Next Article in Journal
Malignant Epithelioid Neoplasm versus Dedifferentiated Malignant Melanoma: A Case Report
Previous Article in Journal
The COVID-19 Pandemic and Emergencies in Otolaryngology–Head and Neck Surgery: An Analysis of Patients Presenting to Emergency Rooms in South-West Germany: A Bi-Center Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ceramides—Emerging Biomarkers of Lipotoxicity in Obesity, Diabetes, Cardiovascular Diseases, and Inflammation

Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
*
Author to whom correspondence should be addressed.
Diseases 2024, 12(9), 195; https://doi.org/10.3390/diseases12090195
Submission received: 12 July 2024 / Revised: 17 August 2024 / Accepted: 23 August 2024 / Published: 23 August 2024

Abstract

:
Abnormalities in lipid homeostasis have been associated with many human diseases, and the interrelation between lipotoxicity and cellular dysfunction has received significant attention in the past two decades. Ceramides (Cers) are bioactive lipid molecules that serve as precursors of all complex sphingolipids. Besides their function as structural components in cell and mitochondrial membranes, Cers play a significant role as key mediators in cell metabolism and are involved in numerous cellular processes, such as proliferation, differentiation, inflammation, and induction of apoptosis. The accumulation of various ceramides in tissues causes metabolic and cellular disturbances. Recent studies suggest that Cer lipotoxicity has an important role in obesity, metabolic syndrome, type 2 diabetes, atherosclerosis, and cardiovascular diseases (CVDs). In humans, elevated plasma ceramide levels are associated with insulin resistance and impaired cardiovascular and metabolic health. In this review, we summarize the role of ceramides as key mediators of lipotoxicity in obesity, diabetes, cardiovascular diseases, and inflammation and their potential as a promising diagnostic tool.

1. Introduction

Abnormalities in lipid homeostasis have been associated with many human diseases, leading to the rise of lipidomics as an emerging field studying lipids and factors that interact with lipids [1,2]. The term “lipotoxicity” was first adopted in 1994 by Lee et al. in the context of the pathogenesis of obesity-related β-cell alterations. The study described how lipid overload in pancreatic β-cells led to loss of function and the onset of type 2 diabetes mellitus (T2DM) in rats [3]. Lipotoxicity refers to the process of excess accumulation of lipids in non-adipose tissues and over-activation of lipid signaling pathways that induce cellular dysfunction. The increased concentrations of lipids and lipid derivatives result in many simultaneously occurring metabolic and functional disturbances in multiple organs and systems. The main organs affected include the pancreas, liver, skeletal muscles, heart, and kidneys [4]. The interrelation between lipotoxicity and cellular dysfunction has focused significant attention in the past two decades on a wide range of prevalent diseases, including obesity, diabetes, CVDs, cancer, and neurodegenerative diseases [4,5,6,7].
Ceramides (Cers) are bioactive lipid molecules that serve as precursors of all complex sphingolipids. Ceramides are composed of sphingosine acylated with a fatty acid, usually saturated or monounsaturated, with a chain length ranging from 14 to 26 carbon [8]. Besides their function as structural components in cell and mitochondrial membranes, Cers play significant role as key mediators in cell metabolism and are involved in numerous cellular processes, such as proliferation, differentiation, inflammation, and induction of apoptosis. Ceramides are considered potential lipotoxic agents that modulate various processes and cause metabolic dysfunction [9,10,11,12]. Some of the effects of ceramide accumulation on several pathways include inhibition of PI3K/Akt pathway (impairing insulin signaling and glucose uptake), inhibition of hormone-sensitive lipase (directly leading to excessive storage of triglycerides), overproduction of ROS in mitochondria via the inhibition of complex III and IV of the respiratory chain (reducing intracellular ATP production and causing oxidative stress and apoptosis), and activation of proinflammatory mediator synthesis, such as TNF-α, interferon-γ, and IL-1β (inducing inflammation) [8,9,10,11,12,13].

2. Ceramide Synthesis and Transport

The synthesis and degradation of ceramides play crucial roles in maintaining cell homeostasis. Ceramides, the hub of the sphingolipid pathway, can be produced through three different pathways: (1) de novo synthesis, (2) sphingomyelin hydrolysis, or (3) salvage pathway (Figure 1). De novo synthesis occurs in the endoplasmic reticulum and starts with the condensation of serine with palmitoyl-CoA to form 3-ketosphinganine at the cytosolic surface of the ER, which is the rate-limiting step that is catalyzed by serine palmitoyltransferase. The carbonyl group of 3-ketosphinganine is then reduced by 3-ketosphinganine reductase to form sphinganine, which can be acylated with a fatty acyl-CoA by one of six (dihydro)ceramide synthases to form dihydroceramide. Dihydroceramide desaturase inserts a double bond into the sphingoid base to form ceramides. Once synthesized, Cers are transported into the Golgi apparatus via ceramide transfer protein or transport vesicles, where they can be converted to complex sphingolipids. In the second pathway, Cers are synthesized via the hydrolysis of the phosphocholine head groups of sphingomyelin (SM) by five different isoforms of sphingomyelinases. Because the amount of SM is much higher than the amount of total ceramide in the cell, hydrolysis of a small portion of SM significantly changes ceramide levels. In the salvage pathway, Cers are recycled from sphingosine re-acylation that is catalyzed by ceramide synthase [8,11,12,13,14,15,16,17].
Twenty years ago, ceramides were thought to be a homogenous class of sphingolipids, but now it is known that ceramides exert fundamentally different biological roles depending on the fatty acyl chain lengths [6]. Ceramides are largely synthesized by six different fatty acyl selective ceramide synthase (CerS) isoforms (CerS1-CerS6) with specific tissue distribution. Each CerS exhibits substrate specificity toward acyl-CoAs carrying different chain lengths (CerS1 is mainly expressed in muscles, C18; CerS2 is mainly expressed in liver, kidney, small intestine, and heart, C22 and C24; CerS4, C20, C22, C24, CerS5, C16, and CerS6 are mainly expressed in adipose tissue, C16; and only CerS3 exhibits broad substrate specificity toward medium- to long-chain fatty acyl-CoAs) (Table 1) [11,12,17,18,19,20,21,22,23]. In humans, CerS enzymes are involved in the regulation of various cellular and metabolic functions. For example, C18:0 ceramide produced by CerS1 is essential for brain development and neuronal signaling, while ceramides produced by CerS2 maintain the normal functions of liver, lungs, brain, heart, and kidney. However, it has been found that dysregulation of CerS leads to increased levels of various ceramide subspecies that have a key role in the pathophysiology of various human diseases [12].
In plasma, ceramides are carried by low-density and high-density lipoproteins (LDL and HDL). LDL is the major carrier of Cers in the plasma (about 60% of total plasma Cers), whereas HDL carries about 24% of plasma Cers. In addition, Cers are also found in extracellular vesicles (lipid vesicles secreted by cells) [24].
The accumulation of various ceramides in tissues causes metabolic and cellular dysfunction [25]. In recent years, Cers have received attention for their prominent role in obesity, metabolic syndrome, T2DM, atherosclerosis, and cardiovascular diseases (CVDs) [6,8,10,11,13,26,27]. Plasma levels of ceramides are increased in patients with obesity and type 2 diabetes mellitus, and are associated with insulin resistance and impaired cardiovascular and metabolic health [10,27,28,29,30,31].

