mass Spectrometry/methods , Signal Transduction , Humans , Sphingolipids/analysis , Sphingolipids/metabolism , Tandem Mass Spectrometry/methods , Liquid Chromatography-Mass Spectrometry , Methyl Ethers
7.
Methods Mol Biol ; 2816: 25-33, 2024.
Article in English | MEDLINE | ID: mdl-38977585

ABSTRACT

Interconvertible sphingolipid metabolites represent germane constituents of eukaryotic membranes and are vital in the regulation of cellular homeostasis, proliferation, survival, and induction of autophagy. This protocol describes a step-by-step method for extractions of sphingosine and sphinganine from mammalian tissue samples, particularly from the murine optic nerve. These lipids are partitioned into a binary mixture of chloroform and methanol in a modified Bligh and Dyer method. This is followed with reverse phase ultrahigh-performance liquid chromatography fractionation with a C18+ column and subsequent tandem mass spectrometry (UHPLC-MS-MS) analysis of the biological abundance. These free sphingoid bases dissociate to form structurally distinctive carbocation product ions that can be confirmed with annotations of lipidomic databases or in-house fragmentation software.


Subject(s)
Lipidomics , Optic Nerve , Sphingosine , Tandem Mass Spectrometry , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/isolation & purification , Animals , Lipidomics/methods , Tandem Mass Spectrometry/methods , Mice , Optic Nerve/metabolism , Chromatography, High Pressure Liquid/methods , Signal Transduction
8.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928268

ABSTRACT

Human corneal fibrosis can lead to opacity and ultimately partial or complete vision loss. Currently, corneal transplantation is the only treatment for severe corneal fibrosis and comes with the risk of rejection and donor shortages. Sphingolipids (SPLs) are known to modulate fibrosis in various tissues and organs, including the cornea. We previously reported that SPLs are tightly related to both, transforming growth factor beta (TGF-ß) signaling and corneal fibrogenesis. The aim of this study was to investigate the effects of sphingosine-1-phosphate (S1P) and S1P inhibition on specific TGF-ß and SPL family members in corneal fibrosis. Healthy human corneal fibroblasts (HCFs) were isolated and cultured in EMEM + FBS + VitC (construct medium) on 3D transwells for 4 weeks. The following treatments were prepared in a construct medium: 0.1 ng/mL TGF-ß1 (ß1), 1 µM sphingosine-1-phosphate (S1P), and 5 µM Sphingosine kinase inhibitor 2 (I2). Five groups were tested: (1) control (no treatment); rescue groups; (2) ß1/S1P; (3) ß1/I2; prevention groups; (4) S1P/ß1; and (5) I2/ß1. Each treatment was administered for 2 weeks with one treatment and switched to another for 2 weeks. Using Western blot analysis, the 3D constructs were examined for the expression of fibrotic markers, SPL, and TGF-ß signaling pathway members. Scratch assays from 2D cultures were also utilized to evaluate cell migration We observed reduced fibrotic expression and inactivation of latent TGF-ß binding proteins (LTBPs), TGF-ß receptors, Suppressor of Mothers Against Decapentaplegic homologs (SMADs), and SPL signaling following treatment with I2 prevention and rescue compared to S1P prevention and rescue, respectively. Furthermore, we observed increased cell migration following stimulation with I2 prevention and rescue groups, with decreased cell migration following stimulation with S1P prevention and rescue groups after 12 h and 18 h post-scratch. We have demonstrated that I2 treatment reduced fibrosis and modulated the inactivation of LTBPs, TGF-ß receptors, SPLs, and the canonical downstream SMAD pathway. Further investigations are warranted in order to fully uncover the potential of utilizing SphK I2 as a novel therapy for corneal fibrosis.


