1
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025; 22:399-413. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Yan J, Han VX, Jones HF, Couttas TA, Jieu B, Leweke FM, Lee J, Loi C, Webster R, Kothur K, Menezes MP, Antony J, Kandula T, Cardamone M, Patel S, Bandodkar S, Dale RC. Cerebrospinal fluid metabolomics in autistic regression reveals dysregulation of sphingolipids and decreased β-hydroxybutyrate. EBioMedicine 2025; 114:105664. [PMID: 40138886 PMCID: PMC11986237 DOI: 10.1016/j.ebiom.2025.105664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Autism is highly heritable, however actionable genetic findings are only found in a minority of patients. Many people with autism suffer loss of neurodevelopmental skills, known as autistic regression. The cause of regression is poorly understood, and the diagnostic and therapeutic pathways are lacking. METHODS We used untargeted metabolomics using a UPLC-Q-Exactive-HFx Mass Spectrometry to examine cerebrospinal fluid (CSF) from twenty-two patients with autistic regression compared to sixteen controls with neurodevelopmental disorders (but not autistic regression) and thirty-four controls with other neurological disease (headache, encephalitis, epilepsy). The twenty-two patients with autistic regression consisted of two groups: early (infantile) autistic regression <2 years of age (n = 8), and later regression of skills >4 years of age, often in the context of pre-existing developmental concerns (n = 14). Metabolites of interest were then quantified and validated using targeted assays. FINDINGS Untargeted case-control studies revealed good separation of patients from controls using multivariate analysis. β-hydroxybutyrate was significantly decreased in the CSF of patients with autistic regression, and the findings were validated using a targeted β-hydroxybutyrate assay. The sphingolipid, sphingosine-1-phosphate was significantly elevated in the discovery case-control studies, and sphingolipid metabolism pathways were also significantly dysregulated. We therefore developed a targeted metabolite assay of forty sphingolipids. After FDR correction, 21 of the 40 sphingolipids were significantly dysregulated (pFDR < 0.05) (Benjamini-Hochberg correction) in autistic regression compared to the neurodevelopmental controls, and 26 of the 40 sphingolipids were significantly dysregulated in autistic regression compared to other neurological controls, with elevated ceramides, hexosylceramides, sphingosines (including sphingosine-1-phosphate), and sulfatides. By contrast, sphingomyelin levels were generally decreased in autistic regression. INTERPRETATION Our data shows the potential utility of CSF metabolomics in the context of autistic regression, a clinical syndrome which has historically lacked pathophysiological biomarkers and disease modifying therapies. FUNDING Financial support for the study was granted by Dale NHMRC Investigator grant APP1193648, Petre Foundation, Cerebral Palsy Alliance, and Ainsworth and SCHF Neuroscience grant scheme.
Collapse
Affiliation(s)
- Jingya Yan
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Velda X Han
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hannah F Jones
- Starship Hospital, Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Timothy A Couttas
- Neuroscience Research Australia, Randwick, NSW, Australia; Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - Beverly Jieu
- Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - F Markus Leweke
- Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - Jennifer Lee
- Department of Endocrinology, The Children's Hospital at Westmead, NSW, Australia
| | - Catherine Loi
- Department of Endocrinology, The Children's Hospital at Westmead, NSW, Australia
| | - Richard Webster
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Kavitha Kothur
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Manoj P Menezes
- Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Jayne Antony
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Tejaswi Kandula
- Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia
| | - Michael Cardamone
- Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia
| | - Shrujna Patel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Sushil Bandodkar
- Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Department of Biochemistry, The Children's Hospital at Westmead, NSW, Australia
| | - Russell C Dale
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia.
| |
Collapse
|
3
|
Williams KJ. Inflammation in atherosclerosis: a Big Idea that has underperformed so far. Curr Opin Lipidol 2025; 36:78-87. [PMID: 39846349 PMCID: PMC11888836 DOI: 10.1097/mol.0000000000000973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
PURPOSE OF REVIEW For many years, inflammation has been a major concept in basic research on atherosclerosis and in the development of potential diagnostic tools and treatments. The purpose of this review is to assess the performance of this concept with an emphasis on recent clinical trials. In addition, contemporary literature may help identify new therapeutic targets, particularly in the context of the treatment of early, rather than end-stage, arterial disease. RECENT FINDINGS Newly reported clinical trials cast doubt on the efficacy of colchicine, the sole anti-inflammatory agent currently approved for use in patients with atherosclerotic cardiovascular disease (ASCVD). New analyses also challenge the hypothesis that residual ASCVD event risk after optimal management of lipids, blood pressure, and smoking arises primarily from residual inflammatory risk. Current clinical practice to initiate interventions so late in the course of atherosclerotic arterial disease may be a better explanation. Lipid-lowering therapy in early atherosclerosis, possibly combined with novel add-on agents to specifically accelerate resolution of maladaptive inflammation, may be more fruitful than the conventional approach of testing immunosuppressive strategies in end-stage arterial disease. Also discussed is the ongoing revolution in noninvasive technologies to image the arterial wall. These technologies are changing screening, diagnosis, and treatment of atherosclerosis, including early and possibly reversable disease. SUMMARY The burden of proof that the Big Idea of inflammation in atherosclerosis has clinical value remains the responsibility of its advocates. This responsibility requires convincing trial data but still seems largely unmet. Unfortunately, the focus on inflammation as the source of residual ASCVD event risk has distracted us from the need to screen and treat earlier.
Collapse
Affiliation(s)
- Kevin Jon Williams
- Department of Cardiovascular Sciences and Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Seal A, Hughes M, Wei F, Pugazhendhi AS, Ngo C, Ruiz J, Schwartzman JD, Coathup MJ. Sphingolipid-Induced Bone Regulation and Its Emerging Role in Dysfunction Due to Disease and Infection. Int J Mol Sci 2024; 25:3024. [PMID: 38474268 PMCID: PMC10932382 DOI: 10.3390/ijms25053024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
The human skeleton is a metabolically active system that is constantly regenerating via the tightly regulated and highly coordinated processes of bone resorption and formation. Emerging evidence reveals fascinating new insights into the role of sphingolipids, including sphingomyelin, sphingosine, ceramide, and sphingosine-1-phosphate, in bone homeostasis. Sphingolipids are a major class of highly bioactive lipids able to activate distinct protein targets including, lipases, phosphatases, and kinases, thereby conferring distinct cellular functions beyond energy metabolism. Lipids are known to contribute to the progression of chronic inflammation, and notably, an increase in bone marrow adiposity parallel to elevated bone loss is observed in most pathological bone conditions, including aging, rheumatoid arthritis, osteoarthritis, and osteomyelitis. Of the numerous classes of lipids that form, sphingolipids are considered among the most deleterious. This review highlights the important primary role of sphingolipids in bone homeostasis and how dysregulation of these bioactive metabolites appears central to many chronic bone-related diseases. Further, their contribution to the invasion, virulence, and colonization of both viral and bacterial host cell infections is also discussed. Many unmet clinical needs remain, and data to date suggest the future use of sphingolipid-targeted therapy to regulate bone dysfunction due to a variety of diseases or infection are highly promising. However, deciphering the biochemical and molecular mechanisms of this diverse and extremely complex sphingolipidome, both in terms of bone health and disease, is considered the next frontier in the field.
Collapse
Affiliation(s)
- Anouska Seal
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
| | - Megan Hughes
- School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK;
| | - Fei Wei
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Abinaya S. Pugazhendhi
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Christopher Ngo
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | | | - Melanie J. Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| |
Collapse
|
5
|
Risner ML, Ribeiro M, McGrady NR, Kagitapalli BS, Chamling X, Zack DJ, Calkins DJ. Neutral sphingomyelinase inhibition promotes local and network degeneration in vitro and in vivo. Cell Commun Signal 2023; 21:305. [PMID: 37904133 PMCID: PMC10614343 DOI: 10.1186/s12964-023-01291-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/22/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Cell-to-cell communication is vital for tissues to respond, adapt, and thrive in the prevailing milieu. Several mechanisms mediate intercellular signaling, including tunneling nanotubes, gap junctions, and extracellular vesicles (EV). Depending on local and systemic conditions, EVs may contain cargoes that promote survival, neuroprotection, or pathology. Our understanding of pathologic intercellular signaling has been bolstered by disease models using neurons derived from human pluripotent stems cells (hPSC). METHODS Here, we used hPSC-derived retinal ganglion cells (hRGC) and the mouse visual system to investigate the influence of modulating EV generation on intercellular trafficking and cell survival. We probed the impact of EV modulation on cell survival by decreasing the catabolism of sphingomyelin into ceramide through inhibition of neutral sphingomyelinase (nSMase), using GW4869. We assayed for cell survival in vitro by probing for annexin A5, phosphatidylserine, viable mitochondria, and mitochondrial reactive oxygen species. In vivo, we performed intraocular injections of GW4869 and measured RGC and superior colliculus neuron density and RGC anterograde axon transport. RESULTS Following twenty-four hours of dosing hRGCs with GW4869, we found that inhibition of nSMase decreased ceramide and enhanced GM1 ganglioside accumulation. This inhibition also reduced the density of small EVs, increased the density of large EVs, and enriched the pro-apoptotic protein, annexin A5. Reducing nSMase activity increased hRGC apoptosis initiation due to enhanced density and uptake of apoptotic particles, as identified by the annexin A5 binding phospholipid, phosphatidylserine. We assayed intercellular trafficking of mitochondria by developing a coculture system of GW4869-treated and naïve hRGCs. In treated cells, inhibition of nSMase reduced the number of viable mitochondria, while driving mitochondrial reactive oxygen species not only in treated, but also in naive hRGCs added in coculture. In mice, 20 days following a single intravitreal injection of GW4869, we found a significant loss of RGCs and their axonal recipient neurons in the superior colliculus. This followed a more dramatic reduction in anterograde RGC axon transport to the colliculus. CONCLUSION Overall, our data suggest that perturbing the physiologic catabolism of sphingomyelin by inhibiting nSMase reorganizes plasma membrane associated sphingolipids, alters the profile of neuron-generated EVs, and promotes neurodegeneration in vitro and in vivo by shifting the balance of pro-survival versus -degenerative EVs. Video Abstract.
Collapse
Affiliation(s)
- Michael L Risner
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave S., Nashville, TN, 37232, USA.
- Department of Foundational Medical Studies, Eye Research Center, Oakland University William Beaumont School of Medicine, 369 Dodge Hall, 118 Library Dr., Rochester, MI, 48309, USA.
| | - Marcio Ribeiro
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave S., Nashville, TN, 37232, USA
| | - Nolan R McGrady
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave S., Nashville, TN, 37232, USA
| | - Bhanu S Kagitapalli
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave S., Nashville, TN, 37232, USA
| | - Xitiz Chamling
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Donald J Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave S., Nashville, TN, 37232, USA.
| |
Collapse
|
6
|
Jiang S, Yang H, Sun Z, Zhang Y, Li Y, Li J. The basis of complications in the context of SARS-CoV-2 infection: Pathological activation of ADAM17. Biochem Biophys Res Commun 2023; 679:37-46. [PMID: 37666046 DOI: 10.1016/j.bbrc.2023.08.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
The virulence of SARS-CoV-2 decreases with increasing infectivity, the primary approaches for antiviral treatments will be preventing or minimizing the complications resulting from virus infection. ADAM metallopeptidase domain 17 (ADAM17) activation by SARS-CoV-2 infection has a dual effect on the development of the disease: increased release of inflammatory cytokines and dysregulation of Angiotensin converting enzyme II (ACE2) on cell surfaces, inflammatory cytokine infiltration and loss of ACE2 protective function lead to a significant increase in the incidence of related complications. Importantly, pathologically activated ADAM17 showed superior features than S protein in regulating ACE2 expression and participating in the intra cellular replication of SARS-CoV-2. In short, SARS-CoV-2 elicits only a limited immune response when it promotes its own replication and pathogenicity through ADAM17. Therefore, the pathological activation of ADAM17 may also represent a diminished innate antiviral defense and an altered strategy of SARS-CoV-2 infection. In this review, we summarized recent advances in our understanding of the pathophysiology of ADAM17, with a focus on the new findings that SARS-CoV-2 affects ADAM17 expression through Furin protein converting enzyme and Mitogen-activated protein kinase (MAPK) pathway, and raises the hypothesis that SARS-CoV-2 may mediates the pathological activation of ADAM17 by hijacking the actin regulatory pathway, and discussed the underlying biological principles.