3. Ceramides and Obesity

Overweight and obesity are global health problems, and their prevalence is constantly increasing [32]. According to a WHO report, the worldwide prevalence of obesity has tripled since 1975, mainly due to sedentary lifestyle and unhealthy diets [33]. It is estimated that more than 2.1 billion people worldwide (30% of the global population) are obese or overweight, and a total of 380 million children and adolescents are affected [34,35]. Obesity is characterized by excessive accumulation of body fat within adipose tissue, which has detrimental effects on human health [36]. Body mass index (BMI) is the most commonly used measure of overweight and obesity in men and women and is calculated by dividing weight in kilograms by height in meters squared. A BMI between 25 and 29.9 indicates overweight, and a BMI ≥ 30 indicates obesity.
Obesity is a major risk factor for the development of a number of chronic and life-threatening diseases, including dyslipidemia, insulin resistance, type 2 diabetes, and CVDs [8,25,26]. Excess calorie intake results in dyslipidemia and increased fatty acid supply, which leads to accumulation of lipid metabolites in peripheral tissues such as the liver, skeletal muscle, heart, and pancreas. The biologically active lipids that accumulate in obesity include triacylglycerol (TAG), diacylglycerol (DAG), long-chain fatty acids, acylcarnitine, phospholipids, and ceramides [6,25,37,38]. It is reported that ceramides are increased in the skeletal muscle, liver, and hypothalamus of obese rodents and humans. Different mechanisms lead to the increased formation of ceramides in peripheral tissues in obesity and lipid excess [25,39]. One of these is increased plasma-free fatty acids used for de novo ceramide synthesis and another is the increased expression of a number of genes involved in de novo synthesis. Other factors that may impair ceramide metabolism in obesity include pro-inflammatory cytokines, oxidative stress, and hormones.
Previous studies report the role of adipokine adiponectin in the control of ceramide levels [8,10]. Adiponectin is predominantly secreted from mature white adipocytes and regulates glucose and lipid homeostasis acting on many tissues, including the liver, kidney, adipose tissue, and pancreas, and exerting anti-diabetic, anti-inflammatory, and cardioprotective actions [8,10,30]. Adiponectin receptors AdipoR1 and AdipoR2 possess ceramidase activity that is activated by adiponectin binding and degrades ceramide into sphingosine, decreasing ceramide levels [6,8,10,20,30,37,40,41]. Sphingosine can be converted to sphingosine-1-phosphate (S1P) by sphingosine kinase (SphKs) [19]. Two major isoforms of SphKs in mammals have been described, namely, SphK1 and SphK2. S1P is a bioactive lipid signaling molecule that controls many physiological and pathological processes by binding with five different G-protein-coupled S1P receptors. The concentration of S1P is higher in plasma than in tissues and is carried mainly by HDL and albumin [42].
The reciprocal balance of ceramide and S1P has been termed ceramide/S1P rheostat. These two sphingolipid metabolites are important second messengers able to regulate cellular functions by modulating opposing signaling pathways. Ceramide has been shown to induce apoptosis, whereas S1P stimulates cell survival, proliferation, and tissue regeneration. Hence, maintaining the ceramide/sphingosine-1-phosphate balance is crucial for cells and cell fate decisions (Figure 2) [43,44,45]. Because ceramide and S1P are interconvertible metabolites, an increase in ceramide levels causes a decrease in S1P and vice versa. Therefore, the ceramide/S1P rheostat has been explored for the development of new therapeutic strategies for various diseases [46].
Sphingosine and/or sphingosine-1-phosphate prevent apoptosis of pancreatic β-cells and exert an anti-diabetic effect [30]. Sphingosine-1-phosphate binds to its receptors and activates AMPK, a serine/threonine kinase that stimulates glucose uptake and lipid oxidation [37,47,48].
Obesity induces low-grade inflammation that contributes to the development of obesity comorbidities, such as insulin resistance, T2DM, and atherosclerosis [49,50]. White adipose tissue (WAT) is not just a lipid storage organ but also an endocrine organ that affects metabolism and inflammation via the secretion of different cytokines and adipokines. WAT is composed of adipocyte precursors, adipocytes, and immune cells, mainly macrophages [51]. Adipocytes and macrophages that infiltrate adipose tissue secrete proinflammatory cytokines such as IL-1, IL-6, and TNF-α capable of impairing insulin signaling [51]. In obesity, adipocytes can increase either in size (hypertrophy) or in number (hyperplasia). Hypertrophy is associated with hypoxia of adipocytes, which activates the HIF-1 gene, leads to cell stress, and activates cell death and transcription factor NF-κB [19,50]. NF-κB stimulates the production of proinflammatory cytokines and chemokine monocytechemotactic protein-1 (MCP-1), which leads to macrophage recruitment and increased secretion of proinflammatory cytokines [19,50]. This significantly impairs the transduction of insulin signals in adipocytes and causes excessive lipolysis, cell death, and progression of insulin resistance [19]. In obese individuals the plasma levels of leptin are increased, while adiponectin levels are decreased [19,51]. The reduced circulating adiponectin levels in obesity result in reduced stimulation of AdipoRs and decreased ceramide degradation in tissues, which contributes to the development of insulin resistance [8].

4. Ceramides and Type 2 Diabetes

Lipotoxicity interferes with insulin signaling and contributes to the development of numerous diseases, including T2DM. Various lipids have been measured in diabetic patients, and ceramides have been proposed to play an important role in the pathogenesis of diabetes and its associated complications. Previous studies report that individuals with T2DM had higher serum and tissue ceramide levels than their nondiabetic counterparts and that ceramide levels were predictors of insulin resistance in T2DM [52,53]. The plasma concentration of dihydroceramide was elevated years prior to diabetes diagnosis and was associated with disease predisposition [54]. Previous studies report that insulin resistance correlated with ceramide levels in plasma, adipose tissue, liver, and skeletal muscles [55,56]. Research suggests that an accumulation of ceramides in the tissues impairs insulin signaling and glucose uptake by the PI3K pathway, which is the major pathway involved in glucose transport. Ceramides cause inactivation of PKB (Akt), which is needed for insulin signaling, and impair the translocation of the glucose transporter GLUT 4 to the cell membrane. Firstly, ceramides inhibit PKB through activation of its dephosphorylation, catalyzed by protein phosphatase 2A. Secondly, they inactivate PKB through its binding to the inhibitory PKCzeta protein. Ceramides may also decrease GLUT 4 gene transcription [12,57,58]. Therefore, ceramides prevent the uptake of glucose and its storage as glycogen and triglycerides. This results in elevated levels of glucose in the blood and decreased insulin sensitivity [57]. Previous studies show that circulating ceramides may be early diagnostic indicators of metabolic diseases like diabetes. Haus et al. showed that type 2 diabetic subjects had higher plasma concentrations of Cer-C18:0, C20:0, and C24:1 compared to healthy controls and that insulin sensitivity was inversely correlated with these ceramide levels [55]. Brozinick et al. revealed that distinct ceramide species with medium and long fatty acid chains were high in non-human primates (macaque monkeys) with prediabetes and diabetes [59]. Sokolowska et al. found that in adult patients with newly diagnosed diabetes, Cer-C22:0 and Cer-C24:0 positively correlated with total cholesterol, triglycerides, and low-density lipoprotein cholesterol (LDL), particularly in autoimmune diabetes, while in T2DM, there was a positive correlation only between Cer-C24:0 and total cholesterol. The authors conclude that monitoring Cer and sphingomyelins levels may help with more accurate differentiation between autoimmune diabetes and T2DM [60].