Subject(s)
Cornea , Fibrosis , Lysophospholipids , Signal Transduction , Sphingosine , Transforming Growth Factor beta , Humans , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/pharmacology , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Cornea/metabolism , Cornea/pathology , Cornea/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Cells, Cultured , Sphingolipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Corneal Diseases/metabolism , Corneal Diseases/pathology , Corneal Diseases/drug therapy
9.
Nutrients ; 16(11)2024 May 26.
Article in English | MEDLINE | ID: mdl-38892556

ABSTRACT

Sphingosine-1-phosphate lyase (SPL) resides at the endpoint of the sphingolipid metabolic pathway, catalyzing the irreversible breakdown of sphingosine-1-phosphate. Depletion of SPL precipitates compromised muscle morphology and function; nevertheless, the precise mechanistic underpinnings remain elusive. Here, we elucidate a model of SPL functional deficiency in Caenorhabditis elegans using spl-1 RNA interference. Within these SPL-deficient nematodes, we observed diminished motility and perturbed muscle fiber organization, correlated with the accumulation of sphingoid bases, their phosphorylated forms, and ceramides (collectively referred to as the "sphingolipid rheostat"). The disturbance in mitochondrial morphology was also notable, as SPL functional loss resulted in heightened levels of reactive oxygen species. Remarkably, the administration of the antioxidant N-acetylcysteine (NAC) ameliorates locomotor impairment and rectifies muscle fiber disarray, underscoring its therapeutic promise for ceramide-accumulation-related muscle disorders. Our findings emphasize the pivotal role of SPL in preserving muscle integrity and advocate for exploring antioxidant interventions, such as NAC supplementation, as prospective therapeutic strategies for addressing muscle function decline associated with sphingolipid/ceramide metabolism disruption.


Subject(s)
Acetylcysteine , Aldehyde-Lyases , Caenorhabditis elegans , Ceramides , Sphingolipids , Animals , Caenorhabditis elegans/drug effects , Acetylcysteine/pharmacology , Ceramides/metabolism , Aldehyde-Lyases/metabolism , Sphingolipids/metabolism , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Muscles/drug effects , Muscles/metabolism , RNA Interference , Sphingosine/analogs & derivatives , Sphingosine/metabolism
10.
Nat Commun ; 15(1): 4755, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834568

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a severe type of the non-alcoholic fatty liver disease (NAFLD). NASH is a growing global health concern due to its increasing morbidity, lack of well-defined biomarkers and lack of clinically effective treatments. Using metabolomic analysis, the most significantly changed active lipid sphingosine d18:1 [So(d18:1)] is selected from NASH patients. So(d18:1) inhibits macrophage HIF-2α as a direct inhibitor and promotes the inflammatory factors secretion. Male macrophage-specific HIF-2α knockout and overexpression mice verified the protective effect of HIF-2α on NASH progression. Importantly, the HIF-2α stabilizer FG-4592 alleviates liver inflammation and fibrosis in NASH, which indicated that macrophage HIF-2α is a potential drug target for NASH treatment. Overall, this study confirms that So(d18:1) promotes NASH and clarifies that So(d18:1) inhibits the transcriptional activity of HIF-2α in liver macrophages by suppressing the interaction of HIF-2α with ARNT, suggesting that macrophage HIF-2α may be a potential target for the treatment of NASH.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Macrophages , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Sphingosine , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Male , Macrophages/metabolism , Macrophages/drug effects , Humans , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Liver/metabolism , Liver/pathology , Liver/drug effects , Mice, Inbred C57BL , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Disease Models, Animal
11.
Arterioscler Thromb Vasc Biol ; 44(8): 1725-1736, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38899471

ABSTRACT

Ceramides, a group of biologically active sphingolipids, have been described as the new cholesterol given strong evidence linking high plasma ceramide with endothelial damage, risk for early adverse cardiovascular events, and development of cardiometabolic disease. This relationship has sparked great interest in investigating therapeutic targets with the goal of suppressing ceramide formation. However, the growing data challenge this paradigm of ceramide as solely eliciting detrimental effects to the cardiovascular system. Studies show that ceramides are necessary for maintaining proper endothelial redox states, mechanosensation, and membrane integrity. Recent work in preclinical models and isolated human microvessels highlights that the loss of ceramide formation can in fact propagate vascular endothelial dysfunction. Here, we delve into these conflicting findings to evaluate how ceramide may be capable of exerting both beneficial and damaging effects within the vascular endothelium. We propose a unifying theory that while basal levels of ceramide in response to physiological stimuli are required for the production of vasoprotective metabolites such as S1P (sphingosine-1-phosphate), the chronic accumulation of ceramide can promote activation of pro-oxidative stress pathways in endothelial cells. Clinically, the evidence discussed here highlights the potential challenges associated with therapeutic suppression of ceramide formation as a means of reducing cardiovascular disease risk.