Collapse
Affiliation(s)
| | - Hao Yang
- Zunyi Medical University Guizhou, China
| | | | - Yi Zhang
- Zunyi Medical University Guizhou, China
| | - Yan Li
- Zunyi Medical University Guizhou, China
| | - Jida Li
- Zunyi Medical University Guizhou, China; Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi, Guizhou, China.
| |
Collapse
|
7
|
Pfrieger FW. The Niemann-Pick type diseases – A synopsis of inborn errors in sphingolipid and cholesterol metabolism. Prog Lipid Res 2023; 90:101225. [PMID: 37003582 DOI: 10.1016/j.plipres.2023.101225] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Disturbances of lipid homeostasis in cells provoke human diseases. The elucidation of the underlying mechanisms and the development of efficient therapies represent formidable challenges for biomedical research. Exemplary cases are two rare, autosomal recessive, and ultimately fatal lysosomal diseases historically named "Niemann-Pick" honoring the physicians, whose pioneering observations led to their discovery. Acid sphingomyelinase deficiency (ASMD) and Niemann-Pick type C disease (NPCD) are caused by specific variants of the sphingomyelin phosphodiesterase 1 (SMPD1) and NPC intracellular cholesterol transporter 1 (NPC1) or NPC intracellular cholesterol transporter 2 (NPC2) genes that perturb homeostasis of two key membrane components, sphingomyelin and cholesterol, respectively. Patients with severe forms of these diseases present visceral and neurologic symptoms and succumb to premature death. This synopsis traces the tortuous discovery of the Niemann-Pick diseases, highlights important advances with respect to genetic culprits and cellular mechanisms, and exposes efforts to improve diagnosis and to explore new therapeutic approaches.
Collapse
|
8
|
Effect of Sphingomyelinase-Treated LDLs on HUVECs. Molecules 2023; 28:molecules28052100. [PMID: 36903354 PMCID: PMC10004656 DOI: 10.3390/molecules28052100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/12/2023] Open
Abstract
Low-density lipoproteins (LDLs) exert a key role in the transport of esterified cholesterol to tissues. Among the atherogenic modifications of LDLs, the oxidative modification has been mainly investigated as a major risk factor for accelerating atherogenesis. Since LDL sphingolipids are also emerging as important regulators of the atherogenic process, increasing attention is devoted to the effects of sphingomyelinase (SMase) on LDL structural and atherogenic properties. The aims of the study were to investigate the effect of SMase treatment on the physical-chemical properties of LDLs. Moreover, we evaluated cell viability, apoptosis, and oxidative and inflammatory status in human umbilical vein endothelial cells (HUVECs) treated with either ox-LDLs or SMase-treated LDLs (SMase-LDLs). Both treatments were associated with the accrual of the intracellular ROS and upregulation of the antioxidant Paraoxonase 2 (PON2), while only SMase-LDLs induced an increase of superoxide dismutase 2 (SOD2), suggesting the activation of a feedback loop to restrain the detrimental effects of ROS. The increased caspase-3 activity and reduced viability observed in cells treated with SMase-LDLs and ox-LDLs suggest a pro-apoptotic effect of these modified lipoproteins on endothelial cells. Moreover, a strong proinflammatory effect of SMase-LDLs compared to ox-LDLs was confirmed by an increased activation of NF-κB and consequent increased expression of its downstream cytokines IL-8 and IL-6 in HUVECs.
Collapse
|
9
|
Olsson K, Cheng AJ, Al-Ameri M, Tardif N, Melin M, Rooyackers O, Lanner JT, Westerblad H, Gustafsson T, Bruton JD, Rullman E. Sphingomyelinase activity promotes atrophy and attenuates force in human muscle fibres and is elevated in heart failure patients. J Cachexia Sarcopenia Muscle 2022; 13:2551-2561. [PMID: 35852046 PMCID: PMC9530516 DOI: 10.1002/jcsm.13029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/26/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Activation of sphingomyelinase (SMase) as a result of a general inflammatory response has been implicated as a mechanism underlying disease-related loss of skeletal muscle mass and function in several clinical conditions including heart failure. Here, for the first time, we characterize the effects of SMase activity on human muscle fibre contractile function and assess skeletal muscle SMase activity in heart failure patients. METHODS The effects of SMase on force production and intracellular Ca2+ handling were investigated in single intact human muscle fibres. Additional mechanistic studies were performed in single mouse toe muscle fibres. RNA sequencing was performed in human muscle bundles exposed to SMase. Intramuscular SMase activity was measured from heart failure patients (n = 61, age 69 ± 0.8 years, NYHA III-IV, ejection fraction 25 ± 1.0%, peak VO2 14.4 ± 0.6 mL × kg × min) and healthy age-matched control subjects (n = 10, age 71 ± 2.2 years, ejection fraction 60 ± 1.2%, peak VO2 25.8 ± 1.1 mL × kg × min). SMase activity was related to circulatory factors known to be associated with progression and disease severity in heart failure. RESULTS Sphingomyelinase reduced muscle fibre force production (-30%, P < 0.05) by impairing sarcoplasmic reticulum (SR) Ca2+ release (P < 0.05) and reducing myofibrillar Ca2+ sensitivity. In human muscle bundles exposed to SMase, RNA sequencing analysis revealed 180 and 291 genes as up-regulated and down-regulated, respectively, at a FDR of 1%. Gene-set enrichment analysis identified 'proteasome degradation' as an up-regulated pathway (average fold-change 1.1, P = 0.008), while the pathway 'cytoplasmic ribosomal proteins' (average fold-change 0.8, P < 0.0001) and factors involving proliferation of muscle cells (average fold-change 0.8, P = 0.0002) where identified as down-regulated. Intramuscular SMase activity was ~20% higher (P < 0.05) in human heart failure patients than in age-matched healthy controls and was positively correlated with markers of disease severity and progression, and with several circulating inflammatory proteins, including TNF-receptor 1 and 2. In a longitudinal cohort of heart failure patients (n = 6, mean follow-up time 2.5 ± 0.2 years), SMase activity was demonstrated to increase by 30% (P < 0.05) with duration of disease. CONCLUSIONS The present findings implicate activation of skeletal muscle SMase as a mechanism underlying human heart failure-related loss of muscle mass and function. Moreover, our findings strengthen the idea that SMase activation may underpin disease-related loss of muscle mass and function in other clinical conditions, acting as a common patophysiological mechanism for the myopathy often reported in diseases associated with a systemic inflammatory response.
Collapse
Affiliation(s)
- Karl Olsson
- Department of Laboratory Medicine, Section of Clinical Physiology, Karolinska Institutet, Huddinge, Sweden
| | - Arthur J Cheng
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Solna, Sweden.,Muscle Health Research Centre, School of Kinesiology and Health Science, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Mamdoh Al-Ameri
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Nicolas Tardif
- Division of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Huddinge, Sweden.,Anesthesiology and intensive care, Department of Clinical Science Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Michael Melin
- Department of Laboratory Medicine, Section of Clinical Physiology, Karolinska Institutet, Huddinge, Sweden
| | - Olav Rooyackers
- Division of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Huddinge, Sweden.,Anesthesiology and intensive care, Department of Clinical Science Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Solna, Sweden
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Solna, Sweden
| | - Thomas Gustafsson
- Department of Laboratory Medicine, Section of Clinical Physiology, Karolinska Institutet and Department of Clinical Physiology Karolinska Univ Hospital, Huddinge, Sweden
| | - Joseph D Bruton
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Solna, Sweden
| | - Eric Rullman
- Department of Laboratory Medicine, Section of Clinical Physiology, Karolinska Institutet and Department of Clinical Physiology Karolinska Univ Hospital, Huddinge, Sweden
| |
Collapse
|
10
|
Artru F, McPhail MJW, Triantafyllou E, Trovato FM. Lipids in Liver Failure Syndromes: A Focus on Eicosanoids, Specialized Pro-Resolving Lipid Mediators and Lysophospholipids. Front Immunol 2022; 13:867261. [PMID: 35432367 PMCID: PMC9008479 DOI: 10.3389/fimmu.2022.867261] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/08/2022] [Indexed: 12/30/2022] Open
Abstract
Lipids are organic compounds insoluble in water with a variety of metabolic and non-metabolic functions. They not only represent an efficient energy substrate but can also act as key inflammatory and anti-inflammatory molecules as part of a network of soluble mediators at the interface of metabolism and the immune system. The role of endogenous bioactive lipid mediators has been demonstrated in several inflammatory diseases (rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, cancer). The liver is unique in providing balanced immunotolerance to the exposure of bacterial components from the gut transiting through the portal vein and the lymphatic system. This balance is abruptly deranged in liver failure syndromes such as acute liver failure and acute-on-chronic liver failure. In these syndromes, researchers have recently focused on bioactive lipid mediators by global metabonomic profiling and uncovered the pivotal role of these mediators in the immune dysfunction observed in liver failure syndromes explaining the high occurrence of sepsis and subsequent organ failure. Among endogenous bioactive lipids, the mechanistic actions of three classes (eicosanoids, pro-resolving lipid mediators and lysophospholipids) in the pathophysiological modulation of liver failure syndromes will be the topic of this narrative review. Furthermore, the therapeutic potential of lipid-immune pathways will be described.
Collapse
Affiliation(s)
- Florent Artru
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Mark J W McPhail
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | | |
Collapse
|
11
|
Li J, Zheng X, Li X, Yang J, Liu W, Yang L, Liu B. Study on the protective effect and mechanism of Liriodendrin on radiation enteritis in mice. JOURNAL OF RADIATION RESEARCH 2022; 63:213-220. [PMID: 35059715 PMCID: PMC8944324 DOI: 10.1093/jrr/rrab128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/11/2021] [Indexed: 06/01/2023]
Abstract
Patients receiving pelvic or abdominal radiotherapy may experience acute and/or chronic side effects due to gastrointestinal changes. However, effective medicine for treating radiation enteritis has not been found yet. Sargentodoxa cuneata is a famous Chinese medicine used to treat intestinal inflammation, and our research team has found the main biologically active compound through its extraction, which is Liriodendrin. In this study, we found that Liriodendrin can reduce the expression of Cer, Cer1P and S1P in the sphingolipid pathway, thereby reducing the histological damage to the intestinal tract of mice and inhibiting the apoptosis of intestinal tissue cells. In addition, Liriodendrin can reduce the levels of pro-inflammatory cytokines (IL-6 and TNF-α), and it is suggested through flow cytometry that the proportion of neutrophils in the intestinal tissue can decrease due to the existence of Liriodendrin. At the same time, the western blot evaluation revealed that Liriodendrin significantly inhibited the activation of Bcl-2/Bax/Caspase-3 and NF-κB signaling pathways. The results show that Liriodendrin can inhibit intestinal inflammation and intestinal cell apoptosis through the sphingolipid pathway. Therefore, the aforementioned results demonstrated that Liriodendrin may be a promising drug for the treatment of radiation enteritis.