5. Ceramides and Cardiovascular Diseases

Cardiovascular diseases remain the most common cause of death worldwide despite significant advancements in their therapy. In 2021 alone, 20.5 million people died from CVDs, representing approximately one-third of all global deaths [61,62,63]. The main contributors to CVDs include smoking, alcohol intake, hypertension, diabetes mellitus, obesity, and dyslipidemia [64,65].
The lipid profile is used as a predictor of CVD risk. The traditional blood tests of lipid status include LDL-cholesterol, HDL-cholesterol, and triglyceride levels, but these routine biomarkers fail to identify all patients at high risk of cardiovascular events [66,67]. Recent studies have identified ceramides to be involved in the development of CVDs, including atherosclerosis, coronary artery disease, heart failure, and myocardial infarction [10,12,68,69,70]. Dysregulation of ceramide and sphingolipid metabolism is suggested to be a key factor in cardiac lipotoxicity, and data from both animal and human studies show that sphingolipid accumulation in cardiomyocytes is associated with cardiac hypertrophy [71,72]. Ceramide has been considered a new predictor for CV events, with higher predictive value than LDL cholesterol [13,62,66]. It is suggested that Cers are involved in atherogenesis by promoting LDL aggregation, remodeling the vascular wall, and increasing inflammation [13,70]. One of the suggested mechanisms for ceramide-induced cardiovascular manifestations is that sphingomyelin accumulates in atherosclerotic lesions, where it may be hydrolyzed by SMases, thus increasing LDL-ceramide and their faster aggregation in the arterial wall, leading to atherosclerosis [42].
Ceramides were reported to activate TNF-α and NF-κB pathways, initiating an inflammation cascade. Plasma Cers were also found to be strongly correlated with the pro-inflammatory cytokine IL-6. Eventually, Cer-induced inflammation is a key factor for the development of atherosclerosis and CVDs [12,42].
Slijkhuis et al. investigated the lipid content of human carotid atherosclerotic plaque in detail and provided new insights into the role of lipids in atherosclerosis [73]. The authors identified SM and Cers as the lipid species that accumulate prominently in the calcified areas of the plaque. Other authors suggest that Cers have a crucial role in the pathogenesis of CVDs and may serve as new diagnostic markers for the progression of atherosclerosis [74]. Recent studies have reported an association between ceramides and cardiovascular risk factors such as age, arterial hypertension, and obesity and the altered ceramide profile observed with atherosclerosis progression. Certain types of ceramides are identified as proatherogenic mediators and are associated with increased cardiovascular risk [61,70,75,76]. Therefore, measurement of plasma ceramides may improve the identification of patients at high risk for cardiovascular events [62,77,78]. Ceramide risk score, composed of specific ceramides and their corresponding ratios, has been proposed as a promising indicator that provides additional predictive value for the assessment of cardiovascular disease risk in different populations [79,80]. The ceramide-based coronary event risk test (CERT) is determined based on four ceramides and their ratios, increasing with risk level. Ceramide score 1 (CERT 1), composed of Cer (d18:1/16:0), Cer (d18:1/18:0), Cer (d18:1/24:1), and their ratios to Cer (d18:1/24:0), applies a scale of 0 to 12 points, and patients are categorized into four risk groups (low–moderate–increased–high). The low-risk score is 0 to 2, the moderate-risk score is 3 to 6, the increased-risk score is 7 to 9, and high-risk is 10 to 12 [31,67,81]. Ceramides C16:0 and C18:0 are considered harmful, whereas those containing very long chains (e.g., C24) are considered protective. In order to improve the reliability of CERT 1, another ceramide risk score, i.e., CERT 2, has been proposed that is composed of four components—one ceramide/ceramide ratio, two ceramide/phosphatidylcholine (PC) ratios, and a single PC [67]. CERT 2 is a novel updated risk score able to predict major adverse cardiovascular events (MACEs) and cardiovascular diseases. Ceramides and phosphatidylcholines are incorporated into this parameter, which reflects the risk of CVD incidents [76,79]. CERT 1 and CERT 2 have been reported as stronger predictors for the assessment of CV risk compared to traditional biomarkers such as LDL cholesterol and high-sensitivity C-reactive protein. Both scores are effective tools able to predict CV events such as myocardial infarction, stroke, and cardiovascular death and could improve the identification of high-risk patients [11,31,67,76,79,80,82,83,84,85]. Previous studies have shown that blocking ceramide production can be an effective strategy for the treatment of CVD, and pharmacological inhibition of key enzymes and receptors in the three different processes of ceramides biosynthesis can reduce ceramide levels [62,75]. Clinical studies have confirmed that cholesterol-biosynthesis inhibitors, such as statins (which inhibit HMG-CoA reductase), effectively reduce plasma ceramides. Furthermore, proprotein convertase subtilisin/kexin type 9 (PCSK-9) inhibitors, monoclonal antibodies used in the therapy of hypercholesterolemia, and its related cardiovascular diseases were found to reduce both cholesterol and, potentially, ceramide levels and were suggested as a new therapeutic option for the treatment of dyslipidemia [62,67]. On the other hand, glucagon-like peptide 1 (GLP-1) receptor agonists (GLP-1 analogs), a class of medications used as therapeutic agents in T2DM, have shown cardioprotective effects against ceramide accumulation. GLP-1 analogs such as liraglutide lower glucose levels by stimulating pancreatic β-cells to release insulin. These agents also suppress the release of glucagon from α-cells and the hepatic glucose output, promote β-cell proliferation and reduce β-cell apoptosis, reduce appetite, and delay gastric emptying, thus contributing to long-term weight loss [12,86]. Although direct evidence is not yet available, it is suggested that a weight loss of at least 10% is associated with a 21% reduction in cardiovascular events [87]. According to the 2023 American Diabetes Association (ADA) guidelines, GLP-1 receptor agonists are mmended as a novel approach for mitigating cardiovascular risk. It is also reported that liraglutide inhibits the accumulation of C16:0 and C24:0 ceramide in mice liver, preventing subsequent inflammation and fibrosis [12]. Additional therapies targeting ceramide biosynthesis (e.g., myriocin—an inhibitor of serine palmitoyltransferase (SPT), the initial and rate-limiting enzyme in de novo synthesis of ceramide) and ceramide inhibitors (e.g., fumonisin B1, fungin FTY720, etc.), are under investigation [11,12]. Myriocin, a natural specific inhibitor of SPT, has shown promising results in the treatment of several diseases, including CVDs. It was reported to reduce atherosclerosis, hepatic steatosis, and fibrosis in mice induced by elevated ceramide levels. Moreover, it could restore normal vasodilation of blood vessels via activation of endothelial NO synthase (eNOS) and NO release [12]. However, future laboratory and clinical research on the therapeutic options for ceramide reduction is necessary. Traditional therapies such as lipid-lowering drugs, lifestyle changes, increased physical activity, and dietary interventions may be employed to control ceramide levels while specific ceramide- targeted drugs are being developed [31].