Subject(s)
Cardiovascular Diseases , Ceramides , Endothelium, Vascular , Oxidative Stress , Sphingosine , Ceramides/metabolism , Humans , Endothelium, Vascular/metabolism , Animals , Cardiovascular Diseases/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Signal Transduction
12.
Mol Metab ; 86: 101971, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38925249

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality, and its incidence is increasing due to endemic obesity. HCC is sexually dimorphic in both humans and rodents with higher incidence in males, although the mechanisms contributing to these correlations remain unclear. Here, we examined the role of sphingosine kinase 2 (SphK2), the enzyme that regulates the balance of bioactive sphingolipid metabolites, sphingosine-1-phosphate (S1P) and ceramide, in gender specific MASH-driven HCC. METHODS: Male and female mice were fed a high fat diet with sugar water, a clinically relevant model that recapitulates MASH-driven HCC in humans followed by physiological, biochemical cellular and molecular analyses. In addition, correlations with increased risk of HCC recurrence were determined in patients. RESULTS: Here, we report that deletion of SphK2 protects both male and female mice from Western diet-induced weight gain and metabolic dysfunction without affecting hepatic lipid accumulation or fibrosis. However, SphK2 deficiency decreases chronic diet-induced hepatocyte proliferation in males but increases it in females. Remarkably, SphK2 deficiency reverses the sexual dimorphism of HCC, as SphK2-/- male mice are protected whereas the females develop liver cancer. Only in male mice, chronic western diet induced accumulation of the autophagy receptor p62 and its downstream mediators, the antioxidant response target NQO1, and the oncogene c-Myc. SphK2 deletion repressed these known drivers of HCC development. Moreover, high p62 expression correlates with poor survival in male HCC patients but not in females. In hepatocytes, lipotoxicity-induced p62 accumulation is regulated by sex hormones and prevented by SphK2 deletion. Importantly, high SphK2 expression in male but not female HCC patients is associated with a more aggressive HCC differentiation status and increased risk of cancer recurrence. CONCLUSIONS: This work identifies SphK2 as a potential regulator of HCC sexual dimorphism and suggests SphK2 inhibitors now in clinical trials could have opposing, gender-specific effects in patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Phosphotransferases (Alcohol Group Acceptor) , Sex Characteristics , Animals , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Male , Female , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Mice , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Humans , Mice, Inbred C57BL , Diet, High-Fat/adverse effects , Mice, Knockout , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Cell Proliferation
13.
Chem Biol Interact ; 398: 111085, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38823539

ABSTRACT

Sepsis-induced acute lung injury (SALI) is the common complication of sepsis, resulting in high incidence and mortality rates. The primary pathogenesis of SALI is the interplay between acute inflammation and endothelial barrier damage. Studies have shown that kaempferol (KPF) has anti-sepsis properties. Sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P) signaling pathway's significance in acute lung damage and S1P receptor 1 (S1PR1) agonists potential in myosin light chain 2 (MLC2) phosphorylation are documented. Whether KPF can regulate the SphK1/S1P/S1PR1/MLC2 signaling pathway to protect the lung endothelial barrier remains unclear. This study investigates the KPF's therapeutic effects and molecular mechanisms in repairing endothelial cell barrier damage in both LPS-induced sepsis mice and human umbilical vein endothelial cells (HUVECs). KPF significantly reduced lung tissue damage and showed anti-inflammatory effects by decreasing IL-6 and TNF-α synthesis in the sepsis mice model. Further, KPF administration can reduce the high permeability of the LPS-induced endothelial cell barrier and alleviate lung endothelial cell barrier injury. Mechanistic studies showed that KPF pretreatment can suppress MLC2 hyperphosphorylation and decrease SphK1, S1P, and S1PR1 levels. The SphK1/S1P/S1PR1/MLC2 signaling pathway controls the downstream proteins linked to endothelial barrier damage, and the Western blot (WB) showed that KPF raised the protein levels. These proteins include zonula occludens (ZO)-1, vascular endothelial (VE)-cadherin and Occludin. The present work revealed that in mice exhibiting sepsis triggered by LPS, KPF strengthened the endothelial barrier and reduced the inflammatory response. The SphK1/S1P/S1PR1/MLC2 pathway's modulation is the mechanism underlying this impact.


