Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.862
Filter
1.
Cell Commun Signal ; 22(1): 304, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831326

ABSTRACT

Elevated concentrations of palmitate in serum of obese individuals can impair endothelial function, contributing to development of cardiovascular disease. Although several molecular mechanisms of palmitate-induced endothelial dysfunction have been proposed, there is no consensus on what signaling event is the initial trigger of detrimental palmitate effects. Here we report that inhibitors of ER stress or ceramid synthesis can rescue palmitate-induced autophagy impairment in macro- and microvascular endothelial cells. Furthermore, palmitate-induced cholesterol synthesis was reverted using these inhibitors. Similar to cell culture data, autophagy markers were increased in serum of obese individuals. Subsequent lipidomic analysis revealed that palmitate changed the composition of membrane phospholipids in endothelial cells and that these effects were not reverted upon application of above-mentioned inhibitors. However, ER stress inhibition in palmitate-treated cells enhanced the synthesis of trilglycerides and restored ceramide levels to control condition. Our results suggest that palmitate induces ER-stress presumably by shift in membrane architecture, leading to impaired synthesis of triglycerides and enhanced production of ceramides and cholesterol, which altogether enhances lipotoxicity of palmitate in endothelial cells.


Subject(s)
Endoplasmic Reticulum Stress , Endothelial Cells , Endoplasmic Reticulum Stress/drug effects , Humans , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Autophagy/drug effects , Triglycerides/metabolism , Cholesterol/metabolism , Palmitates/pharmacology , Ceramides/metabolism
2.
PLoS One ; 19(5): e0303569, 2024.
Article in English | MEDLINE | ID: mdl-38743756

ABSTRACT

There is a phenotype of obese individuals termed metabolically healthy obese that present a reduced cardiometabolic risk. This phenotype offers a valuable model for investigating the mechanisms connecting obesity and metabolic alterations such as Type 2 Diabetes Mellitus (T2DM). Previously, in an untargeted metabolomics analysis in a cohort of morbidly obese women, we observed a different lipid metabolite pattern between metabolically healthy morbid obese individuals and those with associated T2DM. To validate these findings, we have performed a complementary study of lipidomics. In this study, we assessed a liquid chromatography coupled to a mass spectrometer untargeted lipidomic analysis on serum samples from 209 women, 73 normal-weight women (control group) and 136 morbid obese women. From those, 65 metabolically healthy morbid obese and 71 with associated T2DM. In this work, we find elevated levels of ceramides, sphingomyelins, diacyl and triacylglycerols, fatty acids, and phosphoethanolamines in morbid obese vs normal weight. Conversely, decreased levels of acylcarnitines, bile acids, lyso-phosphatidylcholines, phosphatidylcholines (PC), phosphatidylinositols, and phosphoethanolamine PE (O-38:4) were noted. Furthermore, comparing morbid obese women with T2DM vs metabolically healthy MO, a distinct lipid profile emerged, featuring increased levels of metabolites: deoxycholic acid, diacylglycerol DG (36:2), triacylglycerols, phosphatidylcholines, phosphoethanolamines, phosphatidylinositols, and lyso-phosphatidylinositol LPI (16:0). To conclude, analysing both comparatives, we observed decreased levels of deoxycholic acid, PC (34:3), and PE (O-38:4) in morbid obese women vs normal-weight. Conversely, we found elevated levels of these lipids in morbid obese women with T2DM vs metabolically healthy MO. These profiles of metabolites could be explored for the research as potential markers of metabolic risk of T2DM in morbid obese women.


Subject(s)
Diabetes Mellitus, Type 2 , Lipidomics , Obesity, Morbid , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Female , Obesity, Morbid/blood , Obesity, Morbid/metabolism , Obesity, Morbid/complications , Lipidomics/methods , Middle Aged , Adult , Lipids/blood , Metabolomics/methods , Case-Control Studies , Triglycerides/blood , Sphingomyelins/blood , Sphingomyelins/metabolism , Ceramides/blood , Ceramides/metabolism , Lipid Metabolism
4.
Sci Total Environ ; 931: 172993, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38719056