6. Ceramides and Inflammation

Inflammation is part of the body’s response mechanism to infection or tissue injury. It is the process by which the immune system recognizes and destroys harmful and foreign substances and begins the healing of tissues. In chronic conditions, persistent low-grade inflammation is often observed [13,88]. Elevated Cers lead to stimulated synthesis of proinflammatory mediators such as TNF-α, interferon-γ, IL-1β, or platelet-activating factor that enhance the activity of both lysosomal and membrane sphingomyelinases, thus stimulating Cer synthesis and incremental inflammation [11,13,30,40,89]. Ceramide production induces inflammation through activation of the proinflammatory transcription factor NF-κB. NF-κB is a family of transcription factors that induce genes encoding proinflammatory cytokines and proteins, such as IL-1β, IL-6, IL-8, monocyte chemoattractant protein-1, and COX-2, all of which play crucial roles in the inflammatory responses [90]. One of the key modulators of ceramide biosynthesis is Toll-like receptor-4 (TLR4). The receptor is activated on macrophages in response to bacterial infection and tissue damage. Ligands of TLR4 include lipopolysaccharides, low-density lipoproteins, viral proteins, polysaccharides, etc. Previous studies suggest that TLR signaling in macrophages is not only required for innate immunity against pathogens but also contributes to the pathogenesis of many diseases. One of the TLR4 signaling pathways leads to MAP kinases and NF-κB activation and the release of inflammatory molecules. In addition to cytokine production, another response of TLR4-mediated macrophage activation is the imbalance of cellular lipids. Recent studies have revealed that TLR4 agonists induce the expression of several enzymes in the de novo pathway of ceramide biosynthesis, which results in an increase in cellular sphingolipids, including ceramide [40,91,92,93,94].

7. Conclusions

Recent studies reveal the impact of ceramides on metabolic and cardiovascular health and provide evidence that the accumulation of ceramides induces cellular dysfunction. Large cohort human studies convincingly demonstrate that the simultaneous measurement of ceramides together with routine biomarkers of lipid profile may improve the diagnostic accuracy of metabolic diseases like diabetes, and may be a promising diagnostic tool for better assessment of the progression of cardiovascular diseases and a potent predictor of CV events. Therefore, further research on specific ceramide species and their tissue and organ distribution will shed light on the molecular mechanisms of ceramide-induced lipotoxicity. In conclusion, thorough understanding of the dysregulation of ceramide metabolism will add information to clarify the pathogenesis of a variety of diseases, and will help to identify better therapeutic targets and to develop effective treatments.

Author Contributions

G.D. designed the review; G.D., K.S. and T.S. wrote the manuscript; G.D. created the figures and bibliographic contents; G.D. edited and reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

No patient data are used.