Subject(s)
Acute Lung Injury , Cardiac Myosins , Human Umbilical Vein Endothelial Cells , Kaempferols , Lung , Lysophospholipids , Mice, Inbred C57BL , Myosin Light Chains , Sepsis , Signal Transduction , Sphingosine , Animals , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Humans , Myosin Light Chains/metabolism , Signal Transduction/drug effects , Mice , Lysophospholipids/metabolism , Kaempferols/pharmacology , Kaempferols/therapeutic use , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/pharmacology , Male , Human Umbilical Vein Endothelial Cells/metabolism , Cardiac Myosins/metabolism , Lung/pathology , Lung/drug effects , Lung/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Lipopolysaccharides , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Receptors, Lysosphingolipid/metabolism , Interleukin-6/metabolism , Sphingosine-1-Phosphate Receptors/metabolism
14.
Exp Neurol ; 379: 114864, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38866101

ABSTRACT

Platelet-derived growth factor receptor ß positive (PDGFRß+) pericytes detach from the microvascular wall and migrate into the injury center following spinal cord injury (SCI), which has been widely regarded as the main source of fibrotic scar, but the mechanism of migration and fibroblast transition remains elusive. Here we show the associated spatiotemporal distribution between microglia and pericytes at three and seven days post-injury (dpi). The increased expression of Sphingosine kinase-1 (SPHK1) in microglia significantly raised the concentration of Sphingosine-1-phosphate (S1P) in the spinal cord, which promotes migration and fibroblast transition of pericyte. In vitro experiments, we found the elevated Sphingosine 1-phosphate receptor 3 (S1P3), the S1P/S1PR3 axis inhibited the phosphorylation of YAP and promoted its nuclear translocation, which contributed to the formation of alpha-smooth muscle actin (α-SMA) and collagen type I (COL1) protein, This process can be blocked by an S1P3 specific inhibitor TY52156 in vitro. The S1P/S1P3/YAP pathway might be a potential target for treatment in SCI.


Subject(s)
Cell Movement , Fibroblasts , Lysophospholipids , Microglia , Pericytes , Signal Transduction , Sphingosine-1-Phosphate Receptors , Sphingosine , Spinal Cord Injuries , YAP-Signaling Proteins , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Signal Transduction/physiology , Lysophospholipids/metabolism , Animals , Cell Movement/physiology , Pericytes/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Microglia/metabolism , YAP-Signaling Proteins/metabolism , Rats , Fibroblasts/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Rats, Sprague-Dawley , Adaptor Proteins, Signal Transducing/metabolism , Receptors, Lysosphingolipid/metabolism , Cells, Cultured
15.
Cell Signal ; 121: 111252, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38852936

ABSTRACT

BACKGROUND AND AIMS: S1P is an important factor regulating the function of the vascular endothelial barrier. SphK1 is an important limiting enzyme for the synthesis of S1P. However, the role of the SphK1/S1P-mediated vascular endothelial barrier function in atherosclerosis has not been fully revealed. This study explored the roles and mechanisms of SphK1 on atherosclerosis in vivo and in vitro. METHODS: In vivo, ApoE-/- and SphK1-/-ApoE-/- mice were fed a high-fat diet to induce atherosclerosis. In vitro, ox-LDL induced HUVECs to establish a cell model. Aortic histological changes were measured by H&E staining, Oil Red O staining, EVG staining, Sirius scarlet staining, immunofluorescence, and Evans Blue Assay. Western blotting was performed to explore the specific mechanism. RESULTS: We validated that deficiency of SphK1 resulted in a marked amelioration of atherosclerosis, as indicated by the decreased lipid accumulation, inflammatory factors, oxidative stress, aortic plaque area, inflammatory factor infiltration, VCAM-1 expression, and vascular endothelial permeability. Moreover, deficiency of SphK1 downregulated the expression of aortic S1PR3, Rhoa, ROCK, and F-actin. The results of administration with the SphK1 inhibitor PF-543 and the S1PR3 inhibitor VPC23019 in vitro further confirmed the conclusion that deficiency of SphK1 reduced S1P level and S1PR3 protein expression, inhibited Rhoa/ROCK signaling pathway, regulated protein expression of F-actin, improved vascular endothelial dysfunction and permeability, and exerted anti-atherosclerotic effects. CONCLUSIONS: This study revealed that deficiency of SphK1 relieved vascular endothelial barrier function in atherosclerosis mice via SphK1/S1P/S1PR signaling pathway.