Collapse
Affiliation(s)
| | | | - Xiong Li
- Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610051, China
| | - Jing Yang
- The School of Biological Science and Technology, Chengdu Medical College, 610083, Chengdu, China
| | - Wei Liu
- Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610051, China
| | - Lei Yang
- Tianjin key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Bin Liu
- Corresponding author. Bin Liu, Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China. E-mail: ; Tel: +86-13980823937; Fax: +86-028-84771387
| |
Collapse
|
12
|
The acid sphingomyelinase/ceramide system in COVID-19. Mol Psychiatry 2022; 27:307-314. [PMID: 34608263 PMCID: PMC8488928 DOI: 10.1038/s41380-021-01309-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 08/10/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Acid sphingomyelinase (ASM) cleaves sphingomyelin into the highly lipophilic ceramide, which forms large gel-like rafts/platforms in the plasma membrane. We showed that SARS-CoV-2 uses these platforms for cell entry. Lowering the amount of ceramide or ceramide blockade due to inhibitors of ASM, genetic downregulation of ASM, anti-ceramide antibodies or degradation by neutral ceramidase protected against infection with SARS-CoV-2. The addition of ceramide restored infection with SARS-CoV-2. Many clinically approved medications functionally inhibit ASM and are called FIASMAs (functional inhibitors of acid sphingomyelinase). The FIASMA fluvoxamine showed beneficial effects on COVID-19 in a randomized prospective study and a prospective open-label real-world study. Retrospective and observational studies showed favorable effects of FIASMA antidepressants including fluoxetine, and the FIASMA hydroxyzine on the course of COVID-19. The ASM/ceramide system provides a framework for a better understanding of the infection of cells by SARS-CoV-2 and the clinical, antiviral, and anti-inflammatory effects of functional inhibitors of ASM. This framework also supports the development of new drugs or the repurposing of "old" drugs against COVID-19.
Collapse
|
13
|
Sasset L, Di Lorenzo A. Sphingolipid Metabolism and Signaling in Endothelial Cell Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:87-117. [PMID: 35503177 DOI: 10.1007/978-981-19-0394-6_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The endothelium, inner layer of blood vessels, constitutes a metabolically active paracrine, endocrine, and autocrine organ, able to sense the neighboring environment and exert a variety of biological functions important to preserve the health of vasculature, tissues, and organs. Sphingolipids are both fundamental structural components of the eukaryotic membranes and signaling molecules regulating a variety of biological functions. Ceramide and sphingosine-1-phosphate (S1P), bioactive sphingolipids, have emerged as important regulators of cardiovascular functions in health and disease. In this review we discuss recent insights into the role of ceramide and S1P biosynthesis and signaling in regulating endothelial cell functions, in health and diseases. We also highlight advances into the mechanisms regulating serine palmitoyltransferase, the first and rate-limiting enzyme of de novo sphingolipid biosynthesis, with an emphasis on its inhibitors, ORMDL and NOGO-B. Understanding the molecular mechanisms regulating the sphingolipid de novo biosynthesis may provide the foundation for therapeutic modulation of this pathway in a variety of conditions, including cardiovascular diseases, associated with derangement of this pathway.
Collapse
Affiliation(s)
- Linda Sasset
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Feil Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Lin HC, Chiang HP, Jiang WP, Lan YH, Huang GJ, Hsieh MT, Kuo SC, Lo CL, Chiang YT. Exploitation of a rod-shaped, acid-labile curcumin-loaded polymeric nanogel system in the treatment of systemic inflammation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 133:112597. [DOI: 10.1016/j.msec.2021.112597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 10/19/2022]
|
15
|
Rico JE, Sandri EC, Sarmiento AC, Lévesque J, Kenéz Á, Rico DE. Modulation of Plasma and Milk Sphingolipids in Dairy Cows Fed High-Starch Diets. Metabolites 2021; 11:metabo11100711. [PMID: 34677426 PMCID: PMC8540507 DOI: 10.3390/metabo11100711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 11/23/2022] Open
Abstract
Bovine milk is a significant source of sphingolipids, dietary compounds that can exert anti-inflammatory actions, and which can modulate the host’s microbiome. Because sphingolipid synthesis can be modified by diet, we hypothesized that dietary conditions which reduced FFA availability may result in reduced sphingolipid synthesis. Twelve ruminally cannulated cows (120 ± 52 DIM; 35.5 ± 8.9 kg of milk/d; mean ± SD) were randomly assigned to treatment in a crossover design with 21-d periods. Treatments were (1) High starch (HS), (2) Control. The HS diet contained 29% starch, 24% NDF, and 2.8% fatty acids (FA), whereas the Control diet contained 20% starch, 31% NDF, and 2.3% FA. Plasma and milk samples were obtained on d 21 of each period and sphingolipids were quantified using targeted metabolomics. Univariate and multivariate analyses of generalized log-transformed and Pareto-scaled data included ANOVA (fixed effects of treatment) and discriminant analysis. The lipidomics analysis detected 71 sphingolipids across plasma and milk fat, including sphinganines (n = 3), dihydro-ceramides (n = 8), ceramides (Cer; n = 15), sphingomyelins (SM; n = 17), and glycosylated ceramides (n = 28). Followed by Cer, SM were the most abundant sphingolipids detected in milk and plasma, with a preponderance of 16:0-, 23:0-, and 24:0-carbon sidechains. Although no effects of HS diets were observed on plasma sphingolipids, we detected consistent reductions in the concentrations of several milk Cer (e.g., 22:0- and 24:0-Cer) and SM (17:0- and 23:0-SM) in response to HS. Discriminant analysis revealed distinct metabolite separation of HS and Control groups, with several Cer and SM being distinctively predictive of dietary treatment. We conclude that HS diets can reduce the secretion of milk Cer and SM, even in the absence of changes in circulating sphingolipids.
Collapse
Affiliation(s)
- Jorge Eduardo Rico
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
- Correspondence: (J.E.R.); (D.E.R.)
| | | | | | - Janie Lévesque
- CRSAD, Deschambault, QC G0A1S0, Canada; (E.C.S.); (A.C.S.); (J.L.)
| | - Ákos Kenéz
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong, China;
| | - Daniel E. Rico
- CRSAD, Deschambault, QC G0A1S0, Canada; (E.C.S.); (A.C.S.); (J.L.)
- Correspondence: (J.E.R.); (D.E.R.)
| |
Collapse
|
16
|
Vaswani A, Alcazar Magana A, Zimmermann E, Hasan W, Raman J, Maier CS. Comparative liquid chromatography/tandem mass spectrometry lipidomics analysis of macaque heart tissue flash-frozen or embedded in optimal cutting temperature polymer (OCT): Practical considerations. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e9155. [PMID: 34169582 DOI: 10.1002/rcm.9155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 06/13/2023]
Abstract
RATIONALE Biobanks of patient tissues have emerged as essential resources in biomedical research. Optimal cutting temperature compound (OCT) blends have shown to provide stability to the embedded tissue and are compatible with spectroscopic methods, such as infrared (IR) and Raman spectroscopy. Data derived from omics-methods are only useful if tissue damage caused by storage in OCT blends is minimal and well understood. In this context, we investigated the suitability of OCT storage for heart tissue destined for liquid chromatography/tandem mass spectrometry (LC/MS/MS) lipidomic studies. METHODS To determine the compatibility of OCT storage with LC/MS/MS lipidomics studies. The lipid profiles of macaque heart tissue snap-frozen in liquid nitrogen or stored in an OCT blend were evaluated. RESULTS We have evaluated a lipid extraction protocol suitable for OCT-embedded tissue that is compatible with LC/MS/MS. We annotated and evaluated the profiles of 306 lipid species from tissues stored in OCT or liquid nitrogen. For most of the lipid species (95.4%), the profiles were independent of the storage conditions. However, 4.6% of the lipid species; mainly plasmalogens, were affected by the storage method. CONCLUSIONS This study shows that OCT storage is compatible with LC-MS/MS lipidomics of heart tissue, facilitating the use of biobanked tissue samples for future studies.
Collapse
Affiliation(s)
- Ashish Vaswani
- Department of Chemistry at Oregon State University, Corvallis, OR, USA
| | | | | | | | | | - Claudia S Maier
- Department of Chemistry at Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
17
|
Skeletal muscle redox signaling in rheumatoid arthritis. Clin Sci (Lond) 2021; 134:2835-2850. [PMID: 33146370 PMCID: PMC7642299 DOI: 10.1042/cs20190728] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovitis and the presence of serum autoantibodies. In addition, skeletal muscle weakness is a common comorbidity that contributes to inability to work and reduced quality of life. Loss in muscle mass cannot alone account for the muscle weakness induced by RA, but instead intramuscular dysfunction appears as a critical factor underlying the decreased force generating capacity for patients afflicted by arthritis. Oxidative stress and associated oxidative post-translational modifications have been shown to contribute to RA-induced muscle weakness in animal models of arthritis and patients with RA. However, it is still unclear how and which sources of reactive oxygen and nitrogen species (ROS/RNS) that are involved in the oxidative stress that drives the progression toward decreased muscle function in RA. Nevertheless, mitochondria, NADPH oxidases (NOX), nitric oxide synthases (NOS) and phospholipases (PLA) have all been associated with increased ROS/RNS production in RA-induced muscle weakness. In this review, we aim to cover potential ROS sources and underlying mechanisms of oxidative stress and loss of force production in RA. We also addressed the use of antioxidants and exercise as potential tools to counteract oxidative stress and skeletal muscle weakness.
Collapse
|
18
|
Chung HY, Claus RA. Keep Your Friends Close, but Your Enemies Closer: Role of Acid Sphingomyelinase During Infection and Host Response. Front Med (Lausanne) 2021; 7:616500. [PMID: 33553211 PMCID: PMC7859284 DOI: 10.3389/fmed.2020.616500] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022] Open
Abstract
Breakdown of the inert and constitutive membrane building block sphingomyelin to the highly active lipid mediator ceramide by extracellularly active acid sphingomyelinase is tightly regulated during stress response and opens the gate for invading pathogens, triggering the immune response, development of remote organ failure, and tissue repair following severe infection. How do one enzyme and one mediator manage all of these affairs? Under physiological conditions, the enzyme is located in the lysosomes and takes part in the noiseless metabolism of sphingolipids, but following stress the protein is secreted into circulation. When secreted, acid sphingomyelinase (ASM) is able to hydrolyze sphingomyelin present at the outer leaflet of membranes to ceramide. Its generation troubles the biophysical context of cellular membranes resulting in functional assembly and reorganization of proteins and receptors, also embedded in highly conserved response mechanisms. As a consequence of cellular signaling, not only induction of cell death but also proliferation, differentiation, and fibrogenesis are affected. Here, we discuss the current state of the art on both the impact and function of the enzyme during host response and damage control. Also, the potential role of lysosomotropic agents as functional inhibitors of this upstream alarming cascade is highlighted.