ABSTRACT

Inflammation is a key mechanism underlying the adverse health effects of exposure to fine particulate matter (PM2.5). Bioactive lipids in the arachidonic acid (ARA) pathway are important in the regulation of inflammation and are reportedly altered by PM2.5 exposure. Ceramide-1-phosphate (C1P), a class of sphingolipids, is required to initiate ARA metabolism. We examined the role of C1P in the alteration of ARA metabolism after PM2.5 exposure and explored whether changes in the ARA pathway promoted systemic inflammation based on a panel study involving 112 older adults in Beijing, China. Ambient PM2.5 levels were continuously monitored at a fixed station from 2013 to 2015. Serum cytokine levels were measured to assess systemic inflammation. Multiple bioactive lipids in the ARA pathway and three subtypes of C1P were quantified in blood samples. Mediation analyses were performed to test the hypotheses. We observed that PM2.5 exposure was positively associated with inflammatory cytokines and the three subtypes of C1P. Mediation analyses showed that C1P significantly mediated the associations of ARA and 5, 6-dihydroxyeicosatrienoic acid (5, 6-DHET), an ARA metabolite, with PM2.5 exposure. ARA, 5, 6-DHET, and leukotriene B4 mediated systemic inflammatory response to PM2.5 exposure. For example, C1P C16:0 (a subtype of C1P) mediated a 12.9 % (95 % confidence interval: 3.7 %, 32.5 %) increase in ARA associated with 3-day moving average PM2.5 exposure, and ARA mediated a 27.1 % (7.8 %, 61.2 %) change in interleukin-8 associated with 7-day moving average PM2.5 exposure. Our study indicates that bioactive lipids in the ARA and sphingolipid metabolic pathways may mediate systemic inflammation after PM2.5 exposure.


Subject(s)
Air Pollutants , Inflammation , Particulate Matter , Particulate Matter/toxicity , Humans , Inflammation/chemically induced , Air Pollutants/toxicity , Male , Environmental Exposure/statistics & numerical data , Environmental Exposure/adverse effects , Beijing , Female , Aged , Cytokines/blood , Cytokines/metabolism , Arachidonic Acid/metabolism , Ceramides , Middle Aged , Lipids/blood
5.
J Diabetes Investig ; 15(6): 684-692, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38713732

ABSTRACT

AIMS: The aim of this study was to better understand how the chemotherapy drug doxorubicin contributes to the development of ß-cell dysfunction and to explore its relationship with mitochondrial aldehyde dehydrogenase-2 (ALDH2). MATERIALS AND METHODS: In order to investigate this hypothesis, doxorubicin was administered to INS-1 cells, a rat insulinoma cell line, either with or without several target protein activators and inhibitors. ALDH2 activity was detected with a commercial kit and protein levels were determined with western blot. Mitochondrial ROS, membrane potential, and lipid ROS were determined by commercial fluorescent probes. The cell viability was measured by CCK-assay. RESULTS: Exposure of INS-1 cells to doxorubicin decreased active insulin signaling resulting in elevated ALDH2 degradation, compared with control cells by the induction of acid sphingomyelinase mediated ceramide induction. Further, ceramide induction potentiated doxorubicin induced mitochondrial dysfunction. Treatment with the ALDH2 agonist, ALDA1, blocked doxorubicin-induced acid sphingomyelinase activation which significantly blocked ceramide induction and mitochondrial dysfunction mediated cell death. Treatment with the ALDH2 agonist, ALDA1, stimulated casein kinase-2 (CK2) mediated insulin signaling activation. CK2 silencing neutralized the function of ALDH2 in the doxorubicin treated INS-1 cells. CONCLUSIONS: Mitochondrial ALDH2 activation could inhibit the progression of doxorubicin induced pancreatic ß-cell dysfunction by inhibiting the acid sphingomyelinase induction of ceramide, by regulating the activation of CK2 signaling. Our research lays the foundation of ALDH2 activation as a therapeutic target for the precise treatment of chemotherapy drug induced ß-cell dysfunction.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial , Apoptosis , Casein Kinase II , Cell Survival , Doxorubicin , Insulin-Secreting Cells , Mitochondria , Signal Transduction , Doxorubicin/pharmacology , Rats , Animals , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Apoptosis/drug effects , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Aldehyde Dehydrogenase, Mitochondrial/genetics , Signal Transduction/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Cell Survival/drug effects , Casein Kinase II/metabolism , Casein Kinase II/antagonists & inhibitors , Cell Line, Tumor , Ceramides/metabolism , Reactive Oxygen Species/metabolism , Antibiotics, Antineoplastic/pharmacology
6.
PLoS One ; 19(5): e0300844, 2024.
Article in English | MEDLINE | ID: mdl-38809909