Subject(s)
Atherosclerosis , Human Umbilical Vein Endothelial Cells , Phosphotransferases (Alcohol Group Acceptor) , Signal Transduction , Sphingosine-1-Phosphate Receptors , rho-Associated Kinases , rhoA GTP-Binding Protein , Animals , Humans , Male , Mice , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Diet, High-Fat , Human Umbilical Vein Endothelial Cells/metabolism , Lysophospholipids/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptors, Lysosphingolipid/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/metabolism
16.
Lipids Health Dis ; 23(1): 200, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937745

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) causes neuroinflammation and can lead to long-term neurological dysfunction, even in cases of mild TBI (mTBI). Despite the substantial burden of this disease, the management of TBI is precluded by an incomplete understanding of its cellular mechanisms. Sphingolipids (SPL) and their metabolites have emerged as key orchestrators of biological processes related to tissue injury, neuroinflammation, and inflammation resolution. No study so far has investigated comprehensive sphingolipid profile changes immediately following TBI in animal models or human cases. In this study, sphingolipid metabolite composition was examined during the acute phases in brain tissue and plasma of mice following mTBI. METHODS: Wildtype mice were exposed to air-blast-mediated mTBI, with blast exposure set at 50-psi on the left cranium and 0-psi designated as Sham. Sphingolipid profile was analyzed in brain tissue and plasma during the acute phases of 1, 3, and 7 days post-TBI via liquid-chromatography-mass spectrometry. Simultaneously, gene expression of sphingolipid metabolic markers within brain tissue was analyzed using quantitative reverse transcription-polymerase chain reaction. Significance (P-values) was determined by non-parametric t-test (Mann-Whitney test) and by Tukey's correction for multiple comparisons. RESULTS: In post-TBI brain tissue, there was a significant elevation of 1) acid sphingomyelinase (aSMase) at 1- and 3-days, 2) neutral sphingomyelinase (nSMase) at 7-days, 3) ceramide-1-phosphate levels at 1 day, and 4) monohexosylceramide (MHC) and sphingosine at 7-days. Among individual species, the study found an increase in C18:0 and a decrease in C24:1 ceramides (Cer) at 1 day; an increase in C20:0 MHC at 3 days; decrease in MHC C18:0 and increase in MHC C24:1, sphingomyelins (SM) C18:0, and C24:0 at 7 days. Moreover, many sphingolipid metabolic genes were elevated at 1 day, followed by a reduction at 3 days and an absence at 7-days post-TBI. In post-TBI plasma, there was 1) a significant reduction in Cer and MHC C22:0, and an increase in MHC C16:0 at 1 day; 2) a very significant increase in long-chain Cer C24:1 accompanied by significant decreases in Cer C24:0 and C22:0 in MHC and SM at 3 days; and 3) a significant increase of C22:0 in all classes of SPL (Cer, MHC and SM) as well as a decrease in Cer C24:1, MHC C24:1 and MHC C24:0 at 7 days. CONCLUSIONS: Alterations in sphingolipid metabolite composition, particularly sphingomyelinases and short-chain ceramides, may contribute to the induction and regulation of neuroinflammatory events in the early stages of TBI, suggesting potential targets for novel diagnostic, prognostic, and therapeutic strategies in the future.