Collapse
Affiliation(s)
- Ha-Yeun Chung
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Ralf A Claus
- Department for Anaesthesiology and Intensive Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
19
|
Yakah W, Ramiro-Cortijo D, Singh P, Brown J, Stoll B, Kulkarni M, Oosterloo BC, Burrin D, Maddipati KR, Fichorova RN, Freedman SD, Martin CR. Parenteral Fish-Oil Containing Lipid Emulsions Limit Initial Lipopolysaccharide-Induced Host Immune Responses in Preterm Pigs. Nutrients 2021; 13:205. [PMID: 33445698 PMCID: PMC7828127 DOI: 10.3390/nu13010205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 11/30/2022] Open
Abstract
Multicomponent lipid emulsions are available for critical care of preterm infants. We sought to determine the impact of different lipid emulsions on early priming of the host and its response to an acute stimulus. Pigs delivered 7d preterm (n = 59) were randomized to receive different lipid emulsions for 11 days: 100% soybean oil (SO), mixed oil emulsion (SO, medium chain olive oil and fish oil) including 15% fish oil (MO15), or 100% fish oil (FO100). On day 11, pigs received an 8-h continuous intravenous infusion of either lipopolysaccharide (LPS-lyophilized Escherichia coli) or saline. Plasma was collected for fatty acid, oxylipin, metabolomic, and cytokine analyses. At day 11, plasma omega-3 fatty acid levels in the FO100 groups showed the highest increase in eicosapentaenoic acid, EPA (0.1 ± 0.0 to 9.7 ± 1.9, p < 0.001), docosahexaenoic acid, DHA (day 0 = 2.5 ± 0.7 to 13.6 ± 2.9, p < 0.001), EPA and DHA-derived oxylipins, and sphingomyelin metabolites. In the SO group, levels of cytokine IL1β increased at the first hour of LPS infusion (296.6 ± 308 pg/mL) but was undetectable in MO15, FO100, or in the animals receiving saline instead of LPS. Pigs in the SO group showed a significant increase in arachidonic acid (AA)-derived prostaglandins and thromboxanes in the first hour (p < 0.05). No significant changes in oxylipins were observed with either fish-oil containing group during LPS infusion. Host priming with soybean oil in the early postnatal period preserves a higher AA:DHA ratio and the ability to acutely respond to an external stimulus. In contrast, fish-oil containing lipid emulsions increase DHA, exacerbate a deficit in AA, and limit the initial LPS-induced inflammatory responses in preterm pigs.
Collapse
Affiliation(s)
- William Yakah
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| | - David Ramiro-Cortijo
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Pratibha Singh
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Joanne Brown
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Barbara Stoll
- United States Department of Agriculture-Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Houston, TX 77030, USA
| | - Madhulika Kulkarni
- Section Neonatology, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Houston, TX 77030, USA
| | - Berthe C Oosterloo
- United States Department of Agriculture-Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Houston, TX 77030, USA
| | - Doug Burrin
- United States Department of Agriculture-Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Houston, TX 77030, USA
| | - Krishna Rao Maddipati
- Department of Pathology, Lipidomics Core Facility, Wayne State University, 42 W Warren Avenue, Detroit, MI 48202, USA
| | - Raina N Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Steven D Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Camilia R Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| |
Collapse
|
20
|
Ferchaud-Roucher V, Zair Y, Aguesse A, Krempf M, Ouguerram K. Omega 3 Improves Both apoB100-containing Lipoprotein Turnover and their Sphingolipid Profile in Hypertriglyceridemia. J Clin Endocrinol Metab 2020; 105:5893579. [PMID: 32805740 DOI: 10.1210/clinem/dgaa459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/08/2020] [Indexed: 01/17/2023]
Abstract
CONTEXT Evidence for an association between sphingolipids and metabolic disorders is increasingly reported. Omega-3 long-chain polyunsaturated fatty acids (n-3 LC-PUFAs) improve apolipoprotein B100 (apoB100)-containing lipoprotein metabolism, but their effects on the sphingolipid content in lipoproteins remain unknown. OBJECTIVES In subjects with hypertriglyceridemia, we analyzed the effect of n-3 LC-PUFAs on the turnover apoB100-containing lipoproteins and on their sphingolipid content and looked for the possible association between these lipid levels and apoB100-containing lipoprotein turnover parameters. METHODS Six subjects underwent a kinetic study before and after n-3 supplementation for 2 months with 1 g of fish oil 3 times day containing 360 mg of eicosapentaenoic acid (EPA) and 240 mg of docosahexaenoic acid (DHA) in the form of triglycerides. We examined apoB100-containing lipoprotein turnover by primed perfusion labeled [5,5,5-2H3]-leucine and determined kinetic parameters using a multicompartmental model. We quantified sphingolipid species content in lipoproteins using mass spectrometry. RESULTS Supplementation decreased very low-density lipoprotein (VLDL), triglyceride, and apoB100 concentrations. The VLDL neutral and polar lipids showed increased n-3 LC-PUFA and decreased n-6 LC-PUFA content. The conversion rate of VLDL1 to VLDL2 and of VLDL2 to LDL was increased. We measured a decrease in total apoB100 production and VLDL1 production. Supplementation reduced the total ceramide concentration in VLDL while the sphingomyelin content in LDL was increased. We found positive correlations between plasma palmitic acid and VLDL ceramide and between VLDL triglyceride and VLDL ceramide, and inverse correlations between VLDL n-3 LC-PUFA and VLDL production. CONCLUSION Based on these results, we hypothesize that the improvement in apoB100 metabolism during n-3 LC-PUFA supplementation is contributed to by changes in sphingolipids.
Collapse
Affiliation(s)
- Véronique Ferchaud-Roucher
- University of Nantes, CHU Nantes, INRAe, UMR 1280 Physiopathology of Nutritional Adaptations, Nantes, France
- CRNH, West Human Nutrition Research Center, Nantes, France
| | - Yassine Zair
- CRNH, West Human Nutrition Research Center, Nantes, France
| | - Audrey Aguesse
- University of Nantes, CHU Nantes, INRAe, UMR 1280 Physiopathology of Nutritional Adaptations, Nantes, France
- CRNH, West Human Nutrition Research Center, Nantes, France
| | - Michel Krempf
- University of Nantes, CHU Nantes, INRAe, UMR 1280 Physiopathology of Nutritional Adaptations, Nantes, France
- CRNH, West Human Nutrition Research Center, Nantes, France
| | - Khadija Ouguerram
- University of Nantes, CHU Nantes, INRAe, UMR 1280 Physiopathology of Nutritional Adaptations, Nantes, France
- CRNH, West Human Nutrition Research Center, Nantes, France
| |
Collapse
|
21
|
Leonetti D, Estéphan H, Ripoche N, Dubois N, Aguesse A, Gouard S, Brossard L, Chiavassa S, Corre I, Pecqueur C, Neunlist M, Hadchity E, Gaugler MH, Mahé MM, Paris F. Secretion of Acid Sphingomyelinase and Ceramide by Endothelial Cells Contributes to Radiation-Induced Intestinal Toxicity. Cancer Res 2020; 80:2651-2662. [PMID: 32291318 DOI: 10.1158/0008-5472.can-19-1527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 12/16/2019] [Accepted: 04/08/2020] [Indexed: 11/16/2022]
Abstract
Ceramide-induced endothelial cell apoptosis boosts intestinal stem cell radiosensitivity. However, the molecular connection between these two cellular compartments has not been clearly elucidated. Here we report that ceramide and its related enzyme acid sphingomyelinase (ASM) are secreted by irradiated endothelial cells and act as bystander factors to enhance the radiotoxicity of intestinal epithelium. Ceramide and the two isoforms of ASM were acutely secreted in the blood serum of wild-type mice after 15 Gy radiation dose, inducing a gastrointestinal syndrome. Interestingly, serum ceramide was not enhanced in irradiated ASMKO mice, which are unable to develop intestinal failure injury. Because ASM/ceramide were secreted by primary endothelial cells, their contribution was studied in intestinal epithelium dysfunction using coculture of primary endothelial cells and intestinal T84 cells. Adding exogenous ASM or ceramide enhanced epithelial cell growth arrest and death. Conversely, blocking their secretion by endothelial cells using genetic, pharmacologic, or immunologic approaches abolished intestinal T84 cell radiosensitivity. Use of enteroid models revealed ASM and ceramide-mediated deleterious mode-of-action: when ceramide reduced the number of intestinal crypt-forming enteroids without affecting their structure, ASM induced a significant decrease of enteroid growth without affecting their number. Identification of specific and different roles for ceramide and ASM secreted by irradiated endothelial cells opens new perspectives in the understanding of intestinal epithelial dysfunction after radiation and defines a new class of potential therapeutic radiomitigators. SIGNIFICANCE: This study identifies secreted ASM and ceramide as paracrine factors enhancing intestinal epithelial dysfunction, revealing a previously unknown class of mediators of radiosensitivity.
Collapse
Affiliation(s)
| | - Hala Estéphan
- Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France.,Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadath, Lebanon
| | | | - Nolwenn Dubois
- Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France.,ICO, Saint-Herblain, France
| | - Audrey Aguesse
- Université de Nantes, INRA UMR 1280 Physiologie des Adaptations Nutritionnelles, Nantes, France.,CRNHO, West Human Nutrition Research Center, Nantes, France
| | | | - Lisa Brossard
- The Enteric Nervous System in Gut and Brain Disorders, INSERM, Université de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | | | - Isabelle Corre
- Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France
| | | | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain Disorders, INSERM, Université de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Elie Hadchity
- Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadath, Lebanon
| | | | - Maxime M Mahé
- The Enteric Nervous System in Gut and Brain Disorders, INSERM, Université de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - François Paris
- Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France. .,ICO, Saint-Herblain, France
| |
Collapse
|
22
|
Association of proteome and metabolome signatures with severity in patients with community-acquired pneumonia. J Proteomics 2020; 214:103627. [DOI: 10.1016/j.jprot.2019.103627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/29/2019] [Accepted: 12/22/2019] [Indexed: 01/09/2023]
|
23
|
McFadden JW, Rico JE. Invited review: Sphingolipid biology in the dairy cow: The emerging role of ceramide. J Dairy Sci 2019; 102:7619-7639. [PMID: 31301829 DOI: 10.3168/jds.2018-16095] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/30/2019] [Indexed: 01/12/2023]
Abstract
The physiological control of lactation through coordinated adaptations is of fundamental importance for mammalian neonatal life. The putative actions of reduced insulin sensitivity and responsiveness and enhanced adipose tissue lipolysis spare glucose for the mammary synthesis of milk. However, severe insulin antagonism and body fat mobilization may jeopardize hepatic health and lactation in dairy cattle. Interestingly, lipolysis- and dietary-derived fatty acids may impair insulin sensitivity in cows. The mechanisms are undefined yet have major implications for the development of postpartum fatty liver disease. In nonruminants, the sphingolipid ceramide is a potent mediator of saturated fat-induced insulin resistance that defines in part the mechanisms of type 2 diabetes mellitus and nonalcoholic fatty liver disease. In ruminants including the lactating dairy cow, the functions of ceramide had remained virtually undescribed. Through a series of hypothesis-centered studies, ceramide has emerged as a potential antagonist of insulin-stimulated glucose utilization by adipose and skeletal muscle tissues in dairy cattle. Importantly, bovine data suggest that the ability of ceramide to inhibit insulin action likely depends on the lipolysis-dependent hepatic synthesis and secretion of ceramide during early lactation. Although these mechanisms appear to fade as lactation advances beyond peak milk production, early evidence suggests that palmitic acid feeding is a means to augment ceramide supply. Herein, we review a body of work that focuses on sphingolipid biology and the role of ceramide in the dairy cow within the framework of hepatic and fatty acid metabolism, insulin function, and lactation. The potential involvement of ceramide within the endocrine control of lactation is also considered.