Data Availability Statement

Review, no original data sheets are available.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Barranco-Altirriba, M.; Alonso, N.; Weber, R.J.M.; Lloyd, G.R.; Hernandez, M.; Yanes, O.; Capellades, J.; Jankevics, A.; Winder, C.; Falguera, M.; et al. Lipidome characterisation and sex-specific differences in type 1 and type 2 diabetes mellitus. Cardiovasc. Diabetol. 2024, 23, 109. [Google Scholar] [CrossRef] [PubMed]
  2. Malý, M.; Kučerka, O.; Bechyňská, K.; Kočí, K.; Mandys, V.; Hajšlová, J.; Kosek, V. Plasma lipidome differences in patients with and without significant carotid plaque. Vasc. Pharmacol. 2024, 155, 107377. [Google Scholar] [CrossRef] [PubMed]
  3. Lee, Y.; Hirose, H.; Ohneda, M.; Johnson, J.H.; McGarry, J.D.; Unger, R.H. β-Cell Lipotoxicity in the Pathogenesis of Non-Insulin-Dependent Diabetes Mellitus of Obese Rats: Impairment in Adipocyte-β-Cell Relationships. Proc. Natl. Acad. Sci. USA 1994, 91, 10878–10882. [Google Scholar] [CrossRef] [PubMed]
  4. Lipke, K.; Kubis-Kubiak, A.; Piwowar, A. Molecular Mechanism of Lipotoxicity as an Interesting Aspect in the Development of Pathological States—Current View of Knowledge. Cells 2022, 11, 844. [Google Scholar] [CrossRef]
  5. Angarita-Rodríguez, A.; Matiz-González, J.M.; Pinzón, A.; Aristizabal, A.F.; Ramírez, D.; Barreto, G.E.; González, J. Enzymatic Metabolic Switches of Astrocyte Response to Lipotoxicity as Potential Therapeutic Targets for Nervous System Diseases. Pharmaceuticals 2024, 17, 648. [Google Scholar] [CrossRef]
  6. Turpin-Nolan, S.M.; Brüning, J.C. The role of ceramides in metabolic disorders: When size and localization matters. Nat. Rev. Endocrinol. 2020, 16, 224–233. [Google Scholar] [CrossRef]
  7. Nicholson, R.J.; Norris, M.K.; Poss, A.M.; Holland, W.L.; Summers, S.A. The Lard Works in Mysterious Ways: Ceramides in Nutrition-Linked Chronic Disease. Annu. Rev. Nutr. 2022, 42, 115–144. [Google Scholar] [CrossRef]
  8. Hammerschmidt, P.; Brüning, J.C. Contribution of specific ceramides to obesity-associated metabolic diseases. Cell Mol. Life Sci. 2022, 79, 395. [Google Scholar] [CrossRef]
  9. Kauhanen, D.; Sysi-Aho, M.; Koistinen, K.M.; Laaksonen, R.; Sinisalo, J.; Ekroos, K. Development and validation of a high-throughput LC-MS/MS assay for routine measurement of molecular ceramides. Anal. Bioanal. Chem. 2016, 408, 3475–3483. [Google Scholar] [CrossRef]
  10. Field, B.C.; Gordillo, R.; Scherer, P.E. The Role of Ceramides in Diabetes and Cardiovascular Disease Regulation of Ceramides by Adipokines. Front. Endocrinol. 2020, 11, 569250. [Google Scholar] [CrossRef]
  11. Gaggini, M.; Ndreu, R.; Michelucci, E.; Rocchiccioli, S.; Vassalle, C. Ceramides as Mediators of Oxidative Stress and Inflammation in Cardiometabolic Disease. Int. J. Mol. Sci. 2022, 23, 2719. [Google Scholar] [CrossRef]
  12. Shalaby, Y.M.; Al Aidaros, A.; Valappil, A.; Ali, B.R.; Akawi, N. Role of Ceramides in the Molecular Pathogenesis and Potential Therapeutic Strategies of Cardiometabolic Diseases: What we Know so Far. Front. Cell Dev. Biol. 2022, 9, 816301. [Google Scholar] [CrossRef] [PubMed]
  13. Walchuk, C.; Wang, Y.; Suh, M. The impact of EPA and DHA on ceramide lipotoxicity in the metabolic syndrome. Br. J. Nutr. 2021, 125, 863–875. [Google Scholar] [CrossRef] [PubMed]
  14. Seal, A.; Hughes, M.; Wei, F.; Pugazhendhi, A.S.; Ngo, C.; Ruiz, J.; Schwartzman, J.D.; Coathup, M.J. Sphingolipid-Induced Bone Regulation and Its Emerging Role in Dysfunction Due to Disease and Infection. Int. J. Mol. Sci. 2024, 25, 3024. [Google Scholar] [CrossRef]
  15. Adada, M.; Luberto, C.; Canals, D. Inhibitors of the sphingomyelin cycle: Sphingomyelin synthases and sphingomyelinases. Chem. Phys. Lipids 2016, 197, 45–59. [Google Scholar] [CrossRef]
  16. Sessa, L.; Nardiello, A.M.; Santoro, J.; Concilio, S.; Piotto, S. Hydroxylated Fatty Acids: The Role of the Sphingomyelin Synthase and the Origin of Selectivity. Membranes 2021, 11, 787. [Google Scholar] [CrossRef]
  17. Raichur, S. Ceramide Synthases Are Attractive Drug Targets for Treating Metabolic Diseases. Front. Endocrinol. 2020, 11, 483. [Google Scholar] [CrossRef]
  18. Gaggini, M.; Sabatino, L.; Vassalle, C. Conventional and innovative methods to assess oxidative stress biomarkers in the clinical cardiovascular setting. Biotechniques 2020, 68, 223–231. [Google Scholar] [CrossRef]
  19. Sokolowska, E.; Blachnio-Zabielska, A. The Role of Ceramides in Insulin Resistance. Front. Endocrinol. 2019, 10, 577. [Google Scholar] [CrossRef]
  20. Li, W.-h. Targeting Ceramides and Adiponectin Receptors in the Islet of Langerhans for Treating Diabetes. Molecules 2022, 27, 6117. [Google Scholar] [CrossRef]
  21. Ding, S.; Li, G.; Fu, T.; Zhang, T.; Lu, X.; Li, N.; Geng, Q. Ceramides and mitochondrial homeostasis. Cell Signal. 2024, 117, 111099. [Google Scholar] [CrossRef]
  22. Richardson, W.J.; Humphrey, S.B.; Sears, S.M.; Hoffman, N.A.; Orwick, A.J.; Doll, M.A.; Doll, C.L.; Xia, C.; Hernandez-Corbacho, M.; Snider, J.M.; et al. Expression of Ceramide Synthases in Mice and Their Roles in Regulating Acyl-Chain Sphingolipids: A Framework for Baseline Levels and Future Implications in Aging and Disease. Mol. Pharmacol. 2024, 105, 131–143. [Google Scholar] [CrossRef]
  23. Qian, H.; Deng, J.; Lu, C.; Hou, G.; Zhang, H.; Zhang, M.; Fang, Z.; Lv, X.-D. Ceramide synthases: Insights into the expression and prognosis of lung cancer. Exp. Lung Res. 2021, 47, 37–53. [Google Scholar] [CrossRef]
  24. Zeng, Y.; Fu, M.B. Sphingolipids and their carriers. Curr. Opin. Physiol. 2023, 34, 100671. [Google Scholar] [CrossRef]
  25. Aburasayn, H.; Al Batran, R.; Ussher, J.R. Targeting ceramide metabolism in obesity. Am. J. Physiol. Endocrinol. Metab. 2016, 311, E423–E435. [Google Scholar] [CrossRef]
  26. Hanamatsu, H.; Ohnishi, S.; Sakai, S.; Yuyama, K.; Mitsutake, S.; Takeda, H.; Hashino, S.; Igarashi, Y. Altered levels of serum sphingomyelin and ceramide containing distinct acyl chains in young obese adults. Nutr. Diabetes 2014, 4, e141. [Google Scholar] [CrossRef]
  27. Chathoth, S.; Ismail, M.H.; Alghamdi, H.M.; Zakaria, H.M.; Hassan, K.A.; Alshomimi, S.; Vatte, C.; Cyrus, C.; Alsaif, H.S.; Mostafa, A.; et al. Insulin resistance induced by de novo pathway-generated C16-ceramide is associated with type 2 diabetes in an obese population. Lipids Health Dis. 2022, 21, 24. [Google Scholar] [CrossRef]