Subject(s)
Brain , Ceramides , Sphingolipids , Sphingomyelin Phosphodiesterase , Sphingosine , Animals , Mice , Sphingolipids/blood , Sphingolipids/metabolism , Brain/metabolism , Brain/pathology , Ceramides/blood , Ceramides/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelin Phosphodiesterase/blood , Sphingomyelin Phosphodiesterase/genetics , Sphingosine/analogs & derivatives , Sphingosine/blood , Sphingosine/metabolism , Disease Models, Animal , Male , Sphingomyelins/blood , Sphingomyelins/metabolism , Brain Concussion/blood , Brain Concussion/metabolism , Mice, Inbred C57BL , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/pathology , Lysophospholipids/blood , Lysophospholipids/metabolism
17.
JCI Insight ; 9(11)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38855867

ABSTRACT

In rheumatoid arthritis, inflammatory mediators extravasate from blood into joints via gaps between endothelial cells (ECs), but the contribution of ECs is not known. Sphingosine 1-phosphate receptor 1 (S1PR1), widely expressed on ECs, maintains the vascular barrier. Here, we assessed the contribution of vascular integrity and EC S1PR1 signaling to joint damage in mice exposed to serum-induced arthritis (SIA). EC-specific deletion of S1PR1 or pharmacological blockade of S1PR1 promoted vascular leak and amplified SIA, whereas overexpression of EC S1PR1 or treatment with an S1PR1 agonist delayed SIA. Blockade of EC S1PR1 induced membrane metalloproteinase-dependent cleavage of vascular endothelial cadherin (VE-cadherin), a principal adhesion molecule that maintains EC junctional integrity. We identified a disintegrin and a metalloproteinase domain 10 (ADAM10) as the principal VE-cadherin "sheddase." Mice expressing a stabilized VE-cadherin construct had decreased extravascular VE-cadherin and vascular leakage in response to S1PR1 blockade, and they were protected from SIA. Importantly, patients with active rheumatoid arthritis had decreased circulating S1P and microvascular expression of S1PR1, suggesting a dysregulated S1P/S1PR1 axis favoring vascular permeability and vulnerability. We present a model in which EC S1PR1 signaling maintains homeostatic vascular barrier function by limiting VE-cadherin shedding mediated by ADAM10 and suggest this signaling axis as a therapeutic target in inflammatory arthritis.


Subject(s)
ADAM10 Protein , Antigens, CD , Arthritis, Experimental , Arthritis, Rheumatoid , Cadherins , Endothelial Cells , Sphingosine-1-Phosphate Receptors , Animals , Cadherins/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Mice , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Antigens, CD/metabolism , Antigens, CD/genetics , Endothelial Cells/metabolism , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/genetics , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/metabolism , Signal Transduction , Mice, Knockout , Membrane Proteins/metabolism , Membrane Proteins/genetics , Male , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Capillary Permeability , Female
18.
J Mol Med (Berl) ; 102(7): 947-959, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38780771

ABSTRACT

Pancreas ductal adenocarcinoma belongs to the most common cancers, but also to the tumors with the poorest prognosis. Here, we pharmacologically targeted a mitochondrial potassium channel, namely mitochondrial Kv1.3, and investigated the role of sphingolipids and mutated Kirsten Rat Sarcoma Virus (KRAS) in Kv1.3-mediated cell death. We demonstrate that inhibition of Kv1.3 using the Kv1.3-inhibitor PAPTP results in an increase of sphingosine and superoxide in membranes and/or membranes associated with mitochondria, which is enhanced by KRAS mutation. The effect of PAPTP on sphingosine and mitochondrial superoxide formation as well as cell death is prevented by sh-RNA-mediated downregulation of Kv1.3. Induction of sphingosine in human pancreas cancer cells by PAPTP is mediated by activation of sphingosine-1-phosphate phosphatase and prevented by an inhibitor of sphingosine-1-phosphate phosphatase. A rapid depolarization of isolated mitochondria is triggered by binding of sphingosine to cardiolipin, which is neutralized by addition of exogenous cardiolipin. The significance of these findings is indicated by treatment of mutated KRAS-harboring metastasized pancreas cancer with PAPTP in combination with ABC294640, a blocker of sphingosine kinases. This treatment results in increased formation of sphingosine and death of pancreas cancer cells in vitro and, most importantly, prolongs in vivo survival of mice challenged with metastatic pancreas cancer. KEY MESSAGES: Pancreatic ductal adenocarcinoma (PDAC) is a common tumor with poor prognosis. The mitochondrial Kv1.3 ion channel blocker induced mitochondrial sphingosine. Sphingosine binds to cardiolipin thereby mediating mitochondrial depolarization. Sphingosine is formed by a PAPTP-mediated activation of S1P-Phosphatase. Inhibition of sphingosine-consumption amplifies PAPTP effects on PDAC in vivo.


Subject(s)
Mitochondria , Pancreatic Neoplasms , Sphingosine , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Kv1.3 Potassium Channel/metabolism , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/antagonists & inhibitors , Mice , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Cell Death/drug effects , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics
19.
J Lipid Res ; 65(6): 100557, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719152

ABSTRACT

Dietary sphingomyelin (SM) has been reported to favorably modulate postprandial lipemia. Mechanisms underlying these beneficial effects on cardiovascular risk markers are not fully elucidated. Rodent studies showed that tritiated SM was hydrolyzed in the intestinal lumen into ceramides (Cer) and further to sphingosine (SPH) and fatty acids (FA) that were absorbed by the intestine. Our objective was to investigate the uptake and metabolism of SPH and/or tricosanoic acid (C23:0), the main FA of milk SM, as well as lipid secretion in Caco-2/TC7 cells cultured on semipermeable inserts. Mixed micelles (MM) consisting of different digested lipids and taurocholate were prepared without or with SPH, SPH and C23:0 (SPH+C23:0), or C23:0. Triglycerides (TG) were quantified in the basolateral medium, and sphingolipids were analyzed by tandem mass spectrometry. TG secretion increased 11-fold in all MM-incubated cells compared with lipid-free medium. Apical supply of SPH-enriched MM led to increased concentrations of total Cer in cells, and coaddition of C23:0 in SPH-enriched MM led to a preferential increase of C23:0 Cer and C23:0 SM. Complementary experiments using deuterated SPH demonstrated that SPH-d9 was partly converted to sphingosine-1-phosphate-d9, Cer-d9, and SM-d9 within cells incubated with SPH-enriched MM. A few Cer-d9 (2% of added SPH-d9) was recovered in the basolateral medium of (MM+SPH)-incubated cells, especially C23:0 Cer-d9 in (MM+SPH+C23:0)-enriched cells. In conclusion, present results indicate that MM enriched with (SPH+C23:0), such as found in postprandial micelles formed after milk SM ingestion, directly impacts sphingolipid endogenous metabolism in enterocytes, resulting in the secretion of TG-rich particles enriched with C23:0 Cer.


Subject(s)
Ceramides , Intestinal Absorption , Sphingosine , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Humans , Ceramides/metabolism , Caco-2 Cells , Micelles , Triglycerides/metabolism , Isotope Labeling , Animals
20.
PLoS One ; 19(5): e0303296, 2024.
Article in English | MEDLINE | ID: mdl-38753743

ABSTRACT

AIM: Metabolic dysfunction-associated steatohepatitis (MASH) is one of the most prevalent liver diseases and is characterized by steatosis and the accumulation of bioactive lipids. This study aims to understand the specific lipid species responsible for the progression of liver fibrosis in MASH. METHODS: Changes in bioactive lipid levels were examined in the livers of MASH mice fed a choline-deficient diet (CDD). Additionally, sphingosine kinase (SphK)1 mRNA, which generates sphingosine 1 phosphate (S1P), was examined in the livers of patients with MASH. RESULTS: CDD induced MASH and liver fibrosis were accompanied by elevated levels of S1P and increased expression of SphK1 in capillarized liver sinusoidal endothelial cells (LSECs) in mice. SphK1 mRNA also increased in the livers of patients with MASH. Treatment of primary cultured mouse hepatic stellate cells (HSCs) with S1P stimulated their activation, which was mitigated by the S1P receptor (S1PR)2 inhibitor, JTE013. The inhibition of S1PR2 or its knockout in mice suppressed liver fibrosis without reducing steatosis or hepatocellular damage. CONCLUSION: S1P level is increased in MASH livers and contributes to liver fibrosis via S1PR2.


Subject(s)
Fatty Liver , Hepatic Stellate Cells , Liver Cirrhosis , Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor) , Sphingosine-1-Phosphate Receptors , Sphingosine , Animals , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/etiology , Mice , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Humans , Sphingosine-1-Phosphate Receptors/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Male , Mice, Knockout , Mice, Inbred C57BL , Liver/metabolism , Liver/pathology , Choline Deficiency/complications , Choline Deficiency/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/genetics , Pyrazoles , Pyridines
SELECTION OF CITATIONS
SEARCH DETAIL