Collapse
Affiliation(s)
- J W McFadden
- Department of Animal Science, Cornell University, Ithaca, NY 14853.
| | - J E Rico
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| |
Collapse
|
24
|
Acid sphingomyelinase plays a critical role in LPS- and cytokine-induced tissue factor procoagulant activity. Blood 2019; 134:645-655. [PMID: 31262782 DOI: 10.1182/blood.2019001400] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022] Open
Abstract
Tissue factor (TF) is a cofactor for factor VIIa and the primary cellular initiator of coagulation. Typically, most TF on cell surfaces exists in a cryptic coagulant-inactive state but are transformed to a procoagulant form (decryption) following cell activation. Our recent studies in cell model systems showed that sphingomyelin (SM) in the outer leaflet of the plasma membrane is responsible for maintaining TF in an encrypted state in resting cells, and the hydrolysis of SM leads to decryption of TF. The present study was carried out to investigate the relevance of this novel mechanism in the regulation of TF procoagulant activity in pathophysiology. As observed in cell systems, administration of adenosine triphosphate (ATP) to mice enhanced lipopolysaccharide (LPS)-induced TF procoagulant activity in monocytes. Treatment of mice with pharmacological inhibitors of acid sphingomyelinase (ASMase), desipramine and imipramine, attenuated ATP-induced TF decryption. Interestingly, ASMase inhibitors also blocked LPS-induced TF procoagulant activity without affecting the LPS-induced de novo synthesis of TF protein. Additional studies showed that LPS induced translocation of ASMase to the outer leaflet of the plasma membrane and reduced SM levels in monocytes. Studies using human monocyte-derived macrophages and endothelial cells further confirmed the role of ASMase in LPS- and cytokine-induced TF procoagulant activity. Overall, our data indicate that LPS- or cytokine-induced TF procoagulant activity requires the decryption of newly synthesized TF protein by ASMase-mediated hydrolysis of SM. The observation that ASMase inhibitors attenuate TF-induced coagulation raises the possibility of their therapeutic use in treating thrombotic disorders associated with aberrant expression of TF.
Collapse
|
25
|
Ansari SA, Pendurthi UR, Rao LVM. Role of Cell Surface Lipids and Thiol-Disulphide Exchange Pathways in Regulating the Encryption and Decryption of Tissue Factor. Thromb Haemost 2019; 119:860-870. [PMID: 30861549 DOI: 10.1055/s-0039-1681102] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tissue factor (TF), a transmembrane glycoprotein, is the cellular receptor of the coagulation factors VII (FVII) and VIIa (FVIIa). The formation of TF-FVIIa complex triggers the initiation of the blood coagulation pathway. TF plays an essential role in haemostasis, but an aberrant expression of TF activity contributes to thrombotic disorders. In health, TF pro-coagulant activity on cells is controlled tightly to allow sufficient coagulant activity to achieve haemostasis but not to cause thrombosis. It is achieved largely by selective localization of TF in the body and encryption of TF at the cell surface. A vast majority of TF on resting cells exists in an encrypted state with minimal pro-coagulant activity but becomes pro-thrombotic following cell injury or activation. At present, the mechanisms that are responsible for TF encryption and activation (decryption) are not entirely clear, but recent studies provide important mechanistic insights into these processes. To date, externalization of phosphatidylserine to the outer leaflet and thiol-disulphide exchange pathways that either turn on and off the allosteric disulphide bond in TF are shown to play a major role in regulating TF pro-coagulant activity on cell surfaces. Recent studies showed that sphingomyelin, a major phospholipid in the outer leaflet of plasma membrane, plays a critical role in the encryption of TF in resting cells. The present review provides an overview of recent literature on the above-described mechanisms of TF encryption and decryption with a particular emphasis on our recent findings.
Collapse
Affiliation(s)
- Shabbir A Ansari
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| |
Collapse
|
26
|
Qin N, Kokkonen T, Salin S, Seppänen-Laakso T, Taponen J, Vanhatalo A, Elo K. Prepartal high-energy feeding with grass silage-based diets does not disturb the hepatic adaptation of dairy cows during the periparturient period. J Dairy Sci 2018; 101:8929-8943. [DOI: 10.3168/jds.2017-13153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/08/2018] [Indexed: 12/15/2022]
|
27
|
Kumar V. Dendritic cells in sepsis: Potential immunoregulatory cells with therapeutic potential. Mol Immunol 2018; 101:615-626. [DOI: 10.1016/j.molimm.2018.07.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/01/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022]
|
28
|
Ahn WG, Jung JS, Song DK. Lipidomic analysis of plasma lipids composition changes in septic mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:399-408. [PMID: 29962854 PMCID: PMC6019871 DOI: 10.4196/kjpp.2018.22.4.399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 01/16/2023]
Abstract
A lipidomic study on extensive plasma lipids in bacterial peritonitis (cecal ligation and puncture, CLP)-induced sepsis in mice was done at 24 h post-CLP. The effects of administration of lysophosphatidylcholine (LPC) and lysophosphatidic acid (LPA), compounds known to have beneficial effects in CLP, on the sepsis-induced plasma lipid changes were also examined. Among the 147 plasma lipid species from 13 lipid subgroups (fatty acid [FA], LPA, LPC, lysophosphatidylethanolamine [LPE], phosphatidic acid [PA], phosphatidylcholine [PC], phosphatidylethanolamine [PE], phosphatidylinositol [PI], monoacylglyceride [MG], diacylglyceride [DG], triacylglyceride [TG], sphingomyelin [SM], and ceramide [Cer]) analyzed in this study, 40 and 70 species were increased, and decreased, respectively, in the CLP mice. Treatments with LPC and LPA affected 14 species from 7 subgroups, and 25 species from 9 subgroups, respectively. These results could contribute to finding the much needed reliable biomarkers of sepsis.
Collapse
Affiliation(s)
- Won-Gyun Ahn
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon 24252, Korea
| | - Jun-Sub Jung
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon 24252, Korea
| | - Dong-Keun Song
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
29
|
Lallemand T, Rouahi M, Swiader A, Grazide MH, Geoffre N, Alayrac P, Recazens E, Coste A, Salvayre R, Nègre-Salvayre A, Augé N. nSMase2 (Type 2-Neutral Sphingomyelinase) Deficiency or Inhibition by GW4869 Reduces Inflammation and Atherosclerosis in Apoe -/- Mice. Arterioscler Thromb Vasc Biol 2018; 38:1479-1492. [PMID: 29794115 PMCID: PMC6039418 DOI: 10.1161/atvbaha.118.311208] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/07/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Atherosclerosis is a chronic multifactorial and inflammatory disease of large and medium arteries and the leading cause of cardiovascular diseases worldwide. The aim of this study was to investigate whether and how the nSMase2 (type 2-neutral sphingomyelinase), a key enzyme of sphingolipid metabolism, may contribute to the development of atherosclerotic lesions. APPROACH AND RESULTS The role of nSMase2 in atherosclerosis was investigated in Apoe-/-;Smpd3fro/fro mice, mutant for nSMase2, and in Apoe-/-;Smpd3+/+ mice intraperitoneally injected with GW4869, a pharmacological nSMase2 inhibitor. The defect or inhibition of nSMase2 resulted in a reduction of atherosclerotic lesions and a decrease in macrophage infiltration and lipid deposition, although cholesterolemia remained unchanged. nSMase2 inhibition decreased the inflammatory response of murine endothelial cells to oxLDL (oxidized low-density lipoprotein), as assessed by the significant reduction of MCP-1 (monocyte chemoattractant protein 1), ICAM-1 (intercellular adhesion molecule-1), and VCAM-1 (vascular cell adhesion molecule-1) mRNA expressions and macrophage recruitment. Likewise, in RAW264.7 or in macrophages isolated from Apoe-/-/Smpd3fro/fro or Apoe-/-/Smpd3+/+ mice stimulated by lipopolysaccharides, nSMase2 inhibition resulted in a decrease in the expression of inflammatory molecules. Mechanistically, the anti-inflammatory response resulting from nSMase2 inhibition involves Nrf2 (nuclear factor [erythroid-derived 2]-like 2 or NF-E2-related factor-2) activation in both endothelial cells and macrophages, as assessed by the lack of protective effect of GW4869 in endothelial cells silenced for Nrf2 by small interfering RNAs, and in lipopolysaccharide-stimulated macrophages issued from Nrf2-KO mice. CONCLUSIONS The genetic deficiency or inhibition of nSMase2 strongly decreases the development of atherosclerotic lesions in Apoe-/- mice, by reducing inflammatory responses through a mechanism involving the Nrf2 pathway. Inhibitors of nSMase2 may, therefore, constitute a novel approach to slow down atherosclerosis progression.
Collapse
Affiliation(s)
- Tom Lallemand
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Myriam Rouahi
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Audrey Swiader
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Marie-Hélène Grazide
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Nancy Geoffre
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Paul Alayrac
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Emeline Recazens
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Agnès Coste
- PHARMA-DEV, IRD UMR 152, Toulouse, France (A.C.)
| | - Robert Salvayre
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Anne Nègre-Salvayre
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.).,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| | - Nathalie Augé
- From the INSERM U-1048 Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.) .,Université Paul Sabatier, Toulouse, France (T.L., M.R., A.S., M.-H.G., N.G., P.A., E.R., R.S., A.N.-S., N.A.)
| |
Collapse
|
30
|
Abstract
Studies of bioactive lipids in general and sphingolipids in particular have intensified over the past several years, revealing an unprecedented and unanticipated complexity of the lipidome and its many functions, which rivals, if not exceeds, that of the genome or proteome. These results highlight critical roles for bioactive sphingolipids in most, if not all, major cell biological responses, including all major cell signalling pathways, and they link sphingolipid metabolism to key human diseases. Nevertheless, the fairly nascent field of bioactive sphingolipids still faces challenges in its biochemical and molecular underpinnings, including defining the molecular mechanisms of pathway and enzyme regulation, the study of lipid-protein interactions and the development of cellular probes, suitable biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Yusuf A Hannun
- Stony Brook Cancer Center and Department of Medicine, Stony Brook University, New York 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and Department of Medicine, Stony Brook University, New York 11794, USA
- Northport Veterans Affairs Medical Center, Northport, New York 11768, USA
| |
Collapse
|
31
|
Dietary and Endogenous Sphingolipid Metabolism in Chronic Inflammation. Nutrients 2017; 9:nu9111180. [PMID: 29143791 PMCID: PMC5707652 DOI: 10.3390/nu9111180] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/21/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Abstract
Chronic inflammation is a common underlying factor in many major metabolic diseases afflicting Western societies. Sphingolipid metabolism is pivotal in the regulation of inflammatory signaling pathways. The regulation of sphingolipid metabolism is in turn influenced by inflammatory pathways. In this review, we provide an overview of sphingolipid metabolism in mammalian cells, including a description of sphingolipid structure, biosynthesis, turnover, and role in inflammatory signaling. Sphingolipid metabolites play distinct and complex roles in inflammatory signaling and will be discussed. We also review studies examining dietary sphingolipids and inflammation, derived from in vitro and rodent models, as well as human clinical trials. Dietary sphingolipids appear to influence inflammation-related chronic diseases through inhibiting intestinal lipid absorption, altering gut microbiota, activation of anti-inflammatory nuclear receptors, and neutralizing responses to inflammatory stimuli. The anti-inflammatory effects observed with consuming dietary sphingolipids are in contrast to the observation that most cellular sphingolipids play roles in augmenting inflammatory signaling. The relationship between dietary sphingolipids and low-grade chronic inflammation in metabolic disorders is complex and appears to depend on sphingolipid structure, digestion, and metabolic state of the organism. Further research is necessary to confirm the reported anti-inflammatory effects of dietary sphingolipids and delineate their impacts on endogenous sphingolipid metabolism.
Collapse
|
32
|
Apigenin-7-O-β-D-glucuronide inhibits modified low-density lipoprotein uptake and foam cell formation in macrophages. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
33
|
Yamada T, Steinz MM, Kenne E, Lanner JT. Muscle Weakness in Rheumatoid Arthritis: The Role of Ca 2+ and Free Radical Signaling. EBioMedicine 2017; 23:12-19. [PMID: 28781131 PMCID: PMC5605300 DOI: 10.1016/j.ebiom.2017.07.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
In addition to the primary symptoms arising from inflammatory processes in the joints, muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Muscle weakness not only reduces the quality of life for the affected patients, but also dramatically increases the burden on society since patients' work ability decreases. A 25–70% reduction in muscular strength has been observed in pateints with RA when compared with age-matched healthy controls. The reduction in muscle strength is often larger than what could be explained by the reduction in muscle size in patients with RA, which indicates that intracellular (intrinsic) muscle dysfunction plays an important role in the underlying mechanism of muscle weakness associated with RA. In this review, we highlight the present understanding of RA-associated muscle weakness with special focus on how enhanced Ca2 + release from the ryanodine receptor and free radicals (reactive oxygen/nitrogen species) contributes to muscle weakness, and recent developments of novel therapeutic interventions. Muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Intrinsic muscle weakness is important in the underlying mechanisms of muscle weakness associated with rheumatoid arthritis. Enhanced Ca2 + release and peroxynitrite-induced stress contributes to RA-induced muscle weakness.
Collapse
Affiliation(s)
- Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Maarten M Steinz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
34
|
Metabolite Profiles in Sepsis: Developing Prognostic Tools Based on the Type of Infection. Crit Care Med 2017; 44:1649-62. [PMID: 27097292 DOI: 10.1097/ccm.0000000000001740] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Currently used biomarkers insufficiently discriminate between patients with systemic inflammatory response syndrome of non-infectious origin and sepsis. The aim of this study was to identify surrogate markers that distinguish between systemic inflammatory response syndrome and sepsis as well as the underlying type of infection by targeted metabolomics. DESIGN Retrospective analysis. SETTINGS Six sites of the Hellenic Sepsis Study Group and at Jena University Hospital. PATIENTS A total of 406 patients were analyzed: 66 fulfilling criteria for diagnosis of systemic inflammatory response syndrome, 100 for community-acquired pneumonia, 112 for urinary tract infection, 83 for intra-abdominal infection and 45 for bloodstream infection. Patients were divided into test cohort (n = 268) and confirmation cohort (n = 138). INTERVENTIONS A total of 186 metabolites were determined by liquid chromatography tandem mass spectrometry. MEASUREMENTS AND MAIN RESULTS Serum concentrations of most acylcarnitines, glycerophospholipids and sphingolipids were altered in sepsis compared to systemic inflammatory response syndrome. A regression model combining the sphingolipid SM C22:3 and the glycerophospholipid lysoPCaC24:0 was discovered for sepsis diagnosis with a sensitivity of 84.1% and specificity of 85.7%. Furthermore, specific metabolites could be used for the discrimination of different types of infection. The glycerophospholipid lysoPCaC26:1 identified patients with community-acquired pneumonia in sepsis or severe sepsis/septic shock. Within severe sepsis/septic shock, patients with bloodstream infection could be discriminated by a decrease of acetylornithine. Changes of metabolites between sepsis and severe sepsis/septic shock also varied according to the underlying type of infection, showing that putrescine, lysoPCaC18:0 and SM C16:1 are associated with unfavorable outcome in community-acquired pneumonia, intra-abdominal infections and bloodstream infections, respectively. CONCLUSIONS Using a metabolomics approach, single metabolites are identified that allow a good, albeit at about 14% false positive rate of sepsis diagnosis. Additionally, metabolites might be also useful for differentiation and prognosis according to the type of underlying infection. However, confirmation of the findings in ongoing studies is mandatory before they can be applied in the development of novel diagnostic tools for the management of sepsis.
Collapse
|
35
|
Wang J, Pendurthi UR, Rao LVM. Sphingomyelin encrypts tissue factor: ATP-induced activation of A-SMase leads to tissue factor decryption and microvesicle shedding. Blood Adv 2017; 1:849-862. [PMID: 28758160 PMCID: PMC5531194 DOI: 10.1182/bloodadvances.2016003947] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 04/09/2017] [Indexed: 11/20/2022] Open
Abstract
A majority of tissue factor (TF) on cell surfaces exists in an encrypted state with minimal to no procoagulant activity. At present, it is unclear whether limited availability of phosphatidylserine (PS) and/or a specific membrane lipid in the outer leaflet of the plasma membrane contributes to TF encryption. Sphingomyelin (SM) is a major phospholipid in the outer leaflet, and SM metabolism is shown to be altered in many disease settings that cause thrombotic disorders. The present study is carried out to investigate the effect of SM metabolism on TF activity and TF+ microvesicles (MVs) release. In vitro studies using TF reconstituted into liposomes containing varying molar ratios of SM showed that a high molar ratio of SM in the proteoliposomes inhibits TF coagulant activity. Treatment of macrophages with sphingomyelinase (SMase) that hydrolyzes SM in the outer leaflet results in increased TF activity at the cell surface and TF+ MVs release without increasing PS externalization. Adenosine triphosphate (ATP) stimulation of macrophages that activates TF and induces MV shedding also leads to translocation of acid-sphingomyelinase (A-SMase) to the plasma membrane. ATP stimulation increases the hydrolysis of SM in the outer leaflet. Inhibition of A-SMase expression or activity not only attenuates ATP-induced SM hydrolysis, but also inhibits ATP-induced TF decryption and TF+ MVs release. Overall, our novel findings show that SM plays a role in maintaining TF in an encrypted state in resting cells and hydrolysis of SM following cell injury removes the inhibitory effect of SM on TF activity, thus leading to TF decryption.
Collapse
Affiliation(s)
- Jue Wang
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| |
Collapse
|
36
|
Zhan X, Cox C, Ander BP, Liu D, Stamova B, Jin LW, Jickling GC, Sharp FR. Inflammation Combined with Ischemia Produces Myelin Injury and Plaque-Like Aggregates of Myelin, Amyloid-β and AβPP in Adult Rat Brain. J Alzheimers Dis 2016; 46:507-23. [PMID: 25790832 PMCID: PMC4878315 DOI: 10.3233/jad-143072] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background: Ischemia, white matter injury, and Alzheimer’s disease (AD) pathologies often co-exist in aging brain. How one condition predisposes to, interacts with, or perhaps causes the others remains unclear. Objectives: To better understand the link between ischemia, white matter injury, and AD, adult rats were administered lipopolysaccharide (LPS) to serve as an inflammatory stimulus, and 24 h later subjected to 20-min focal cerebral ischemia (IS) followed by 30-min hypoxia (H). Methods: Myelin and axonal damage, as well as amyloid-β (Aβ) and amyloid-β protein precursor (AβPP) deposition were examined by Western blot and immunocytochemistry following LPS/IS/H. Findings were compared to the 5XFAD mouse AD brain. Results: Myelin/axonal injury was observed bilaterally in cortex following LPS/IS/H, along with an increase in IL-1, granzyme B, and LPS. AβPP deposition was present in ischemic striatum in regions of myelin loss. Aβ1-42 and AβPP were deposited in small foci in ischemic cortex that co-localized with myelin aggregates. In the 5XFAD mouse AD model, cortical amyloid plaques also co-localized with myelin aggregates. Conclusions: LPS/IS/H produce myelin injury and plaque-like aggregates of myelin. AβPP and Aβ co-localize with these myelin aggregates.
Collapse
Affiliation(s)
- Xinhua Zhan
- Department of Neurology, MIND Institute, University of California at Davis, Sacramento, CA, USA
| | - Christopher Cox
- Department of Neurology, MIND Institute, University of California at Davis, Sacramento, CA, USA
| | - Bradley P Ander
- Department of Neurology, MIND Institute, University of California at Davis, Sacramento, CA, USA
| | - Dazhi Liu
- Department of Neurology, MIND Institute, University of California at Davis, Sacramento, CA, USA
| | - Boryana Stamova
- Department of Neurology, MIND Institute, University of California at Davis, Sacramento, CA, USA
| | - Lee-Way Jin
- Alzheimer's Disease Center, University of California at Davis, Sacramento, CA, USA.,Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, CA, USA
| | - Glen C Jickling
- Department of Neurology, MIND Institute, University of California at Davis, Sacramento, CA, USA
| | - Frank R Sharp
- Department of Neurology, MIND Institute, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
37
|
Adada M, Luberto C, Canals D. Inhibitors of the sphingomyelin cycle: Sphingomyelin synthases and sphingomyelinases. Chem Phys Lipids 2016. [DOI: 10.1016/j.chemphyslip.2015.07.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
38
|
Vito CD, Hadi LA, Navone SE, Marfia G, Campanella R, Mancuso ME, Riboni L. Platelet-derived sphingosine-1-phosphate and inflammation: from basic mechanisms to clinical implications. Platelets 2016; 27:393-401. [PMID: 26950429 DOI: 10.3109/09537104.2016.1144179] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Beyond key functions in hemostasis and thrombosis, platelets are recognized as key players of inflammation, an underlying feature of a variety of diseases. In this regard, platelets act as a circulating source of several pro- and anti-inflammatory molecules, which are secreted from their intracellular stores upon activation. Among them, mounting evidence highlights a crucial role of sphingosine-1-phosphate (S1P), a multifunctional sphingoid mediator. S1P-induced pleiotropic effects include those crucial in inflammatory processes, such as the maintenance of the endothelial barrier integrity, and leukocyte activation and recruitment at the injured site. This review outlines the peculiar features and molecular mechanisms that allow platelets for acting as a unique factory that produces and stores S1P in large quantities. A particular emphasis is placed on the autocrine and paracrine roles of S1P derived from the "inflamed" platelets, highlighting the role of its cross-talk with endothelial and blood cells involved in inflammation, and the mechanisms of its contribution to the development and progression of inflammatory diseases. Finally, potential clinical implications of platelet-derived S1P as diagnostic tool of inflammatory severity, and as therapeutic target in inflammation are discussed.
Collapse
Affiliation(s)
- Clara Di Vito
- a Department of Medical Biotechnology and Translational Medicine, LITA-Segrate , University of Milan , Milan , Italy
| | - Loubna Abdel Hadi
- a Department of Medical Biotechnology and Translational Medicine, LITA-Segrate , University of Milan , Milan , Italy
| | - Stefania Elena Navone
- b Neurosurgery Unit, Laboratory of Experimental Neurosurgery and Cell Therapy, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico , University of Milan , Milan , Italy
| | - Giovanni Marfia
- b Neurosurgery Unit, Laboratory of Experimental Neurosurgery and Cell Therapy, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico , University of Milan , Milan , Italy
| | - Rolando Campanella
- c Division of Neurosurgery, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico , University of Milan , Milan , Italy
| | - Maria Elisa Mancuso
- d Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico , Milan , Italy
| | - Laura Riboni
- a Department of Medical Biotechnology and Translational Medicine, LITA-Segrate , University of Milan , Milan , Italy
| |
Collapse
|
39
|
Rodríguez-Sureda V, Crovetto F, Triunfo S, Sánchez O, Crispi F, Llurba E, Gratacós E, Figueras F, Domínguez C. Increased secretory sphingomyelinase activity in the first trimester of pregnancy in women later developing preeclampsia: a nested case-control study. Biol Chem 2016; 397:269-79. [DOI: 10.1515/hsz-2015-0266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/28/2015] [Indexed: 11/15/2022]
Abstract
Abstract
The pathogenic basis of abnormal placentation and dysfunction in preeclampsia (PE) is highly complex and incompletely understood. Secretory sphyngomyelinase activity (S-ASM) was analyzed in plasma samples from 158 pregnant women developing PE and 112 healthy pregnant controls. Serum PlGF, sFlt-1, s-Endoglin and sVCAM were measured. Results showed S-ASM activity to be higher in women who later developed PE than in those with uncomplicated pregnancies (40.6% and 28.8% higher in the late- and early-onset groups, respectively). Plasma S-ASM activity correlated significantly with circulating markers of endothelial damage in the late-PE group (endoglin and sVCAM-1), with plasma cholesterol and total lipid levels. However, these significant associations were not observed in the early-PE or control groups. This work provides the first evidence of significantly elevated circulating S-ASM activity in the first trimester of pregnancy in women who go on to develop PE; thus, it may be deduced that the circulating form of ASM is biologically active in PE and could contribute to promoting endothelial dysfunction and cardiovascular programming. Plasma S-ASM measurement may have clinical relevance as a further potential biomarker contributing to the earliest identification of women at risk of developing preeclampsia.
Collapse
|
40
|
Mizuno S, Ogishima S, Kitatani K, Kikuchi M, Tanaka H, Yaegashi N, Nakaya J. Network Analysis of a Comprehensive Knowledge Repository Reveals a Dual Role for Ceramide in Alzheimer's Disease. PLoS One 2016; 11:e0148431. [PMID: 26849355 PMCID: PMC4752297 DOI: 10.1371/journal.pone.0148431] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/18/2016] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of senile dementia. Many inflammatory factors such as amyloid-β and pro-inflammatory cytokines are known to contribute to the inflammatory response in the AD brain. Sphingolipids are widely known to have roles in the pathogenesis of inflammatory diseases, where the precise roles for sphingolipids in inflammation-associated pathogenesis of AD are not well understood. Here we performed a network analysis to clarify the importance of sphingolipids and to model relationships among inflammatory factors and sphingolipids in AD. In this study, we have updated sphingolipid signaling and metabolic cascades in a map of AD signaling networks that we named “AlzPathway,” a comprehensive knowledge repository of signaling pathways in AD. Our network analysis of the updated AlzPathway indicates that the pathways related to ceramide are one of the primary pathways and that ceramide is one of the important players in the pathogenesis of AD. The results of our analysis suggest the following two prospects about inflammation in AD: (1) ceramide could play important roles in both inflammatory and anti-inflammatory pathways of AD, and (2) several factors such as Sphingomyelinase and Siglec-11 may be associated with ceramide related inflammation and anti-inflammation pathways in AD. In this study, network analysis of comprehensive knowledge repository reveals a dual role for ceramide in AD. This result provides a clue to clarify sphingolipids related inflammatory and anti-inflammatory pathways in AD.
Collapse
Affiliation(s)
- Satoshi Mizuno
- Department of Clinical Informatics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Clinical Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- * E-mail: (SM); (SO)
| | - Soichi Ogishima
- Department of Clinical Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- * E-mail: (SM); (SO)
| | - Kazuyuki Kitatani
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masataka Kikuchi
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Tanaka
- Department of Clinical Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Nobuo Yaegashi
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jun Nakaya
- Department of Clinical Informatics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
41
|
Kornhuber J, Rhein C, Müller CP, Mühle C. Secretory sphingomyelinase in health and disease. Biol Chem 2016; 396:707-36. [PMID: 25803076 DOI: 10.1515/hsz-2015-0109] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/16/2015] [Indexed: 01/12/2023]
Abstract
Acid sphingomyelinase (ASM), a key enzyme in sphingolipid metabolism, hydrolyzes sphingomyelin to ceramide and phosphorylcholine. In mammals, the expression of a single gene, SMPD1, results in two forms of the enzyme that differ in several characteristics. Lysosomal ASM (L-ASM) is located within the lysosome, requires no additional Zn2+ ions for activation and is glycosylated mainly with high-mannose oligosaccharides. By contrast, the secretory ASM (S-ASM) is located extracellularly, requires Zn2+ ions for activation, has a complex glycosylation pattern and has a longer in vivo half-life. In this review, we summarize current knowledge regarding the physiology and pathophysiology of S-ASM, including its sources and distribution, molecular and cellular mechanisms of generation and regulation and relevant in vitro and in vivo studies. Polymorphisms or mutations of SMPD1 lead to decreased S-ASM activity, as detected in patients with Niemann-Pick disease B. Thus, lower serum/plasma activities of S-ASM are trait markers. No genetic causes of increased S-ASM activity have been identified. Instead, elevated activity is the result of enhanced release (e.g., induced by lipopolysaccharide and cytokine stimulation) or increased enzyme activation (e.g., induced by oxidative stress). Increased S-ASM activity in serum or plasma is a state marker of a wide range of diseases. In particular, high S-ASM activity occurs in inflammation of the endothelium and liver. Several studies have demonstrated a correlation between S-ASM activity and mortality induced by severe inflammatory diseases. Serial measurements of S-ASM reveal prolonged activation and, therefore, the measurement of this enzyme may also provide information on past inflammatory processes. Thus, S-ASM may be both a promising clinical chemistry marker and a therapeutic target.
Collapse
|
42
|
Konno Y, Takahashi I, Narita A, Takeda O, Koizumi H, Tamura M, Kikuchi W, Komatsu A, Tamura H, Tsuchida S, Noguchi A, Takahashi T. Elevation of Serum Acid Sphingomyelinase Activity in Acute Kawasaki Disease. TOHOKU J EXP MED 2015; 237:133-40. [PMID: 26447086 DOI: 10.1620/tjem.237.133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Kawasaki disease (KD) is an acute systemic vasculitis that affects both small and medium-sized vessels including the coronary arteries in infants and children. Acid sphingomyelinase (ASM) is a lysosomal glycoprotein that hydrolyzes sphingomyelin to ceramide, a lipid, that functions as a second messenger in the regulation of cell functions. ASM activation has been implicated in numerous cellular stress responses and is associated with cellular ASM secretion, either through alternative trafficking of the ASM precursor protein or by means of an unidentified mechanism. Elevation of serum ASM activity has been described in several human diseases, suggesting that patients with diseases involving vascular endothelial cells may exhibit a preferential elevation of serum ASM activity. As acute KD is characterized by systemic vasculitis that could affect vascular endothelial cells, the elevation of serum ASM activity should be considered in these patients. In the present study, serum ASM activity in the sera of 15 patients with acute KD was determined both before and after treatment with infusion of high-dose intravenous immunoglobulin (IVIG), a first-line treatment for acute KD. Serum ASM activity before IVIG was significantly elevated in KD patients when compared to the control group (3.85 ± 1.46 nmol/0.1 ml/6 h vs. 1.15 ± 0.10 nmol/0.1 ml/6 h, p < 0.001), suggesting that ASM activation may be involved in the pathophysiology of this condition. Serum ASM activity before IVIG was significantly correlated with levels of C-reactive protein (p < 0.05). These results suggest the involvement of sphingolipid metabolism in the pathophysiology of KD.
Collapse
Affiliation(s)
- Yuuki Konno
- Department of Pediatrics, Akita University Graduate School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Supinski GS, Alimov AP, Wang L, Song XH, Callahan LA. Neutral sphingomyelinase 2 is required for cytokine-induced skeletal muscle calpain activation. Am J Physiol Lung Cell Mol Physiol 2015; 309:L614-24. [PMID: 26138644 DOI: 10.1152/ajplung.00141.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/01/2015] [Indexed: 12/22/2022] Open
Abstract
Calpain contributes to infection-induced diaphragm dysfunction but the upstream mechanism(s) responsible for calpain activation are poorly understood. It is known, however, that cytokines activate neutral sphingomyelinase (nSMase) and nSMase has downstream effects with the potential to increase calpain activity. We tested the hypothesis that infection-induced skeletal muscle calpain activation is a consequence of nSMase activation. We administered cytomix (20 ng/ml TNF-α, 50 U/ml IL-1β, 100 U/ml IFN-γ, 10 μg/ml LPS) to C2C12 muscle cells to simulate the effects of infection in vitro and studied mice undergoing cecal ligation puncture (CLP) as an in vivo model of infection. In cell studies, we assessed sphingomyelinase activity, subcellular calcium levels, and calpain activity and determined the effects of inhibiting sphingomyelinase using chemical (GW4869) and genetic (siRNA to nSMase2 and nSMase3) techniques. We assessed diaphragm force and calpain activity and utilized GW4869 to inhibit sphingomyelinase in mice. Cytomix increased cytosolic and mitochondrial calcium levels in C2C12 cells (P < 0.001); addition of GW4869 blocked these increases (P < 0.001). Cytomix also activated calpain, increasing calpain activity (P < 0.02), and the calpain-mediated cleavage of procaspase 12 (P < 0.001). Procaspase 12 cleavage was attenuated by either GW4869 (P < 0.001), BAPTA-AM (P < 0.001), or siRNA to nSMase2 (P < 0.001) but was unaffected by siRNA to nSMase3. GW4869 prevented CLP-induced diaphragm calpain activation and diaphragm weakness in mice. These data suggest that nSMase2 activation is required for the development of infection-induced diaphragm calpain activation and muscle weakness. As a consequence, therapies that inhibit nSMase2 in patients may prevent infection-induced skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Gerald S Supinski
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky; and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Alexander P Alimov
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky; and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Lin Wang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky; and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Xiao-Hong Song
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky; and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Leigh A Callahan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky; and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
44
|
Loehr JA, Abo-Zahrah R, Pal R, Rodney GG. Sphingomyelinase promotes oxidant production and skeletal muscle contractile dysfunction through activation of NADPH oxidase. Front Physiol 2015; 5:530. [PMID: 25653619 PMCID: PMC4300905 DOI: 10.3389/fphys.2014.00530] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/27/2014] [Indexed: 11/13/2022] Open
Abstract
Elevated concentrations of sphingomyelinase (SMase) have been detected in a variety of diseases. SMase has been shown to increase muscle derived oxidants and decrease skeletal muscle force; however, the sub-cellular site of oxidant production has not been elucidated. Using redox sensitive biosensors targeted to the mitochondria and NADPH oxidase (Nox2), we demonstrate that SMase increased Nox2-dependent ROS and had no effect on mitochondrial ROS in isolated FDB fibers. Pharmacological inhibition and genetic knockdown of Nox2 activity prevented SMase induced ROS production and provided protection against decreased force production in the diaphragm. In contrast, genetic overexpression of superoxide dismutase within the mitochondria did not prevent increased ROS production and offered no protection against decreased diaphragm function in response to SMase. Our study shows that SMase induced ROS production occurs in specific sub-cellular regions of skeletal muscle; however, the increased ROS does not completely account for the decrease in muscle function.