  28. Hilvo, M.; Salonurmi, T.; Havulinna, A.S.; Kauhanen, D.; Pedersen, E.R.; Tell, G.S.; Meyer, K.; Teeriniemi, A.M.; Laatikainen, T.; Jousilahti, P.; et al. Ceramide stearic to palmitic acid ratio predicts incident diabetes. Diabetologia 2018, 61, 1424–1434. [Google Scholar] [CrossRef]
  29. Chaurasia, B.; Summers, S.A. Ceramides-Lipotoxic Inducers of Metabolic Disorders. Trends Endocrinol. Metab. 2015, 26, 538–550. [Google Scholar] [CrossRef]
  30. Ying, L.; Tippetts, T.S.; Chaurasia, B. Ceramide Dependent Lipotoxicity in Metabolic Diseases. Nutr. Healthy Aging 2019, 5, 1–12. [Google Scholar] [CrossRef]
  31. Hilvo, M.; Vasile, V.C.; Donato, L.J.; Hurme, R.; Laaksonen, R. Ceramides and Ceramide Scores: Clinical Applications for Cardiometabolic Risk Stratification. Front. Endocrinol. 2020, 11, 570628. [Google Scholar] [CrossRef]
  32. San Martin, R.; Brandao, C.F.C.; Junqueira-Franco, M.V.M.; Junqueira, G.P.; de Freitas, E.C.; de Carvalho, F.G.; Rodrigues, C.H.P.; Aguesse, A.; Billon-Crossouard, S.; Krempf, M.; et al. Untargeted lipidomic analysis of plasma from obese women submitted to combined physical exercise. Sci. Rep. 2022, 12, 11541. [Google Scholar] [CrossRef]
  33. Boutari, C.; Mantzoros, C.S. A 2022 update on the epidemiology of obesity and a call to action: As its twin COVID-19 pandemic appears to be receding, the obesity and dysmetabolism pandemic continues to rage on. Metabolism 2022, 133, 155217. [Google Scholar] [CrossRef]
  34. Landovská, P.; Karbanová, M. Social costs of obesity in the Czech Republic. Eur. J. Health Econ. 2023, 24, 1321–1341. [Google Scholar] [CrossRef] [PubMed]
  35. Yin, R.; Wang, X.; Li, K.; Yu, K.; Yang, L. Lipidomic profiling reveals distinct differences in plasma lipid composition in overweight or obese adolescent students. BMC Endocr. Disord. 2021, 21, 201. [Google Scholar] [CrossRef] [PubMed]
  36. Shafiee, A.; Nakhaee, Z.; Bahri, R.A.; Amini, M.J.; Salehi, A.; Jafarabady, K.; Seighali, N.; Rashidian, P.; Fathi, H.; Esmaeilpur Abianeh, F.; et al. Global prevalence of obesity and overweight among medical students: A systematic review and meta-analysis. BMC Public Health 2024, 24, 1673. [Google Scholar] [CrossRef]
  37. Bikman, B.T.; Summers, S.A. Ceramides as modulators of cellular and whole-body metabolism. J. Clin. Investig. 2011, 121, 4222–4230. [Google Scholar] [CrossRef] [PubMed]
  38. Auguet, T.; Bertran, L.; Capellades, J.; Abelló, S.; Aguilar, C.; Sabench, F.; del Castillo, D.; Correig, X.; Yanes, O.; Richart, C. LC/MS-Based Untargeted Metabolomics Analysis in Women with Morbid Obesity and Associated Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2023, 24, 7761. [Google Scholar] [CrossRef]
  39. Ali-Berrada, S.; Guitton, J.; Tan-Chen, S.; Gyulkhandanyan, A.; Hajduch, E.; Le Stunff, H. Circulating Sphingolipids and Glucose Homeostasis: An Update. Int. J. Mol. Sci. 2023, 24, 12720. [Google Scholar] [CrossRef]
  40. Chaurasia, B.; Summers, S.A. Ceramides in Metabolism: Key Lipotoxic Players. Annu. Rev. Physiol. 2021, 83, 303–330. [Google Scholar] [CrossRef]
  41. Hammad, S.M.; Lopes-Virella, M.F. Circulating Sphingolipids in Insulin Resistance, Diabetes and Associated Complications. Int. J. Mol. Sci. 2023, 24, 14015. [Google Scholar] [CrossRef] [PubMed]
  42. Iqbal, J.; Walsh, M.T.; Hammad, S.M.; Hussain, M.M. Sphingolipids and Lipoproteins in Health and Metabolic Disorders. Trends Endocrinol. Metab. 2017, 28, 506–518. [Google Scholar] [CrossRef] [PubMed]
  43. Parveen, F.; Bender, D.; Law, S.-H.; Mishra, V.K.; Chen, C.-C.; Ke, L.-Y. Role of Ceramidases in Sphingolipid Metabolism and Human Diseases. Cells 2019, 8, 1573. [Google Scholar] [CrossRef] [PubMed]
  44. Zaibaq, F.; Dowdy, T.; Larion, M. Targeting the Sphingolipid Rheostat in Gliomas. Int. J. Mol. Sci. 2022, 23, 9255. [Google Scholar] [CrossRef]
  45. Navone, S.E.; Guarnaccia, L.; Rizzaro, M.D.; Begani, L.; Barilla, E.; Alotta, G.; Garzia, E.; Caroli, M.; Ampollini, A.; Violetti, A.; et al. Role of Luteolin as Potential New Therapeutic Option for Patients with Glioblastoma through Regulation of Sphingolipid Rheostat. Int. J. Mol. Sci. 2024, 25, 130. [Google Scholar] [CrossRef]
  46. Piccoli, M.; Cirillo, F.; Ghiroldi, A.; Rota, P.; Coviello, S.; Tarantino, A.; La Rocca, P.; Lavota, I.; Creo, P.; Signorelli, P.; et al. Sphingolipids and Atherosclerosis: The Dual Role of Ceramide and Sphingosine-1-Phosphate. Antioxidants 2023, 12, 143. [Google Scholar] [CrossRef]
  47. Holland, W.L.; Miller, R.A.; Wang, Z.V.; Sun, K.; Barth, B.M.; Bui, H.H.; Davis, K.E.; Bikman, B.T.; Halberg, N.; Rutkowski, J.M.; et al. Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin. Nat. Med. 2011, 17, 55–63. [Google Scholar] [CrossRef]
  48. Juszczak, F.; Pierre, L.; Decarnoncle, M.; Jadot, I.; Martin, B.; Botton, O.; Caron, N.; Dehairs, J.; Swinnen, J.V.; Declèves, A.E. Sex differences in obesity-induced renal lipid accumulation revealed by lipidomics: A role of adiponectin/AMPK axis. Biol. Sex. Differ. 2023, 14, 63. [Google Scholar] [CrossRef]
  49. Garske, K.M.; Kar, A.; Comenho, C.; Balliu, B.; Pan, D.Z.; Bhagat, Y.V.; Rosenberg, G.; Koka, A.; Das, S.S.; Miao, Z.; et al. Increased body mass index is linked to systemic inflammation through altered chromatin co-accessibility in human preadipocytes. Nat. Commun. 2023, 14, 4214. [Google Scholar] [CrossRef]
  50. Huerta-Canseco, C.; Caba, M.; Camacho-Morales, A. Obesity-mediated Lipoinflammation Modulates Food Reward Responses. Neuroscience 2023, 529, 37–53. [Google Scholar] [CrossRef]
  51. Hildebrandt, X.; Ibrahim, M.; Peltzer, N. Cell death and inflammation during obesity: “Know my methods, WAT(son)”. Cell Death Differ. 2023, 30, 279–292. [Google Scholar] [CrossRef] [PubMed]
  52. Ofori, E.K.; Buabeng, A.; Amanquah, S.D.; Danquah, K.O.; Amponsah, S.K.; Dziedzorm, W.; Dogodzi, F.K.; Adusu-Donkor, L.X.; Bernard, S.K.; Asare-Anane, H. Effect of circulating ceramides on adiposity and insulin resistance in patients with type 2 diabetes: An observational cross-sectional study. Endocrinol. Diab Metab. 2023, 6, e418. [Google Scholar] [CrossRef] [PubMed]
  53. Broskey, N.T.; Obanda, D.N.; Burton, J.H.; Cefalu, W.T.; Ravussin, E. Skeletal muscle ceramides and daily fat oxidation in obesity and diabetes. Metabolism 2018, 82, 118–123. [Google Scholar] [CrossRef] [PubMed]