ABSTRACT

BACKGROUND: A randomized clinical trial to evaluate the effect of a Mediterranean-style diet on vascular health indices such as endothelial function indices, serum lipid and ceramide plasma and some adipokine serum levels. We recruited all consecutive patients at high risk of cardiovascular diseases admitted to the Internal Medicine and Stroke Care ward at the University Hospital of Palermo between September 2017 and December 2020. MATERIALS AND METHODS: The enrolled subjects, after the evaluation of the degree of adherence to a dietary regimen of the Mediterranean-style diet, were randomised to a Mediterranean Diet (group A) assessing the adherence to a Mediterranean-style diet at each follow up visit (every three months) for the entire duration of the study (twelve months) and to a Low-fat diet (group B) with a dietary "counselling" starting every three months for the entire duration of the study (twelve months).The aims of the study were to evaluate: the effects of adherence to Mediterranean Diet on some surrogate markers of vascular damage, such as endothelial function measured by means of the reactive hyperaemia index (RHI) and augmentation index (AIX), at the 6-(T1) and 12-month (T2) follow-ups; the effects of adherence to Mediterranean Diet on the lipidaemic profile and on serum levels of ceramides at T1 and T2 follow-ups; the effects of adherence to Mediterranean Diet on serum levels of visfatin, adiponectin and resistin at the 6- and 12-month follow-ups. RESULTS: A total of 101 patients were randomised to a Mediterranean Diet style and 52 control subjects were randomised to a low-fat diet with a dietary "counselling". At the six-month follow-up (T1), subjects in the Mediterranean Diet group showed significantly lower mean serum total cholesterol levels, and significantly higher increase in reactive hyperaemia index (RHI) values compared to the low-fat diet group. Patients in the Mediterranean Diet group also showed lower serum levels of resistin and visfatin at the six-month follow-up compared to the control group, as well as higher values ​​of adiponectin, lower values of C24:0, higher values of C22:0 and higher values of the C24:0/C16:0 ratio. At the twelve-month follow-up (T2), subjects in the Mediterranean Diet group showed lower serum total cholesterol levels and lower serum LDL cholesterol levels than those in the control group. At the twelve-month follow-up, we also observed a further significant increase in the mean RHI in the Mediterranean Diet group, lower serum levels of resistin and visfatin, lower values of C24:0 and of C:18:0,and higher values of the C24:0/C16:0 ratio. DISCUSSION: The findings of our current study offer a further possible explanation with regard to the beneficial effects of a higher degree of adherence to a Mediterranean-style diet on multiple cardiovascular risk factors and the underlying mechanisms of atherosclerosis. Moreover, these findings provide an additional plausible interpretation of the results from observational and cohort studies linking high adherence to a Mediterranean-style diet with lower total mortality and a decrease in cardiovascular events and cardiovascular mortality. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04873167. https://classic.clinicaltrials.gov/ct2/show/NCT04873167.


Subject(s)
Adipokines , Ceramides , Diet, Mediterranean , Humans , Male , Female , Middle Aged , Ceramides/blood , Adipokines/blood , Aged , Cardiovascular Diseases/blood , Cardiovascular Diseases/prevention & control , Resistin/blood , Diet, Fat-Restricted , Biomarkers/blood , Nicotinamide Phosphoribosyltransferase/blood
8.
Bioorg Med Chem Lett ; 107: 129792, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38734389

ABSTRACT

Ceramide 1-phosphate (C1P) is a lipid mediator that specifically binds and activates cytosolic phospholipase A2α (cPLA2α). To elucidate the structure-activity relationship of the affinity of C1P for cPLA2α in lipid environments, we prepared a series of C1P analogs containing structural modifications in the hydrophilic parts and subjected them to surface plasmon resonance (SPR). The results suggested the presence of a specific binding site for cPLA2α on the amide, 3-OH and phosphate groups in C1P structure. Especially, dihydro-C1P exhibited enhanced affinity for cPLA2α, suggesting the hydrogen bonding ability of 3-hydroxy group is important for interactions with cPLA2α. This study helps to understand the influence of specific structural moieties of C1P on the interaction with cPLA2α at the atomistic level and may lead to the design of drugs that regulate cPLA2α activation.