Collapse
Affiliation(s)
- James A Loehr
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine Houston, TX, USA
| | - Reem Abo-Zahrah
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine Houston, TX, USA
| | - Rituraj Pal
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine Houston, TX, USA
| | - George G Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine Houston, TX, USA
| |
Collapse
|
45
|
Mastronardi CA, Paz-Filho G, Zanoni M, Molano-González N, Arcos-Burgos M, Licinio J, Wong ML. Temporal gene expression in the hippocampus and peripheral organs to endotoxin-induced systemic inflammatory response in caspase-1-deficient mice. Neuroimmunomodulation 2015; 22:263-73. [PMID: 25633245 PMCID: PMC4710542 DOI: 10.1159/000368310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/05/2014] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES Caspase-1 (casp1), a key protease involved in the systemic inflammatory response syndrome (SIRS), controls the brain expression of a set of eight genes: Nos2 and Ptgs2 (nitric oxide synthase 2 and prostaglandin-endoperoxide synthase 2, two inducible enzymes), Cxcl1 and Cxcl10 (C-X-C motif chemokine ligand 1 and ligand 10), Tgtp and Gbp2 (T cell-specific GTPase 1 and guanylate-binding protein 2, two GTPases), Adamts1 (a disintegrin-like and metallopeptidase with thrombospondin type 1 motif, 1, a metalloprotease) and Il1rn (interleukin-1 receptor antagonist). Our objective was to ascertain whether casp1 also controlled the peripheral expression of these genes and, if so, to compare their central versus peripheral patterns of gene expression in immune and endocrine tissues during SIRS. METHODS Wild-type (wt) and casp1 knockout (casp1(-/-)) mice were injected with either saline or a high dose of endotoxin/lipopolysaccharide (LPS; 800 μg/mice i.p.). Saline-injected mice were immediately euthanized after injection, whereas LPS-injected mice were sacrificed 6 and 12 h after LPS administration. Hippocampal, splenic and adrenal gene expressions were determined by real-time PCR. RESULTS Overall, casp1(-/-) mice showed a lower inflammatory response than wt mice. The expression levels of powerful proinflammatory factors such as Nos2 and Ptgs2 was reduced in casp1(-/-) mice. Moreover, a hierarchical clustering analysis aimed at studying patterns of gene coexpression revealed large alterations in the hippocampal pattern of casp1(-/-) mice. Surprisingly, the expression of Adamts1 was increased in the hippocampus and adrenals of casp1(-/-) mice. CONCLUSIONS The resilience of casp1(-/-) mice to SIRS lethality is associated with a lower inflammatory response, loss of hippocampal gene coexpression patterns, and increased hippocampal Adamts1 gene expression. The latter might be beneficial for casp1(-/-) mice, since ADAMTS1 is likely to play a role in neuronal plasticity. The mechanisms described here may help the development of either novel biomarkers or therapeutic targets against SIRS/sepsis.
Collapse
Affiliation(s)
- Claudio Alberto Mastronardi
- Genomics and Predictive Medicine Group, Genome Biology Department, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Gilberto Paz-Filho
- Genomics and Predictive Medicine Group, Genome Biology Department, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Martina Zanoni
- Department of Psychiatry, University of Verona, Verona, Italy
| | - Nicolas Molano-González
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mauricio Arcos-Burgos
- Genomics and Predictive Medicine Group, Genome Biology Department, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Julio Licinio
- Mind and Brain Theme, South Australian Health and Medical Research Institute and Flinders University of South Australia, Adelaide, Australia
| | - Ma-Li Wong
- Mind and Brain Theme, South Australian Health and Medical Research Institute and Flinders University of South Australia, Adelaide, Australia
| |
Collapse
|
46
|
Beckmann N, Sharma D, Gulbins E, Becker KA, Edelmann B. Inhibition of acid sphingomyelinase by tricyclic antidepressants and analogons. Front Physiol 2014; 5:331. [PMID: 25228885 PMCID: PMC4151525 DOI: 10.3389/fphys.2014.00331] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/12/2014] [Indexed: 11/13/2022] Open
Abstract
Amitriptyline, a tricyclic antidepressant, has been used in the clinic to treat a number of disorders, in particular major depression and neuropathic pain. In the 1970s the ability of tricyclic antidepressants to inhibit acid sphingomyelinase (ASM) was discovered. The enzyme ASM catalyzes the hydrolysis of sphingomyelin to ceramide. ASM and ceramide were shown to play a crucial role in a wide range of diseases, including cancer, cystic fibrosis, diabetes, Alzheimer's disease, and major depression, as well as viral (e.g., measles virus) and bacterial (e.g., Staphylococcus aureus, Pseudomonas aeruginosa) infections. Ceramide molecules may act in these diseases by the alteration of membrane biophysics, the self-association of ceramide molecules within the cell membrane and the ultimate formation of larger ceramide-enriched membrane domains/platforms. These domains were shown to serve the clustering of certain receptors such as CD95 and may also act in the above named diseases. The potential to block the generation of ceramide by inhibiting the ASM has opened up new therapeutic approaches for the treatment of these conditions. Since amitriptyline is one of the longest used clinical drugs and side effects are well studied, it could potentially become a cheap and easily accessible medication for patients suffering from these diseases. In this review, we aim to provide an overview of current in vitro and in vivo studies and clinical trials utilizing amitriptyline to inhibit ASM and contemplate possible future applications of the drug.
Collapse
Affiliation(s)
- Nadine Beckmann
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Deepa Sharma
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Bärbel Edelmann
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| |
Collapse
|
47
|
Inhibition of endothelial lipase activity by sphingomyelin in the lipoproteins. Lipids 2014; 49:987-96. [PMID: 25167836 DOI: 10.1007/s11745-014-3944-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 08/14/2014] [Indexed: 10/24/2022]
Abstract
Endothelial lipase (EL) is a major determinant of plasma HDL concentration, its activity being inversely proportional to HDL levels. Although it is known that it preferentially acts on HDL compared to LDL and VLDL, the basis for this specificity is not known. Here we tested the hypothesis that sphingomyelin, a major phospholipid in lipoproteins is a physiological inhibitor of EL, and that the preference of the enzyme for HDL may be due to low sphingomyelin/phosphatidylcholine (PtdCho) ratio in HDL, compared to other lipoproteins. Using recombinant human EL, we showed that sphingomyelin inhibits the hydrolysis of PtdCho in the liposomes in a concentration-dependent manner. While the enzyme showed lower hydrolysis of LDL PtdCho, compared to HDL PtdCho, this difference disappeared after the degradation of lipoprotein sphingomyelin by bacterial sphingomyelinase. Analysis of molecular species of PtdCho hydrolyzed by EL in the lipoproteins showed that the enzyme preferentially hydrolyzed PtdCho containing polyunsaturated fatty acids (PUFA) such as 22:6, 20:5, 20:4 at the sn-2 position, generating the corresponding PUFA-lyso PtdCho. This specificity for PUFA-PtdCho species was not observed after depletion of sphingomyelin by sphingomyelinase. These results show that sphingomyelin not only plays a role in regulating EL activity, but also influences its specificity towards PtdCho species.
Collapse
|
48
|
Acid sphingomyelinase is activated in sickle cell erythrocytes and contributes to inflammatory microparticle generation in SCD. Blood 2014; 124:1941-50. [PMID: 25075126 DOI: 10.1182/blood-2014-01-543652] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Sphingolipids are a class of lipids containing a backbone of sphingoid bases that can be produced de novo through the reaction of palmitate and serine and further metabolized through the activity of various enzymes to produce intermediates with diverse roles in cellular processes and signal transduction. One of these intermediates, sphingosine 1-phosphate (S1P), is stored at high concentrations (1 μM) in red blood cells (RBCs) and directs a wide array of cellular processes mediated by 5 known G-protein coupled receptors (S1P1-S1P5). In this study, we show that RBC membrane alterations in sickle cell disease enhance the activation acid sphingomyelinase by 13%, resulting in increased production and storage of sphingosine (2.6-fold) and S1P (3.5-fold). We also show that acid sphingomyelinase enhances RBC-derived microparticle (MP) generation. These MPs are internalized by myeloid cells and promote proinflammatory cytokine secretion and endothelial cell adhesion, suggesting that potential crosstalk between circulating inflammatory cells and MPs may contribute to the inflammation-rooted pathogenesis of the disease. Treatment with amitriptyline reduces MP generation in vitro and in vivo and might be used to mitigate inflammatory processes in sickle cell disease.
Collapse
|
49
|
Wallace M, Morris C, O'Grada CM, Ryan M, Dillon ET, Coleman E, Gibney ER, Gibney MJ, Roche HM, Brennan L. Relationship between the lipidome, inflammatory markers and insulin resistance. ACTA ACUST UNITED AC 2014; 10:1586-95. [DOI: 10.1039/c3mb70529c] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The objectives of the present study were to (1) examine the effects of the phenotypic factors age, gender and BMI on the lipidomic profile and (2) investigate the relationship between the lipidome, inflammatory markers and insulin resistance.
Collapse
Affiliation(s)
- Martina Wallace
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research
- University College Dublin
| | - Ciara Morris
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research
- University College Dublin
| | - Colm M. O'Grada
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research
- University College Dublin
| | - Miriam Ryan
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
| | - Eugene T. Dillon
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research
- University College Dublin
| | - Eilish Coleman
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research
- University College Dublin
| | - Eileen R. Gibney
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
| | - Michael J. Gibney
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
| | - Helen M. Roche
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research
- University College Dublin
| | - Lorraine Brennan
- UCD Institute of Food and Health
- University College Dublin
- Belfield, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research
- University College Dublin
| |
Collapse
|
50
|
Jenkins RW, Clarke CJ, Lucas JT, Shabbir M, Wu BX, Simbari F, Mueller J, Hannun YA, Lazarchick J, Shirai K. Evaluation of the role of secretory sphingomyelinase and bioactive sphingolipids as biomarkers in hemophagocytic lymphohistiocytosis. Am J Hematol 2013; 88:E265-72. [PMID: 23828274 DOI: 10.1002/ajh.23535] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/24/2013] [Accepted: 06/28/2013] [Indexed: 12/27/2022]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a rare systemic inflammatory syndrome that results from unrestrained immune cell activation. Despite significant advances in the understanding of the pathophysiology of HLH, interventions remain limited for this often-fatal condition. Secretory sphingomyelinase (S-SMase) is a pro-inflammatory lipid hydrolase that is upregulated in several inflammatory conditions, including HLH. S-SMase promotes the formation of ceramide, a bioactive lipid implicated in several human disease states. However, the role of the S-SMase/ceramide pathway in HLH remains unexplored. To further evaluate the role of S-SMase upregulation in HLH, we tested the serum of patients with HLH (n = 16; primary = 3, secondary = 13) and healthy control patients (n = 25) for serum S-SMase activity with tandem sphingolipid metabolomic profiling. Patients with HLH exhibited elevated levels of serum S-SMase activity, with concomitant elevations in several ceramide species and sphingosine, while levels of sphingosine-1-phosphate were significantly decreased. Importantly, the ratio of C16 -ceramide:sphingosine was uniquely elevated in HLH patients that died despite appropriate treatment, but remained low in HLH patients that survived, suggesting that this ratio may be of prognostic significance. Together, these results demonstrate upregulation of the S-SMase/ceramide pathway in HLH, and suggest that the balance of ceramide and sphingosine determine clinical outcomes in HLH. .
Collapse
Affiliation(s)
- Russell W. Jenkins
- Department of Medicine; Massachusetts General Hospital; Boston Massachusetts
| | | | - John Thomas Lucas
- Department of Radiation Oncology; Wake Forest University; Winston-Salem North Carolina
| | - Munira Shabbir
- Department of Medicine; Division of Hematology and Oncology; Medical University of South Carolina; Charleston South Carolina
- Aga Khan University Hospital; Karachi Pakistan
| | - Bill X. Wu
- Department of Microbiology and Immunology; Medical University of South Carolina; Charleston South Carolina
| | - Fabio Simbari
- Department of Biomedicinal Chemistry; Institute for Advanced Chemistry of Catalonia (CSIC); Jordi Girona 18-26 Barcelona Spain
| | - Joan Mueller
- Department of Pathology; Medical University of South Carolina; Charleston South Carolina
| | - Yusuf A. Hannun
- Department of Medicine; Stony Brook University; Stony Brook New York
| | - John Lazarchick
- Department of Pathology; Medical University of South Carolina; Charleston South Carolina
| | - Keisuke Shirai
- Department of Medicine; Division of Hematology and Oncology; Medical University of South Carolina; Charleston South Carolina
| |
Collapse
|