  54. Wigger, L.; Cruciani-Guglielmacci, C.; Nicolas, A.; Denom, J.; Fernandez, N.; Fumeron, F.; Marques-Vidal, P.; Ktorza, A.; Kramer, W.; Schulte, A.; et al. Plasma dihydroceramides are diabetes susceptibility biomarker candidates in mice and humans. Cell Rep. 2017, 18, 2269–2279. [Google Scholar] [CrossRef]
  55. Haus, J.M.; Kashyap, S.R.; Kasumov, T.; Zhang, R.; Kelly, K.R.; Defronzo, R.A.; Kirwan, J.P. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes 2009, 58, 337–343. [Google Scholar] [CrossRef]
  56. Boon, J.; Hoy, A.J.; Stark, R.; Brown, R.D.; Meex, R.C.; Henstridge, D.C.; Schenk, S.; Meikle, P.J.; Horowitz, J.F.; Kingwell, B.A.; et al. Ceramides contained in LDL are elevated in type 2 diabetes and promote inflammation and skeletal muscle insulin resistance. Diabetes 2013, 62, 401–410. [Google Scholar] [CrossRef]
  57. Galadari, S.; Rahman, A.; Pallichankandy, S.; Galadari, A.; Thayyullathil, F. Role of ceramide in diabetes mellitus: Evidence and mechanisms. Lipids Health Dis. 2013, 12, 98. [Google Scholar] [CrossRef]
  58. Hajduch, E.; Balendran, A.; Batty, I.H.; Litherland, G.J.; Blair, A.S.; Downes, C.P.; Hundal, H.S. Ceramide impairs the insulin-dependent membrane recruitment of protein kinase B leading to a loss in downstream signalling in L6 skeletal muscle cells. Diabetologia 2001, 44, 173–183. [Google Scholar] [CrossRef]
  59. Brozinick, J.T.; Hawkins, E.; Hoang Bui, H.; Kuo, M.S.; Tan, B.; Kievit, P.; Grove, K. Plasma sphingolipids are biomarkers of metabolic syndrome in non-human primates maintained on a Western-style diet. Int. J. Obes. 2013, 37, 1064–1070. [Google Scholar] [CrossRef]
  60. Sokołowska, E.; Car, H.; Szelachowska, M.; Adamska, A. Comparative characteristics of newly diagnosed adult diabetic patients: A pilot study. Acta Pol. Pharm. 2023, 80, 501–508. [Google Scholar] [CrossRef]
  61. Düsing, P.; Heinrich, N.N.; Al-Kassou, B.; Gutbrod, K.; Dörmann, P.; Nickenig, G.; Jansen, F.; Zietzer, A. Analysis of circulating ceramides and hexosylceramides in patients with coronary artery disease and type II diabetes mellitus. BMC Cardiovasc. Disord. 2023, 23, 454. [Google Scholar] [CrossRef] [PubMed]
  62. Guo, J.; Feng, J.; Qu, H.; Xu, H.; Zhou, H. Potential Drug Targets for Ceramide Metabolism in Cardiovascular Disease. J. Cardiovasc. Dev. Dis. 2022, 9, 434. [Google Scholar] [CrossRef] [PubMed]
  63. Di Cesare, M.; Perel, P.; Taylor, S.; Kabudula, C.; Bixby, H.; Gaziano, T.A.; McGhie, D.V.; Mwangi, J.; Pervan, B.; Narula, J.; et al. The Heart of the World. Glob. Heart 2024, 19, 11. [Google Scholar] [CrossRef]
  64. Alqahtani, B.A.; Alenazi, A.M. A national perspective on cardiovascular diseases in Saudi Arabia. BMC Cardiovasc. Disord. 2024, 24, 184. [Google Scholar] [CrossRef] [PubMed]
  65. He, L.; Zhang, M.; Zhao, Y.; Li, W.; Zhang, Y. Association between new Life’s Essential 8 and the risk of all-cause and cardiovascular mortality in patients with hypertension: A cohort study. BMC Public Health 2024, 24, 1730. [Google Scholar] [CrossRef]
  66. McGurk, K.A.; Keavney, B.D.; Nicolaou, A. Circulating ceramides as biomarkers of cardiovascular disease: Evidence from phenotypic and genomic studies. Atherosclerosis 2021, 327, 18–30. [Google Scholar] [CrossRef]
  67. Bosak, K.; Sauer, A.; Meeusen, J. Clinical Update: Ceramides As Novel Biomarkers of Cardiovascular Disease Risk. J. Nurse Pract. 2024, 20, 104838. [Google Scholar] [CrossRef]
  68. Choi, R.H.; Tatum, S.M.; Symons, J.D.; Summers, S.A.; Holland, W.L. Ceramides and other sphingolipids as drivers of cardiovascular disease. Nat. Rev. Cardiol. 2021, 18, 701–711. [Google Scholar] [CrossRef]
  69. Magaye, R.R.; Savira, F.; Hua, Y.; Kelly, D.J.; Reid, C.; Flynn, B.; Liew, D.; Wang, B.H. The role of dihydrosphingolipids in disease. Cell Mol. Life Sci. 2019, 76, 1107–1134. [Google Scholar] [CrossRef]
  70. Zietzer, A.; Düsing, P.; Reese, L.; Nickenig, G.; Jansen, F. Ceramide Metabolism in Cardiovascular Disease: A Network with High Therapeutic Potential. Arterioscler. Thromb. Vasc. Biol. 2022, 42, 1220–1228. [Google Scholar] [CrossRef]
  71. Brady, E.M.; Cao, T.H.; Moss, A.J.; Athithan, L.; Ayton, S.L.; Redman, E.; Argyridou, S.; Graham-Brown, M.P.M.; Maxwell, C.B.; Jones, D.J.L.; et al. Circulating sphingolipids and relationship to cardiac remodelling before and following a low-energy diet in asymptomatic Type 2 Diabetes. BMC Cardiovasc. Disord. 2024, 24, 25. [Google Scholar] [CrossRef]
  72. Sletten, A.C.; Peterson, L.R.; Schaffer, J.E. Manifestations and mechanisms of myocardial lipotoxicity in obesity. J. Intern. Med. 2018, 284, 478–491. [Google Scholar] [CrossRef] [PubMed]
  73. Slijkhuis, N.; Towers, M.; Mirzaian, M.; Korteland, S.A.; Heijs, B.; van Gaalen, K.; Nieuwenhuizen, I.; Nigg, A.; van der Heiden, K.; de Rijke, Y.B.; et al. Identifying lipid traces of atherogenic mechanisms in human carotid plaque. Atherosclerosis 2023, 385, 117340. [Google Scholar] [CrossRef] [PubMed]
  74. Law, S.H.; Chan, H.C.; Ke, G.M.; Kamatam, S.; Marathe, G.K.; Ponnusamy, V.K.; Ke, L.Y. Untargeted Lipidomic Profiling Reveals Lysophosphatidylcholine and Ceramide as Atherosclerotic Risk Factors in apolipoprotein E Knockout Mice. Int. J. Mol. Sci. 2023, 24, 6956. [Google Scholar] [CrossRef]
  75. Gruzdeva, O.; Dyleva, Y.; Belik, E.; Uchasova, E.; Ponasenko, A.; Ivanov, S.; Zinets, M.; Stasev, A.; Kutikhin, A.; Markova, V.; et al. Expression of Ceramide-Metabolizing Enzymes in the Heart Adipose Tissue of Cardiovascular Disease Patients. Int. J. Mol. Sci. 2023, 24, 9494. [Google Scholar] [CrossRef] [PubMed]
  76. Shoghli, M.; Lokki, A.I.; Lääperi, M.; Sinisalo, J.; Lokki, M.L.; Hilvo, M.; Jylhä, A.; Tuomilehto, J.; Laaksonen, R. The Novel Ceramide- and Phosphatidylcholine-Based Risk Score for the Prediction of New-Onset of Hypertension. J. Clin. Med. 2023, 12, 7524. [Google Scholar] [CrossRef]
  77. Yu, J.; Pan, W.; Shi, R.; Yang, T.; Li, Y.; Yu, G.; Bai, Y.; Schuchman, E.H.; He, X.; Zhang, G. Ceramide is upregulated and associated with mortality in patients with chronic heart failure. Can. J. Cardiol. 2015, 31, 357–363. [Google Scholar] [CrossRef]
  78. Havulinna, A.S.; Sysi-Aho, M.; Hilvo, M.; Kauhanen, D.; Hurme, R.; Ekroos, K.; Salomaa, V.; Laaksonen, R. Circulating Ceramides Predict Cardiovascular Outcomes in the Population-Based FINRISK 2002 Cohort. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 2424–2430. [Google Scholar] [CrossRef]