Subject(s)
Ceramides , Drug Design , Surface Plasmon Resonance , Ceramides/chemistry , Ceramides/chemical synthesis , Ceramides/metabolism , Structure-Activity Relationship , Group IV Phospholipases A2/metabolism , Group IV Phospholipases A2/antagonists & inhibitors , Humans , Molecular Structure , Binding Sites
9.
Nanoscale ; 16(21): 10350-10365, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38739006

ABSTRACT

Tumour cells secrete various proangiogenic factors like VEGF, PDGF, and EGF that result in the formation of highly vascularized tumours with an immunosuppressive tumour microenvironment. As tumour growth and metastasis are highly dependent on angiogenesis, targeting tumour vasculature along with rapidly dividing tumour cells is a potential approach for cancer treatment. Here, we specifically engineered sub-100 sized nanomicelles (DTX-CA4 NMs) targeting proliferation and angiogenesis using an esterase-sensitive phosphocholine-tethered docetaxel conjugate of lithocholic acid (LCA) (PC-LCA-DTX) and a poly(ethylene glycol) (PEG) derivative of an LCA-combretastatin A4 conjugate (PEG-LCA-CA4). DTX-CA4 NMs effectively inhibit the tumour growth in syngeneic (CT26) and xenograft (HCT116) colorectal cancer models, inhibit tumour recurrence, and enhance the percentage survival in comparison with individual drug-loaded NMs. DTX-CA4 NMs enhance the T cell-mediated anti-tumour immune response and DTX-CA4 NMs in combination with an immune checkpoint inhibitor, anti-PDL1 antibody, enhance the anti-tumour response. We additionally showed that DTX-CA4 NMs effectively attenuate the production of ceramide-1-phosphate, a key metabolite of the sphingolipid pathway, by downregulating the expression of ceramide kinase at both transcriptional and translational levels. Therefore, this study presents the engineering of effective DTX-CA4 NMs for targeting the tumour microenvironment that can be explored further for clinical applications.


Subject(s)
Cell Proliferation , Ceramides , Docetaxel , Micelles , Neovascularization, Pathologic , Animals , Ceramides/chemistry , Ceramides/pharmacology , Humans , Mice , Cell Proliferation/drug effects , Docetaxel/pharmacology , Docetaxel/chemistry , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Lithocholic Acid/chemistry , Lithocholic Acid/pharmacology , Polyethylene Glycols/chemistry , Cell Line, Tumor , Mice, Inbred BALB C , Stilbenes/chemistry , Stilbenes/pharmacology , HCT116 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Female , Angiogenesis
10.
Int J Mol Sci ; 25(7)2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38612601

ABSTRACT

Cutaneous wound healing is a complex biological process involving a series of well-coordinated events aimed at restoring skin integrity and function. Various experimental models have been developed to study the mechanisms underlying skin wound repair and to evaluate potential therapeutic interventions. This review explores the diverse array of skin wound healing models utilized in research, ranging from rodent excisional wounds to advanced tissue engineering constructs and microfluidic platforms. More importantly, the influence of lipids on the wound healing process is examined, emphasizing their role in enhancing barrier function restoration, modulating inflammation, promoting cell proliferation, and promoting remodeling. Lipids, such as phospholipids, sphingolipids, and ceramides, play crucial roles in membrane structure, cell signaling, and tissue repair. Understanding the interplay between lipids and the wound microenvironment provides valuable insights into the development of novel therapeutic strategies for promoting efficient wound healing and tissue regeneration. This review highlights the significance of investigating skin wound healing models and elucidating the intricate involvement of lipids in the healing process, offering potential avenues for improving clinical outcomes in wound management.


Subject(s)
Ceramides , Inflammation , Humans , Cell Proliferation , Microfluidics , Phospholipids
11.
Skin Res Technol ; 30(4): e13682, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38616504

ABSTRACT

BACKGROUND: Natural products are often friendly and can be used on children's skin after systematic and careful research. Therefore, in this study, the Royal Oji Complex (ROC), a product with natural ingredients, was used to study their effectiveness on keratinocytes taken from the skin of children from 0 to 3 years old. METHOD: Normal human epidermal keratinocytes and tissue-isolated keratinocytes (TIKC) from young donors were treated with three different concentrations of ROC: 0.1, 1, and 10 ppm. The mRNA expression of the epidermal barrier's essential genes, such as hyaluronic acid synthase 3 (Has3), involucrin (IVL), loricrin (LOR), and claudin-1 (CLD1) was investigated using qRT-PCR. Ceramide content was measured by ELISA, with retinoic acid (R.A.) and amarogentin (AMA) serving as positive controls. RESULTS: ROC significantly elevated HAS3 gene expression in HEKn cells, especially at 10 ppm, indicating potential advantages for skin hydration in young infants. IVL increased at first but decreased as ROC concentrations increased. LOR was upregulated at lower ROC concentrations but reduced at higher doses. CLD1 gene expression increased considerably in HEKn but reduced with increasing ROC doses. Ceramide concentration increased somewhat but not significantly at 10 ppm. CONCLUSION: ROC shows potential in altering keratinocyte gene expression, with unique responses in HEKn and TIKC from young donors. While changes in ceramide content were insignificant, these results help to comprehend ROC's multiple effects on young children's skin.


Subject(s)
Keratinocytes , Skin , Child , Infant , Humans , Child, Preschool , Infant, Newborn , Epidermis , Ceramides , Tissue Donors
12.
Hepatol Commun ; 8(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38619429

ABSTRACT

BACKGROUND: Mutations in the gene MTARC1 (mitochondrial amidoxime-reducing component 1) protect carriers from metabolic dysfunction-associated steatohepatitis (MASH) and cirrhosis. MTARC1 encodes the mARC1 enzyme, which is localized to the mitochondria and has no known MASH-relevant molecular function. Our studies aimed to expand on the published human genetic mARC1 data and to observe the molecular effects of mARC1 modulation in preclinical MASH models. METHODS AND RESULTS: We identified a novel human structural variant deletion in MTARC1, which is associated with various biomarkers of liver health, including alanine aminotransferase levels. Phenome-wide Mendelian Randomization analyses additionally identified novel putatively causal associations between MTARC1 expression, and esophageal varices and cardiorespiratory traits. We observed that protective MTARC1 variants decreased protein accumulation in in vitro overexpression systems and used genetic tools to study mARC1 depletion in relevant human and mouse systems. Hepatocyte mARC1 knockdown in murine MASH models reduced body weight, liver steatosis, oxidative stress, cell death, and fibrogenesis markers. mARC1 siRNA treatment and overexpression modulated lipid accumulation and cell death consistently in primary human hepatocytes, hepatocyte cell lines, and primary human adipocytes. mARC1 depletion affected the accumulation of distinct lipid species and the expression of inflammatory and mitochondrial pathway genes/proteins in both in vitro and in vivo models. CONCLUSIONS: Depleting hepatocyte mARC1 improved metabolic dysfunction-associated steatotic liver disease-related outcomes. Given the functional role of mARC1 in human adipocyte lipid accumulation, systemic targeting of mARC1 should be considered when designing mARC1 therapies. Our data point to plasma lipid biomarkers predictive of mARC1 abundance, such as Ceramide 22:1. We propose future areas of study to describe the precise molecular function of mARC1, including lipid trafficking and subcellular location within or around the mitochondria and endoplasmic reticulum.


Subject(s)
Fatty Liver , Hepatocytes , Animals , Humans , Mice , Adipocytes , Biomarkers , Ceramides , Mendelian Randomization Analysis
13.
Sci Rep ; 14(1): 8398, 2024 04 10.
Article in English | MEDLINE | ID: mdl-38600251

ABSTRACT

Allergic rhinitis (AR) is caused by type I hypersensitivity reaction in the nasal tissues. The interaction between CD300f and its ligand ceramide suppresses immunoglobulin E (IgE)-mediated mast cell activation. However, whether CD300f inhibits the development of allergic rhinitis (AR) remains elusive. We aimed to investigate the roles of CD300f in the development of AR and the effectiveness of intranasal administration of ceramide liposomes on AR in murine models. We used ragweed pollen-induced AR models in mice. Notably, CD300f deficiency did not significantly influence the ragweed-specific IgE production, but increased the frequency of mast cell-dependent sneezing as well as the numbers of degranulated mast cells and eosinophils in the nasal tissues in our models. Similar results were also obtained for MCPT5-exprssing mast cell-specific loss of CD300f. Importantly, intranasal administration of ceramide liposomes reduced the frequency of sneezing as well as the numbers of degranulated mast cells and eosinophils in the nasal tissues in AR models. Thus, CD300f-ceramide interaction, predominantly in mast cells, alleviates the symptoms and progression of AR. Therefore, intranasal administration of ceramide liposomes may be a promising therapeutic approach against AR by targeting CD300f.


Subject(s)
Liposomes , Rhinitis, Allergic , Animals , Mice , Administration, Intranasal , Sneezing , Ceramides , Disease Models, Animal , Rhinitis, Allergic/drug therapy , Immunoglobulin E , Nasal Mucosa , Mice, Inbred BALB C , Ovalbumin
14.
Eur Rev Med Pharmacol Sci ; 28(6): 2322-2328, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38567595

ABSTRACT

OBJECTIVE: Strategies for cancer therapy involve radiation therapy (RT), which accounts for about 40% of all cancer treatment types. As to current chemotherapeutics, cancer cells also develop resistance that remains a clinical problem, such as disease recurrence. Recent studies focused on understanding the molecular mechanisms of radiation-induced cell death. Conventional RT aims at treatment with a single fraction per day of 8-30 Gy per fraction. Radiotherapy increases intracellular ceramide levels that trigger cell death. Additionally, increasing intracellular ceramide by radiation may restore therapeutic sensitivity to cancer treatments. Drugs that inhibit ceramide-metabolizing enzymes like ceramidases are expected to be radiotherapy sensitizers. MATERIALS AND METHODS: In this research, we investigated the proapoptotic effects of SRS alone and in combination with ceranib-2, a ceramidase inhibitor in human breast adenocarcinoma cells. The molecular mechanism of action of RT and ceranib-2 was investigated on MCF-7 cells exposed to 13 µM ceranib-2 for 24 hours following 20 Gy radiation using MTT, radiotherapy, and annexin-V analyses. RESULTS: Results indicated that the dose of 20 Gy radiation induces apoptosis on human breast cancer cells with and without co-treatment with ceranib-2 by causing cytotoxicity in the cells. Based on the results of ceranib-2 exposure, it can be concluded that the mechanism of action may rely on an increase of intracellular ceramides, also called apoptotic lipids. CONCLUSIONS: The study results suggest that co-treatment of human breast adenocarcinoma cells with a ceramidase inhibitor, ceranib-2, and a high dose of radiation of 20 Gy exerted cytotoxicity and apoptosis and might be a solid, potent alternative to current therapy strategies.


Subject(s)
Adenocarcinoma , Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Neoplasm Recurrence, Local , Apoptosis , Ceramidases , Ceramides/pharmacology , Ceramides/metabolism
15.
Food Res Int ; 184: 114253, 2024 May.
Article in English | MEDLINE | ID: mdl-38609231

ABSTRACT

Sea cucumbers are a rich source of bioactive compounds and are gaining popularity as nutrient-rich seafood. They are consumed as a whole organism in Pacific regions. However, limited data are available on the comparison of their lipid composition and nutritional value. In this study, untargeted liquid chromatography/mass spectrometry was applied to comprehensively profile lipids in the skin, meat, and intestinal contents of three color-distinct edible sea cucumbers. Multivariate principal component analysis revealed that the lipid composition of the intestinal contents of red, black, and blue sea cucumbers differs from that of skin, and meats. Polyunsaturated fatty acids (PUFAs) are abundant in the intestinal contents, followed by meats of sea cucumber. Lipid nutritional quality assessments based on fatty acid composition revealed a high P:S ratio, low index of atherogenicity, and high health promotion indices for the intestinal contents of red sea cucumber, suggesting its potential health benefits. In addition, hierarchical cluster analysis revealed that the intestinal contents of sea cucumbers were relatively high in PUFA-enriched phospholipids and lysophospholipids. Ceramides are abundant in black skin, blue meat, and red intestinal content samples. Overall, this study provides the first insights into a comprehensive regio-specific profile of the lipid content of sea cucumbers and their potential use as a source of lipid nutrients in food and nutraceuticals.


Subject(s)
Sea Cucumbers , Animals , Ceramides , Cluster Analysis , Dietary Supplements , Fatty Acids
16.
Biochem Biophys Res Commun ; 715: 149980, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38678780

ABSTRACT

The transport of ceramide from the endoplasmic reticulum (ER) to the Golgi is a key step in the synthesis of complex sphingolipids, the main building blocks of the plasma membrane. In yeast, ceramide is transported to the Golgi either through ATP-dependent COPII vesicles of the secretory pathway or by ATP-independent non-vesicular transport that involves tethering proteins at ER-Golgi membrane contact sites. Studies in both mammalian and yeast cells reported that vesicular transport mainly carries ceramide containing very long chain fatty acids, while the main mammalian non-vesicular ceramide transport protein CERT only transports ceramides containing short chain fatty acids. However, if non-vesicular ceramide transport in yeast similarly favors short chain ceramides remained unanswered. Here we employed a yeast GhLag1 strain in which the endogenous ceramide synthase is replaced by the cotton-derived GhLag1 gene, resulting in the production of short chain C18 rather than C26 ceramides. We show that block of vesicular transport through ATP-depletion or the use of temperature-sensitive sec mutants caused a reduction in inositolphosphorylceramide (IPC) synthesis to similar extent in WT and GhLag1 backgrounds. Since the remaining IPC synthesis is a readout for non-vesicular ceramide transport, our results indicate that non-vesicular ceramide transport is neither blocked nor facilitated when only short chain ceramides are present. Therefore, we propose that the sorting of ceramide into non-vesicular transport is independent of acyl chain length in budding yeast.


Subject(s)
Ceramides , Golgi Apparatus , Saccharomyces cerevisiae , Ceramides/metabolism , Golgi Apparatus/metabolism , Biological Transport , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomycetales/metabolism , Saccharomycetales/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Endoplasmic Reticulum/metabolism , Adenosine Triphosphate/metabolism , Glycosphingolipids
17.
Signal Transduct Target Ther ; 9(1): 97, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38664391

ABSTRACT

Bile acids, once considered mere dietary surfactants, now emerge as critical modulators of macronutrient (lipid, carbohydrate, protein) metabolism and the systemic pro-inflammatory/anti-inflammatory balance. Bile acid metabolism and signaling pathways play a crucial role in protecting against, or if aberrant, inducing cardiometabolic, inflammatory, and neoplastic conditions, strongly influencing health and disease. No curative treatment exists for any bile acid influenced disease, while the most promising and well-developed bile acid therapeutic was recently rejected by the FDA. Here, we provide a bottom-up approach on bile acids, mechanistically explaining their biochemistry, physiology, and pharmacology at canonical and non-canonical receptors. Using this mechanistic model of bile acids, we explain how abnormal bile acid physiology drives disease pathogenesis, emphasizing how ceramide synthesis may serve as a unifying pathogenic feature for cardiometabolic diseases. We provide an in-depth summary on pre-existing bile acid receptor modulators, explain their shortcomings, and propose solutions for how they may be remedied. Lastly, we rationalize novel targets for further translational drug discovery and provide future perspectives. Rather than dismissing bile acid therapeutics due to recent setbacks, we believe that there is immense clinical potential and a high likelihood for the future success of bile acid therapeutics.


Subject(s)
Bile Acids and Salts , Signal Transduction , Bile Acids and Salts/metabolism , Humans , Signal Transduction/drug effects , Animals , Ceramides/metabolism , Ceramides/genetics
18.
Int Immunopharmacol ; 133: 112083, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38648714

ABSTRACT

Japanese encephalitis virus (JEV) infection is considered a global public health emergency. Severe peripheral neuropathy caused by JEV infection has increased disability and mortality rates in recent years. Because there are very few therapeutic options for JEV infection, prompt investigations of the ability of clinically safe, efficacious and globally available drugs to inhibit JEV infection and ameliorate peripheral neuropathy are urgently needed. In this study, we found that high doses of intravenous immunoglobulin, a function inhibitor of acid sphingomyelinase (FIASMA), inhibited acid sphingomyelinase (ASM) and ceramide activity in the serum and sciatic nerve of JEV-infected rats, reduced disease severity, reversed electrophysiological and histological abnormalities, significantly reduced circulating proinflammatory cytokine levels, inhibited Th1 and Th17 cell proliferation, and suppressed the infiltration of inflammatory CD4 + cells into the sciatic nerve. It also maintained the peripheral nerve-blood barrier without causing severe clinical side effects. In terms of the potential mechanisms, ASM was found to participate in immune cell differentiation and to activate immune cells, thereby exerting proinflammatory effects. Therefore, immunoglobulin is a FIASMA that reduces abnormal immune responses and thus targets the ASM/ceramide system to treat peripheral neuropathy caused by JEV infection.


Subject(s)
Ceramides , Encephalitis Virus, Japanese , Encephalitis, Japanese , Immunoglobulins, Intravenous , Peripheral Nervous System Diseases , Sphingomyelin Phosphodiesterase , Animals , Ceramides/metabolism , Immunoglobulins, Intravenous/therapeutic use , Immunoglobulins, Intravenous/pharmacology , Encephalitis Virus, Japanese/immunology , Encephalitis Virus, Japanese/physiology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/virology , Rats , Encephalitis, Japanese/drug therapy , Encephalitis, Japanese/immunology , Male , Sciatic Nerve/pathology , Cytokines/metabolism , Signal Transduction/drug effects , Humans , Th1 Cells/immunology , Rats, Sprague-Dawley , Th17 Cells/immunology
19.
Biochim Biophys Acta Biomembr ; 1866(5): 184324, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38688405

ABSTRACT

The stratum corneum (SC) lipid matrix, composed primarily of ceramides (CERs), cholesterol and free fatty acids (FFA), has an important role for the skin barrier function. The presence of the long periodicity phase (LPP), a unique lamellar phase, is characteristic for the SC. Insight into the lipid molecular arrangement within the LPP unit cell is imperative for understanding the relationship between the lipid subclasses and the skin barrier function. In this study, the impact of the CER head group structure on the lipid arrangement and barrier functionality was investigated using lipid models forming the LPP. The results demonstrate that the positions of CER N-(tetracosanoyl)-sphingosine (CER NS) and CER N-(tetracosanoyl)-phytosphingosine (CER NP), two essentials CER subclasses, are not influenced by the addition of another CER subclass (N-(tetracosanoyl)-dihydrosphingosine (CER NdS), N-(2R-hydroxy-tetracosanoyl)-sphingosine (CER AS) or D-(2R-hydroxy-tetracosanoyl)-phytosphingosine (CER AP)). However, differences are observed in the lipid organization and the hydrogen bonding network of the three different models. A similar localization of CER NP and CER NS is also observed in a more complex lipid model, with the CER subclass composition mimicking that of human SC. These studies show the adaptability and insensitivity of the LPP unit cell structure to changes in the lipid head group structures of the CER subclasses.


Subject(s)
Ceramides , Epidermis , Ceramides/chemistry , Humans , Epidermis/metabolism , Epidermis/chemistry , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Sphingosine/metabolism , Cholesterol/chemistry , Cholesterol/metabolism
20.
Molecules ; 29(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38675558

ABSTRACT

The cannabinoid-type I (CB1) receptor functions as a double-edged sword to decide cell fate: apoptosis/survival. Elevated CB1 receptor expression is shown to cause acute ceramide accumulation to meet the energy requirements of fast-growing cancers. However, the flip side of continual CB1 activation is the initiation of a second ceramide peak that leads to cell death. In this study, we used ovarian cancer cells, PA1, which expressed CB1, which increased threefold when treated with a natural compound, bis(palmitoleic acid) ester of a glycerol (C2). This novel compound is isolated from a marine snail, Conus inscriptus, using hexane and the structural details are available in the public domain PubChem database (ID: 14275348). The compound induced two acute ceramide pools to cause G0/G1 arrest and killed cells by apoptosis. The compound increased intracellular ceramides (C:16 to 7 times and C:18 to 10 times), both of which are apoptotic inducers in response to CB1 signaling and thus the compound is a potent CB1 agonist. The compound is not genotoxic because it did not induce micronuclei formation in non-cancerous Chinese hamster ovarian (CHO) cells. Since the compound induced the cannabinoid pathway, we tested if there was a psychotropic effect in zebrafish models, however, it was evident that there were no observable neurobehavioral changes in the treatment groups. With the available data, we propose that this marine compound is safe to be used in non-cancerous cells as well as zebrafish. Thus, this anticancer compound is non-toxic and triggers the CB1 pathway without causing psychotropic effects.


Subject(s)
Apoptosis , Ceramides , Conus Snail , Fatty Acids , Receptor, Cannabinoid, CB1 , Animals , Female , Humans , Apoptosis/drug effects , Cell Line, Tumor , Ceramides/metabolism , Ceramides/chemistry , Fatty Acids/pharmacology , Fatty Acids/chemistry , Fatty Acids/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/genetics , Signal Transduction/drug effects , Conus Snail/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...