  79. Rigamonti, A.E.; Dei Cas, M.; Caroli, D.; Bondesan, A.; Cella, S.G.; Paroni, R.; Sartorio, A. Ceramide Risk Score in the Evaluation of Metabolic Syndrome: An Additional or Substitutive Biochemical Marker in the Clinical Practice? Int. J. Mol. Sci. 2023, 24, 12452. [Google Scholar] [CrossRef]
  80. Yin, W.; Li, F.; Tan, X.; Wang, H.; Jiang, W.; Wang, X.; Li, S.; Zhang, Y.; Han, Q.; Wang, Y.; et al. Plasma Ceramides and Cardiovascular Events in Hypertensive Patients at High Cardiovascular Risk. Am. J. Hypertens. 2021, 34, 1209–1216. [Google Scholar] [CrossRef]
  81. Nicholls, M. Plasma ceramides and cardiac risk. Eur. Heart J. 2017, 38, 1359–1360. [Google Scholar] [CrossRef]
  82. Wretlind, A.; Curovic, V.R.; Suvitaival, T.; Theilade, S.; Tofte, N.; Winther, S.A.; Vilsbøll, T.; Vestergaard, H.; Rossing, P.; Legido-Quigley, C. Ceramides as Risk Markers for Future Cardiovascular Events and All-Cause Mortality in Long-standing Type 1 Diabetes. Diabetes 2023, 72, 1493–1501. [Google Scholar] [CrossRef]
  83. Laaksonen, R.; Ekroos, K.; Sysi-Aho, M.; Hilvo, M.; Vihervaara, T.; Kauhanen, D.; Suoniemi, M.; Hurme, R.; März, W.; Scharnagl, H.; et al. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur. Heart J. 2016, 37, 1967–1976. [Google Scholar] [CrossRef]
  84. Meeusen, J.W.; Donato, L.J.; Bryant, S.C.; Baudhuin, L.M.; Berger, P.B.; Jaffe, A.S. Plasma Ceramides. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 1933–1939. [Google Scholar] [CrossRef] [PubMed]
  85. Liang, H.; Li, F.; Zhang, L.; Li, L.; Guo, B. Ceramides and pro-inflammatory cytokines for the prediction of acute coronary syndrome: A multi-marker approach. BMC Cardiovasc. Disord. 2024, 24, 47. [Google Scholar] [CrossRef]
  86. Drucker, D.J. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef]
  87. Coutinho, W.; Halpern, B. Pharmacotherapy for obesity: Moving towards efficacy improvement. Diabetol. Metab. Syndr. 2024, 16, 6. [Google Scholar] [CrossRef] [PubMed]
  88. Pahwa, R.; Goyal, A.; Jialal, I. Chronic Inflammation. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  89. Jenkins, R.W.; Canals, D.; Idkowiak-Baldys, J.; Simbari, F.; Roddy, P.; Perry, D.M.; Kitatani, K.; Luberto, C.; Hannun, Y.A. Regulated secretion of acid sphingomyelinase: Implications for selectivity of ceramide formation. J. Biol. Chem. 2010, 285, 35706–35718. [Google Scholar] [CrossRef] [PubMed]
  90. Nixon, G.F. Sphingolipids in inflammation: Pathological implications and potential therapeutic targets. Br. J. Pharmacol. 2009, 158, 982–993. [Google Scholar] [CrossRef]
  91. Schilling, J.D.; Machkovech, H.M.; He, L.; Sidhu, R.; Fujiwara, H.; Weber, K.; Ory, D.S.; Schaffer, J.E. Palmitate and lipopolysaccharide trigger synergistic ceramide production in primary macrophages. J. Biol. Chem. 2013, 288, 2923–2932. [Google Scholar] [CrossRef]
  92. Sims, K.; Haynes, C.A.; Kelly, S.; Allegood, J.C.; Wang, E.; Momin, A.; Leipelt, M.; Reichart, D.; Glass, C.K.; Sullards, M.C.; et al. Kdo2-lipid A, a TLR4-specific agonist, induces de novo sphingolipid biosynthesis in RAW264.7 macrophages, which is essential for induction of autophagy. J. Biol. Chem. 2010, 285, 38568–38579. [Google Scholar] [CrossRef] [PubMed]
  93. Dennis, E.A.; Deems, R.A.; Harkewicz, R.; Quehenberger, O.; Brown, H.A.; Milne, S.B.; Myers, D.S.; Glass, C.K.; Hardiman, G.; Reichart, D.; et al. A mouse macrophage lipidome. J. Biol. Chem. 2010, 285, 39976–39985. [Google Scholar] [CrossRef] [PubMed]
  94. Kim, J.K. Fat uses a TOLL-road to connect inflammation and diabetes. Cell Metab. 2006, 4, 417–419. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The three main pathways of ceramide biosynthesis. The de novo synthesis is a multistep pathway that starts with the condensation of serine with palmitoyl-CoA to form 3-ketosphinganine, which is the rate-limiting step and is catalyzed by serine palmitoyltransferase. In the second pathway, Cers are synthesized via the hydrolysis of the phosphocholine head groups of sphingomyelin by sphingomyelinases. In the salvage pathway, Cers are recycled from sphingosine re-acylation that is catalyzed by ceramide synthase.
Figure 1. The three main pathways of ceramide biosynthesis. The de novo synthesis is a multistep pathway that starts with the condensation of serine with palmitoyl-CoA to form 3-ketosphinganine, which is the rate-limiting step and is catalyzed by serine palmitoyltransferase. In the second pathway, Cers are synthesized via the hydrolysis of the phosphocholine head groups of sphingomyelin by sphingomyelinases. In the salvage pathway, Cers are recycled from sphingosine re-acylation that is catalyzed by ceramide synthase.
Diseases 12 00195 g001
Figure 2. Sphingolipid rheostat and the opposite effects of ceramides and S1P on cell fate.
Figure 2. Sphingolipid rheostat and the opposite effects of ceramides and S1P on cell fate.
Diseases 12 00195 g002
Table 1. Substrate specificity and tissue distribution of ceramide synthase isoforms.
Table 1. Substrate specificity and tissue distribution of ceramide synthase isoforms.
Ceramide Synthase IsoformsAcyl Chain-Length SpecificityTissue Distribution
CerS1C18Muscle tissues, brain, testis, gastrointestinal tract,
female tissues, bone marrow, and lymphoid tissues
CerS2C22-24Mainly expressed in liver, kidney, and small intestine
CerS3Broad substrate specificity toward medium- to long-chain fatty acidsEndocrine tissues, gastrointestinal tract, male tissues,
and female tissues, especially in esophagus, testis, and skin
CerS4C20-C24Nearly all tissues
CerS5C16All tissues except heart muscle and smooth muscle,
especially in adipose tissue
CerS6C16Most tissues, especially in brain and adipose tissue
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Delcheva, G.; Stefanova, K.; Stankova, T. Ceramides—Emerging Biomarkers of Lipotoxicity in Obesity, Diabetes, Cardiovascular Diseases, and Inflammation. Diseases 2024, 12, 195. https://doi.org/10.3390/diseases12090195

AMA Style

Delcheva G, Stefanova K, Stankova T. Ceramides—Emerging Biomarkers of Lipotoxicity in Obesity, Diabetes, Cardiovascular Diseases, and Inflammation. Diseases. 2024; 12(9):195. https://doi.org/10.3390/diseases12090195

Chicago/Turabian Style

Delcheva, Ginka, Katya Stefanova, and Teodora Stankova. 2024. "Ceramides—Emerging Biomarkers of Lipotoxicity in Obesity, Diabetes, Cardiovascular Diseases, and Inflammation" Diseases 12, no. 9: 195. https://doi.org/10.3390/diseases12090195

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop