151
|
Alaamery M, Albesher N, Aljawini N, Alsuwailm M, Massadeh S, Wheeler MA, Chao CC, Quintana FJ. Role of sphingolipid metabolism in neurodegeneration. J Neurochem 2020; 158:25-35. [PMID: 32402091 DOI: 10.1111/jnc.15044] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/15/2020] [Accepted: 04/19/2020] [Indexed: 12/21/2022]
Abstract
Sphingolipids are a class of lipids highly enriched in the central nervous system (CNS), which shows great diversity and complexity, and has been implicated in CNS development and function. Alterations in sphingolipid metabolism have been described in multiple diseases, including those affecting the central nervous system (CNS). In this review, we discuss the role of sphingolipid metabolism in neurodegeneration, evaluating its direct roles in neuron development and health, and also in the induction of neurotoxic activities in CNS-resident astrocytes and microglia in the context of neurologic diseases such as multiple sclerosis and Alzheimer's disease. Finally, we focus on the metabolism of gangliosides and sphingosine-1-phosphate, its contribution to the pathogenesis of neurologic diseases, and its potential as a candidate target for the therapeutic modulation of neurodegeneration.
Collapse
Affiliation(s)
- Manal Alaamery
- KACST-BWH Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia.,Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Nour Albesher
- KACST-BWH Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia.,Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Nora Aljawini
- KACST-BWH Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia.,Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Moneera Alsuwailm
- KACST-BWH Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia.,Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Salam Massadeh
- KACST-BWH Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia.,Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Chun-Cheih Chao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, USA
| |
Collapse
|
152
|
Sudhadevi T, Ha AW, Ebenezer DL, Fu P, Putherickal V, Natarajan V, Harijith A. Advancements in understanding the role of lysophospholipids and their receptors in lung disorders including bronchopulmonary dysplasia. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158685. [PMID: 32169655 PMCID: PMC7206974 DOI: 10.1016/j.bbalip.2020.158685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/25/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a devastating chronic neonatal lung disease leading to serious adverse consequences. Nearly 15 million babies are born preterm accounting for >1 in 10 births globally. The aetiology of BPD is multifactorial and the survivors suffer lifelong respiratory morbidity. Lysophospholipids (LPL), which include sphingosine-1-phosphate (S1P), and lysophosphatidic acid (LPA) are both naturally occurring bioactive lipids involved in a variety of physiological and pathological processes such as cell survival, death, proliferation, migration, immune responses and vascular development. Altered LPL levels have been observed in a number of lung diseases including BPD, which underscores the importance of these signalling lipids under normal and pathophysiological situations. Due to the paucity of information related to LPLs in BPD, most of the ideas related to BPD and LPL are speculative. This article is intended to promote discussion and generate hypotheses, in addition to the limited review of information related to BPD already established in the literature.
Collapse
Affiliation(s)
- Tara Sudhadevi
- Department of Pediatrics, University of Illinois, Chicago, IL, United States of America
| | - Alison W Ha
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, United States of America
| | - David L Ebenezer
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, United States of America
| | - Panfeng Fu
- Department of Pharmacology, University of Illinois, Chicago, IL, United States of America
| | - Vijay Putherickal
- Department of Pharmacology, University of Illinois, Chicago, IL, United States of America
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois, Chicago, IL, United States of America; Department of Medicine, University of Illinois, Chicago, IL, United States of America
| | - Anantha Harijith
- Department of Pediatrics, University of Illinois, Chicago, IL, United States of America; Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, United States of America; Department of Pharmacology, University of Illinois, Chicago, IL, United States of America.
| |
Collapse
|
153
|
Canals D, Salamone S, Santacreu BJ, Nemeth E, Aguilar D, Hernandez-Corbacho MJ, Adada M, Staquicini DI, Arap W, Pasqualini R, Haley J, Obeid LM, Hannun YA. Ceramide launches an acute anti-adhesion pro-migration cell signaling program in response to chemotherapy. FASEB J 2020; 34:7610-7630. [PMID: 32307766 PMCID: PMC8265206 DOI: 10.1096/fj.202000205r] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
Chemotherapy has been reported to upregulate sphingomylinases and increase cellular ceramide, often linked to the induction to cell death. In this work, we show that sublethal doses of doxorubicin and vorinostat still increased cellular ceramide, which was located predominantly at the plasma membrane. To interrogate possible functions of this specific pool of ceramide, we used recombinant enzymes to mimic physiological levels of ceramide at the plasma membrane upon chemotherapy treatment. Using mass spectrometry and network analysis, followed by experimental confirmation, the results revealed that this pool of ceramide acutely regulates cell adhesion and cell migration pathways with weak connections to commonly established ceramide functions (eg, cell death). Neutral sphingomyelinase 2 (nSMase2) was identified as responsible for the generation of plasma membrane ceramide upon chemotherapy treatment, and both ceramide at the plasma membrane and nSMase2 were necessary and sufficient to mediate these "side" effects of chemotherapy on cell adhesion and migration. This is the first time a specific pool of ceramide is interrogated for acute signaling functions, and the results define plasma membrane ceramide as an acute signaling effector necessary and sufficient for regulation of cell adhesion and cell migration under chemotherapeutical stress.
Collapse
Affiliation(s)
- Daniel Canals
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Silvia Salamone
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Bruno Jaime Santacreu
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
- Facultad de Farmacia y Bioquímica, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina
| | - Erika Nemeth
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Daniel Aguilar
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Barcelona, Catalunya, Spain
| | | | - Mohamad Adada
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Daniela I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, United States
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ, United States
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, United States
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - John Haley
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
- Northport VA Hospital
- Stony Brook Cancer Center, Stony Brook, NY, United States
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
- Department of Biochemistry, Stony Brook University
- Stony Brook Cancer Center, Stony Brook, NY, United States
| |
Collapse
|
154
|
Yeast Sphingolipid Phospholipase Gene ISC1 Regulates the Spindle Checkpoint by a CDC55-Dependent Mechanism. Mol Cell Biol 2020; 40:MCB.00340-19. [PMID: 32205408 DOI: 10.1128/mcb.00340-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 03/18/2020] [Indexed: 01/10/2023] Open
Abstract
Defects in the spindle assembly checkpoint (SAC) can lead to aneuploidy and cancer. Sphingolipids have important roles in many cellular functions, including cell cycle regulation and apoptosis. However, the specific mechanisms and functions of sphingolipids in cell cycle regulation have not been elucidated. Using analysis of concordance for synthetic lethality for the yeast sphingolipid phospholipase ISC1, we identified two groups of genes. The first comprises genes involved in chromosome segregation and stability (CSM3, CTF4, YKE2, DCC1, and GIM4) as synthetically lethal with ISC1 The second group, to which ISC1 belongs, comprises genes involved in the spindle checkpoint (BUB1, MAD1, BIM1, and KAR3), and they all share the same synthetic lethality with the first group. We demonstrate that spindle checkpoint genes act upstream of Isc1, and their deletion phenocopies that of ISC1 Reciprocally, ISC1 deletion mutants were sensitive to benomyl, indicating a SAC defect. Similar to BUB1 deletion, ISC1 deletion prevents spindle elongation in hydroxyurea-treated cells. Mechanistically, PP2A-Cdc55 ceramide-activated phosphatase was found to act downstream of Isc1, thus coupling the spindle checkpoint genes and Isc1 to CDC55-mediated nuclear functions.
Collapse
|
155
|
Dai L, Chen J, Lin Z, Wang Z, Mu S, Qin Z. Targeting Sphingosine Kinase by ABC294640 against Diffuse Intrinsic Pontine Glioma (DIPG). J Cancer 2020; 11:4683-4691. [PMID: 32626514 PMCID: PMC7330698 DOI: 10.7150/jca.46269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/11/2020] [Indexed: 01/29/2023] Open
Abstract
As a highly aggressive pediatric brainstem tumor, diffuse intrinsic pontine glioma (DIPG) accounts for 10% to 20% of childhood brain tumors. The survival rate for DIPG remains very low, with a median survival time as less than one year even under radiotherapy, the current standard treatment. Moreover, over than 250 clinical trials have failed when trying to improve the survival compared to radiotherapy. The sphingolipid metabolism and related signaling pathways have been found closely related to cancer cell survival; however, the sphingolipid metabolism targeted therapies have never been investigated in DIPG. In the current study, the anti-DIPG activity of ABC294640, the only first-in-class orally available Sphingosine kinase (SphK) inhibitor was explored. Treatment with ABC294640 significantly repressed DIPG cell growth by inducing intracellular pro-apoptotic ceramides production and cell apoptosis. We also profiled ABC294640-induced changes in gene expression within DIPG cells and identified many new genes tightly controlled by sphingolipid metabolism, such as IFITM1 and KAL1. These genes are required for DIPG cell survival and display clinical relevance in DIPG patients' samples. Together, our findings in this study indicate that targeting sphingolipid metabolism may represent a promising strategy to improve DIPG treatment.
Collapse
Affiliation(s)
- Lu Dai
- Departments of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR 72205, USA
| | - Jungang Chen
- Departments of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR 72205, USA
| | - Zhen Lin
- Department of Pathology, Tulane University Health Sciences Center, Tulane Cancer Center, 1700 Tulane Ave., New Orleans, LA 70112, USA
| | - Zhaoxiong Wang
- Department of Pathology, Tulane University Health Sciences Center, Tulane Cancer Center, 1700 Tulane Ave., New Orleans, LA 70112, USA
| | - Shengyu Mu
- Pharmacology & Toxicology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR 72205, USA
| | - Zhiqiang Qin
- Departments of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR 72205, USA
| |
Collapse
|
156
|
Correnti J, Lin C, Brettschneider J, Kuriakose A, Jeon S, Scorletti E, Oranu A, McIver-Jenkins D, Kaneza I, Buyco D, Saiman Y, Furth EE, Argemi J, Bataller R, Holland WL, Carr RM. Liver-specific ceramide reduction alleviates steatosis and insulin resistance in alcohol-fed mice. J Lipid Res 2020; 61:983-994. [PMID: 32398264 PMCID: PMC7328039 DOI: 10.1194/jlr.ra119000446] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/29/2020] [Indexed: 12/20/2022] Open
Abstract
Alcohol's impairment of both hepatic lipid metabolism and insulin resistance (IR) are key drivers of alcoholic steatosis, the initial stage of alcoholic liver disease (ALD). Pharmacologic reduction of lipotoxic ceramide prevents alcoholic steatosis and glucose intolerance in mice, but potential off-target effects limit its strategic utility. Here, we employed a hepatic-specific acid ceramidase (ASAH) overexpression model to reduce hepatic ceramides in a Lieber-DeCarli model of experimental alcoholic steatosis. We examined effects of alcohol on hepatic lipid metabolism, body composition, energy homeostasis, and insulin sensitivity as measured by hyperinsulinemic-euglycemic clamp. Our results demonstrate that hepatic ceramide reduction ameliorates the effects of alcohol on hepatic lipid droplet (LD) accumulation by promoting VLDL secretion and lipophagy, the latter of which involves ceramide cross-talk between the lysosomal and LD compartments. We additionally demonstrate that hepatic ceramide reduction prevents alcohol's inhibition of hepatic insulin signaling. These effects on the liver are associated with a reduction in oxidative stress markers and are relevant to humans, as we observe peri- LD ASAH expression in human ALD. Together, our results suggest a potential role for hepatic ceramide inhibition in preventing ALD.
Collapse
Affiliation(s)
- Jason Correnti
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Chelsea Lin
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | | | - Amy Kuriakose
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Sookyoung Jeon
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Eleonora Scorletti
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Amanke Oranu
- Division of Gastroenterology, United Health Services, Binghamton, NY
| | - Dru McIver-Jenkins
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Isabelle Kaneza
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Delfin Buyco
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Yedidya Saiman
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Emma E Furth
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Josepmaria Argemi
- Center for Liver Diseases, Pittsburgh Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Ramon Bataller
- Center for Liver Diseases, Pittsburgh Research Center, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| | - Rotonya M Carr
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA. mailto:
| |
Collapse
|
157
|
Chabowski DS, Cohen KE, Abu-Hatoum O, Gutterman DD, Freed JK. Crossing signals: bioactive lipids in the microvasculature. Am J Physiol Heart Circ Physiol 2020; 318:H1185-H1197. [PMID: 32243770 PMCID: PMC7541955 DOI: 10.1152/ajpheart.00706.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The primary function of the arterial microvasculature is to ensure that regional perfusion of blood flow is matched to the needs of the tissue bed. This critical physiological mechanism is tightly controlled and regulated by a variety of vasoactive compounds that are generated and released from the vascular endothelium. Although these substances are required for modulating vascular tone, they also influence the surrounding tissue and have an overall effect on vascular, as well as parenchymal, homeostasis. Bioactive lipids, fatty acid derivatives that exert their effects through signaling pathways, are included in the list of vasoactive compounds that modulate the microvasculature. Although lipids were identified as important vascular messengers over three decades ago, their specific role within the microvascular system is not well defined. Thorough understanding of these pathways and their regulation is not only essential to gain insight into their role in cardiovascular disease but is also important for preventing vascular dysfunction following cancer treatment, a rapidly growing problem in medical oncology. The purpose of this review is to discuss how biologically active lipids, specifically prostanoids, epoxyeicosatrienoic acids, sphingolipids, and lysophospholipids, contribute to vascular function and signaling within the endothelium. Methods for quantifying lipids will be briefly discussed, followed by an overview of the various lipid families. The cross talk in signaling between classes of lipids will be discussed in the context of vascular disease. Finally, the potential clinical implications of these lipid families will be highlighted.
Collapse
Affiliation(s)
- Dawid S. Chabowski
- 1Division of Cardiology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin,2Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Katie E. Cohen
- 1Division of Cardiology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin,2Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ossama Abu-Hatoum
- 4Department of Surgery, HaEmek Medical Center, Technion Medical School, Haifa, Israel
| | - David D. Gutterman
- 1Division of Cardiology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin,2Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Julie K. Freed
- 2Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin,3Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
158
|
Bergman BC, Goodpaster BH. Exercise and Muscle Lipid Content, Composition, and Localization: Influence on Muscle Insulin Sensitivity. Diabetes 2020; 69:848-858. [PMID: 32312901 DOI: 10.2337/dbi18-0042] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/17/2020] [Indexed: 11/13/2022]
Abstract
Accumulation of lipid in skeletal muscle is thought to be related to the development of insulin resistance and type 2 diabetes. Initial work in this area focused on accumulation of intramuscular triglyceride; however, bioactive lipids such as diacylglycerols and sphingolipids are now thought to play an important role. Specific species of these lipids appear to be more negative toward insulin sensitivity than others. Adding another layer of complexity, localization of lipids within the cell appears to influence the relationship between these lipids and insulin sensitivity. This article summarizes how accumulation of total lipids, specific lipid species, and localization of lipids influence insulin sensitivity in humans. We then focus on how these aspects of muscle lipids are impacted by acute and chronic aerobic and resistance exercise training. By understanding how exercise alters specific species and localization of lipids, it may be possible to uncover specific lipids that most heavily impact insulin sensitivity.
Collapse
|
159
|
Yang Y, Yang X, Lin Y, Yang G, Li L. LASS2 regulates hepatocyte steatosis by interacting with NDUFS2/OXPHOS related proteins. Biochem Biophys Res Commun 2020; 526:871-879. [PMID: 32279995 DOI: 10.1016/j.bbrc.2020.02.166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 02/27/2020] [Indexed: 12/30/2022]
Abstract
LAG1 longevity assurance homolog 2 (LASS2), a highly conserved transmembrane protein, has been reported to be associated with nonalcoholic fatty liver disease (NAFLD). However, the effect of LASS2 on energy homeostasis and its mechanism remains unknown. In this study, we found lower expression levels of LASS2 in the livers of mice with liver steatosis induced by a high-fat diet (HFD) and free fatty acids (FFAs)-treated hepatocytes. In FFAs-treated Hepa1-6 cells and mouse primary hepatocytes (MPHs), LASS2 overexpression significantly decreased intracellular lipid content compared with the control cells. LASS2 overexpression also significantly upregulated lipolysis-related proteins, such as ATGL and HSL, and inhibited lipogenesis-related proteins, such as SREBP1 and FAS. In addition, the phosphorylation levels of AMPK and ACC increased significantly. On the contrary, LASS2 knockdown in FFAs-treated hepatocytes aggravated lipid accumulation via facilitating lipogenesis and inhibiting lipolysis. Further, co-IP and LC-MS analysis found that LASS2 might interacted with NDUFS2 to inhibit lipogenesis. The production of mitochondrial reactive oxygen species (mtROS) may be related to the interaction between LASS2 and NDUFS2. Collectively, we are the first time to showed that LASS2 might promote the phosphorylation of AMPK via mtROS produced by interaction with NDUFS2/OXPHOS, thus inhibiting lipogenesis.
Collapse
Affiliation(s)
- Yan Yang
- The Key Laboratory of Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - XiaoLi Yang
- Department of Clinical Laboratory, Affiliated Hospital of Zunyi Medical University, ZunYi, Guizhou, 563003, People's Republic of China; College of Laboratory Medicine, Zunyi Medical University, Zunyi, Guizhou, 563003, People's Republic of China
| | - Yao Lin
- The Key Laboratory of Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - GangYi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Ling Li
- The Key Laboratory of Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
160
|
Iessi E, Marconi M, Manganelli V, Sorice M, Malorni W, Garofalo T, Matarrese P. On the role of sphingolipids in cell survival and death. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:149-195. [PMID: 32247579 DOI: 10.1016/bs.ircmb.2020.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sphingolipids, universal components of biological membranes of all eukaryotic organisms, from yeasts to mammals, in addition of playing a structural role, also play an important part of signal transduction pathways. They participate or, also, ignite several fundamental subcellular signaling processes but, more in general, they directly contribute to key biological activities such as cell motility, growth, senescence, differentiation as well as cell fate, i.e., survival or death. The sphingolipid metabolic pathway displays an intricate network of reactions that result in the formation of multiple sphingolipids, including ceramide, and sphingosine-1-phosphate. Different sphingolipids, that have key roles in determining cell fate, can induce opposite effects: as a general rule, sphingosine-1-phosphate promotes cell survival and differentiation, whereas ceramide is known to induce apoptosis. Furthermore, together with cholesterol, sphingolipids also represent the basic lipid component of lipid rafts, cholesterol- and sphingolipid-enriched membrane microdomains directly involved in cell death and survival processes. In this review, we briefly describe the characteristics of sphingolipids and lipid membrane microdomains. In particular, we will consider the involvement of various sphingolipids per se and of lipid rafts in apoptotic pathway, both intrinsic and extrinsic, in nonapoptotic cell death, in autophagy, and in cell differentiation. In addition, their roles in the most common physiological and pathological contexts either as pathogenetic elements or as biomarkers of diseases will be considered. We would also hint how the manipulation of sphingolipid metabolism could represent a potential therapeutic target to be investigated and functionally validated especially for those diseases for which therapeutic options are limited or ineffective.
Collapse
Affiliation(s)
- Elisabetta Iessi
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Matteo Marconi
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | | | - Maurizio Sorice
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Walter Malorni
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy; Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Tina Garofalo
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
161
|
Barron KA, Jeffries KA, Krupenko NI. Sphingolipids and the link between alcohol and cancer. Chem Biol Interact 2020; 322:109058. [PMID: 32171848 DOI: 10.1016/j.cbi.2020.109058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/20/2019] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Epidemiological evidence underscores alcohol consumption as a strong risk factor for multiple cancer types, with liver cancer being most commonly associated with alcohol intake. While mechanisms linking alcohol consumption to malignant tumor development are not fully understood, the likely players in ethanol-induced carcinogenesis are genotoxic stress caused by formation of acetaldehyde, increased oxidative stress, and altered nutrient metabolism, including the impairment of methyl transfer reactions. Alterations of sphingolipid metabolism and associated signaling pathways are another potential link between ethanol and cancer development. In particular, ceramides are involved in the regulation of cellular proliferation, differentiation, senescence, and apoptosis and are known to function as important regulators of malignant transformation as well as tumor progression. However, to date, the cross-talk between ceramides and alcohol in cancer disease is largely an open question and only limited data are available on this subject. Most studies linking ceramide to cancer considered liver steatosis as the underlying mechanism, which is not surprising taking into consideration that ceramide pathways are an integral part of the overall lipid metabolism. This review summarizes the latest studies pointing to ceramide as an important mediator of cancer-promoting effects of chronic alcohol consumption and underscores the necessity of understanding the role of sphingolipids and lipid signaling in response to alcohol in order to prevent and/or successfully manage diseases caused by alcohol.
Collapse
Affiliation(s)
| | | | - Natalia I Krupenko
- Department of Nutrition, UNC Chapel Hill, USA; Nutrition Research Institute, UNC Chapel Hill, USA.
| |
Collapse
|
162
|
Garate J, Lage S, Martín-Saiz L, Perez-Valle A, Ochoa B, Boyano MD, Fernández R, Fernández JA. Influence of Lipid Fragmentation in the Data Analysis of Imaging Mass Spectrometry Experiments. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:517-526. [PMID: 32126773 DOI: 10.1021/jasms.9b00090] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Imaging mass spectrometry (IMS) is becoming an essential technique in lipidomics. Still, many questions remain open, precluding it from achieving its full potential. Among them, identification of species directly from the tissue is of paramount importance. However, it is not an easy task, due to the abundance and variety of lipid species, their numerous fragmentation pathways, and the formation of a significant number of adducts, both with the matrix and with the cations present in the tissue. Here, we explore the fragmentation pathways of 17 lipid classes, demonstrating that in-source fragmentation hampers identification of some lipid species. Then, we analyze what type of adducts each class is more prone to form. Finally, we use that information together with data from on-tissue MS/MS and MS3 to refine the peak assignment in a real experiment over sections of human nevi, to demonstrate that statistical analysis of the data is significantly more robust if unwanted peaks due to fragmentation, matrix, and other species that only introduce noise in the analysis are excluded.
Collapse
Affiliation(s)
| | | | | | | | | | - M Dolores Boyano
- Health Research Institute, Cruces University Hospital, Barakaldo, Spain
| | | | | |
Collapse
|
163
|
Jeon S, Carr R. Alcohol effects on hepatic lipid metabolism. J Lipid Res 2020; 61:470-479. [PMID: 32029510 DOI: 10.1194/jlr.r119000547] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
Alcoholic liver disease (ALD) is the most prevalent type of chronic liver disease with significant morbidity and mortality worldwide. ALD begins with simple hepatic steatosis and progresses to alcoholic steatohepatitis, fibrosis, and cirrhosis. The severity of hepatic steatosis is highly associated with the development of later stages of ALD. This review explores the disturbances of alcohol-induced hepatic lipid metabolism through altered hepatic lipid uptake, de novo lipid synthesis, fatty acid oxidation, hepatic lipid export, and lipid droplet formation and catabolism. In addition, we review emerging data on the contributions of genetics and bioactive lipid metabolism in alcohol-induced hepatic lipid accumulation.
Collapse
Affiliation(s)
- Sookyoung Jeon
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| | - Rotonya Carr
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
164
|
Amorim NML, Kee A, Coster ACF, Lucas C, Bould S, Daniel S, Weir JM, Mellett NA, Barbour J, Meikle PJ, Cohn RJ, Turner N, Hardeman EC, Simar D. Irradiation impairs mitochondrial function and skeletal muscle oxidative capacity: significance for metabolic complications in cancer survivors. Metabolism 2020; 103:154025. [PMID: 31765667 DOI: 10.1016/j.metabol.2019.154025] [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: 08/22/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Metabolic complications are highly prevalent in cancer survivors treated with irradiation but the underlying mechanisms remain unknown. METHODS Chow or high fat-fed C57Bl/6J mice were irradiated (6Gy) before investigating the impact on whole-body or skeletal muscle metabolism and profiling their lipidomic signature. Using a transgenic mouse model (Tg:Pax7-nGFP), we isolated muscle progenitor cells (satellite cells) and characterised their metabolic functions. We recruited childhood cancer survivors, grouped them based on the use of total body irradiation during their treatment and established their lipidomic profile. RESULTS In mice, irradiation delayed body weight gain and impaired fat pads and muscle weights. These changes were associated with impaired whole-body fat oxidation in chow-fed mice and altered ex vivo skeletal muscle fatty acid oxidation, potentially due to a reduction in oxidative fibres and reduced mitochondrial enzyme activity. Irradiation led to fasting hyperglycaemia and impaired glucose uptake in isolated skeletal muscles. Cultured satellite cells from irradiated mice showed decreased fatty acid oxidation and reduced glucose uptake, recapitulating the host metabolic phenotype. Irradiation resulted in a remodelling of lipid species in skeletal muscles, with the extensor digitorum longus muscle being particularly affected. A large number of lipid species were reduced, with several of these species showing a positive correlation with mitochondrial enzymes activity. In cancer survivors exposed to irradiation, we found a similar decrease in systemic levels of most lipid species, and lipid species that increased were positively correlated with insulin resistance (HOMA-IR). CONCLUSION Irradiation leads to long-term alterations in body composition, and lipid and carbohydrate metabolism in skeletal muscle, and affects muscle progenitor cells. Such changes result in persistent impairment of metabolic functions, providing a new mechanism for the increased prevalence of metabolic diseases reported in irradiated individuals. In this context, changes in the lipidomic signature in response to irradiation could be of diagnostic value.
Collapse
Affiliation(s)
- Nadia M L Amorim
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Anthony Kee
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Adelle C F Coster
- School of Mathematics and Statistics, UNSW Sydney, Sydney, Australia
| | - Christine Lucas
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sarah Bould
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sara Daniel
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Jacquelyn M Weir
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Jayne Barbour
- Mitochondrial Bioenergetics Lab, Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Richard J Cohn
- School of Women's and Children's Health, UNSW Sydney, Randwick, Australia; Kids Cancer Centre, Sydney Children's Hospital Network, Randwick, Australia
| | - Nigel Turner
- Mitochondrial Bioenergetics Lab, Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Edna C Hardeman
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia.
| | - David Simar
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, Australia.
| |
Collapse
|
165
|
Riboni L, Abdel Hadi L, Navone SE, Guarnaccia L, Campanella R, Marfia G. Sphingosine-1-Phosphate in the Tumor Microenvironment: A Signaling Hub Regulating Cancer Hallmarks. Cells 2020; 9:E337. [PMID: 32024090 PMCID: PMC7072483 DOI: 10.3390/cells9020337] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
As a key hub of malignant properties, the cancer microenvironment plays a crucial role intimately connected to tumor properties. Accumulating evidence supports that the lysophospholipid sphingosine-1-phosphate acts as a key signal in the cancer extracellular milieu. In this review, we have a particular focus on glioblastoma, representative of a highly aggressive and deleterious neoplasm in humans. First, we highlight recent advances and emerging concepts for how tumor cells and different recruited normal cells contribute to the sphingosine-1-phosphate enrichment in the cancer microenvironment. Then, we describe and discuss how sphingosine-1-phosphate signaling contributes to favor cancer hallmarks including enhancement of proliferation, stemness, invasion, death resistance, angiogenesis, immune evasion and, possibly, aberrant metabolism. We also discuss the potential of how sphingosine-1-phosphate control mechanisms are coordinated across distinct cancer microenvironments. Further progress in understanding the role of S1P signaling in cancer will depend crucially on increasing knowledge of its participation in the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Loubna Abdel Hadi
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
- Department of Clinical Sciences and Community Health, University of Milan, 20100 Milan, Italy
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| |
Collapse
|
166
|
Targeting alkaline ceramidase 3 alleviates the severity of nonalcoholic steatohepatitis by reducing oxidative stress. Cell Death Dis 2020; 11:28. [PMID: 31949129 PMCID: PMC6965144 DOI: 10.1038/s41419-019-2214-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 01/15/2023]
Abstract
Overload of palmitic acids is linked to the dysregulation of ceramide metabolism in nonalcoholic steatohepatitis (NASH), and ceramides are important bioactive lipids mediating the lipotoxicity of palmitic acid in NASH. However, much remains unclear about the role of ceramidases that catalyze the hydrolysis of ceramides in NASH. By analyzing the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) database, we found that alkaline ceramidase 3 (ACER3) is upregulated in livers of patients with NASH. Consistently, we found that Acer3 mRNA levels and its enzymatic activity were also upregulated in mouse livers with NASH induced by a palmitate-enriched Western diet (PEWD). Moreover, we demonstrated that palmitate treatment also elevated Acer3 mRNA levels and its enzymatic activity in mouse primary hepatocytes. In order to investigate the function of Acer3 in NASH, Acer3 null mice and their wild-type littermates were fed a PEWD to induce NASH. Knocking out Acer3 was found to augment PEWD-induced elevation of C18:1-ceramide and alleviate early inflammation and fibrosis but not steatosis in mouse livers with NASH. In addition, Acer3 deficiency attenuated hepatocyte apoptosis in livers with NASH. These protective effects of Acer3 deficiency were found to be associated with suppression of hepatocellular oxidative stress in NASH liver. In vitro studies further revealed that loss of ACER3/Acer3 increased C18:1-ceramide and inhibited apoptosis and oxidative stress in mouse primary hepatocytes and immortalized human hepatocytes induced by palmitic-acid treatment. These results suggest that ACER3 plays an important pathological role in NASH by mediating palmitic-acid-induced oxidative stress.
Collapse
|
167
|
Sukocheva OA, Lukina E, McGowan E, Bishayee A. Sphingolipids as mediators of inflammation and novel therapeutic target in inflammatory bowel disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 120:123-158. [PMID: 32085881 DOI: 10.1016/bs.apcsb.2019.11.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Morbidity of inflammatory gastrointestinal (GI) diseases continues to grow resulting in worsen quality of life and increased burden on public medical systems. Complex and heterogenous illnesses, inflammatory bowel diseases (IBDs) encompass several inflammation -associated pathologies including Crohn's disease and ulcerative colitis. IBD is often initiated by a complex interplay between host genetic and environmental factors, lifestyle and diet, and intestinal bacterial components. IBD inflammatory signature was linked to the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) signaling pathway that is currently targeted by IBD therapies. Sphingolipid signaling was identified as one of the key mediators and regulators of pro-inflammatory conditions, and, specifically, TNF-α related signaling. All GI tissues and circulating immune/blood cells contain activated sphingolipid-metabolizing enzymes, including sphingosine kinases (SphK1 and SphK2) that generate sphingosine-1-phosphate (S1P), a bioactive lipid and ligand for five G-protein coupled membrane S1P receptors (S1PRs). Numerous normal and pathogenic inflammatory responses are mediated by SphK/S1P/S1PRs signaling axis including lymphocyte trafficking and activation of cytokine signaling machinery. SphK1/S1P/S1PRs axis has recently been defined as a target for the treatment of GI diseases including IBD/colitis. Several SphK1 inhibitors and S1PRs antagonists have been developed as novel anti-inflammatory agents. In this review, we discuss the mechanisms of SphK/S1P signaling in inflammation-linked GI disorders. The potential role of SphK/S1PRs inhibitors in the prevention and treatment of IBD/colitis is critically evaluated.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, Australia
| | - Elena Lukina
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, Australia
| | - Eileen McGowan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| |
Collapse
|
168
|
Dai GY, Yin J, Li KE, Chen DK, Liu Z, Bi FC, Rong C, Yao N. The Arabidopsis AtGCD3 protein is a glucosylceramidase that preferentially hydrolyzes long-acyl-chain glucosylceramides. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49930-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
169
|
Abstract
Sphingosine, ceramide, sphingosine-1-phosphate, and other related sphingolipids have emerged as important bioactive molecules involved in a variety of key cellular processes such as cell growth, differentiation, apoptosis, exosome release, and inter- and intracellular cell communication, making the pathways of sphingolipid metabolism a key domain in maintaining cell homeostasis (Hannun and Obeid, Trends Biochem Sci 20:73-77, 1995; Hannun and Obeid, Nat Rev Mol Cell Biol 9:139-150, 2008; Kosaka et al., J Biol Chem 288:10849-10859, 2013). Various studies have determined that these pathways play a central role in regulating intracellular production of ceramide and the other bioactive sphingolipids and hence are an important component of signaling in various diseases such as cancer, diabetes, and neurodegenerative and cardiovascular diseases (Chaube et al., Biochim Biophys Acta 1821:313-323, 2012; Clarke et al., Adv Enzyme Regul 51:51-58, 2011b; Horres and Hannun, Neurochem Res 37:1137-1149, 2012). In this chapter, we discuss one of the major enzyme classes in producing ceramide, sphingomyelinases (SMases), from a biochemical and structural perspective with an emphasis on their applicability as therapeutic targets.
Collapse
Affiliation(s)
- Prajna Shanbhogue
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Yusuf A Hannun
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.
- Stony Brook University Cancer Center, Stony Brook, NY, USA.
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
170
|
Gaboon NE, Banaganapalli B, Nasser K, Razeeth M, Alsaedi MS, Rashidi OM, Abdelwehab LS, Alahmadi TS, Safdar OY, Shaik J, Choudhry HM, Al-numan HH, Khan MI, Al-Aama JY, Elango R, Shaik NA. Exome sequencing and metabolomic analysis of a chronic kidney disease and hearing loss patient family revealed RMND1 mutation induced sphingolipid metabolism defects. Saudi J Biol Sci 2020; 27:324-334. [PMID: 31889854 PMCID: PMC6933272 DOI: 10.1016/j.sjbs.2019.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial disorders (MIDs) shows overlapping clinical presentations owing to the genetic and metabolic defects of mitochondria. However, specific relationship between inherited mutations in nuclear encoded mitochondrial proteins and their functional impacts in terms of metabolic defects in patients is not yet well explored. Therefore, using high throughput whole exome sequencing (WES), we screened a chronic kidney disease (CKD) and sensorineural hearing loss (SNHL) patient, and her family members to ascertain the mode of inheritance of the mutation, and healthy population controls to establish its rare frequency. The impact of mutation on biophysical characteristics of the protein was further studied by mapping it in 3D structure. Furthermore, LC-MS tandem mass spectrophotometry based untargeted metabolomic profiling was done to study the fluctuations in plasma metabolites relevant to disease causative mutations and kidney damage. We identified a very rare homozygous c.631G > A (p.Val211Met) pathogenic mutation in RMND1 gene in the proband, which is inherited in an autosomal recessive fashion. This gene is involved in the mitochondrial translational pathways and contribute in mitochondrial energy metabolism. The p.Val211Met mutation is found to disturb the structural orientation (RMSD is -2.95 Å) and stability (ΔΔG is -0.552 Kcal/mol) of the RMND1 protein. Plasma metabolomics analysis revealed the aberrant accumulation of metabolites connected to lipid and amino acid metabolism pathways. Of these metabolites, pathway networking has discovered ceramide, a metabolite of sphingolipids, which plays a role in different signaling cascades including mitochondrial membrane biosynthesis, is highly elevated in this patient. This study suggests that genetic defects in RMND1 gene alters the mitochondrial energy metabolism leading to the accumulation of ceramide, and subsequently promote dysregulated apoptosis and tissue necrosis in kidneys.
Collapse
Affiliation(s)
- Nagwa E.A. Gaboon
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Medical Genetics Centre, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Babajan Banaganapalli
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalidah Nasser
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Razeeth
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mosab S. Alsaedi
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Omran M. Rashidi
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Turki Saad Alahmadi
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Osama Y. Safdar
- Pediatric Nephrology Center of Excellence, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jilani Shaik
- Genome Research Chair, College of Science, King Saud University, Saudi Arabia
| | - Hani M.Z. Choudhry
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda Husain Al-numan
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Jumana Y. Al-Aama
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noor A. Shaik
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
171
|
Druggable Sphingolipid Pathways: Experimental Models and Clinical Opportunities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:101-135. [PMID: 32894509 DOI: 10.1007/978-3-030-50621-6_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Intensive research in the field of sphingolipids has revealed diverse roles in cell biological responses and human health and disease. This immense molecular family is primarily represented by the bioactive molecules ceramide, sphingosine, and sphingosine 1-phosphate (S1P). The flux of sphingolipid metabolism at both the subcellular and extracellular levels provides multiple opportunities for pharmacological intervention. The caveat is that perturbation of any single node of this highly regulated flux may have effects that propagate throughout the metabolic network in a dramatic and sometimes unexpected manner. Beginning with S1P, the receptors for which have thus far been the most clinically tractable pharmacological targets, this review will describe recent advances in therapeutic modulators targeting sphingolipids, their chaperones, transporters, and metabolic enzymes.
Collapse
|
172
|
Abstract
There is substantial evidence that the enzymes, sphingosine kinase 1 and 2, which catalyse the formation of the bioactive lipid sphingosine 1-phosphate, are involved in pathophysiological processes. In this chapter, we appraise the evidence that both enzymes are druggable and describe how isoform-specific inhibitors can be developed based on the plasticity of the sphingosine-binding site. This is contextualised with the effect of sphingosine kinase inhibitors in cancer, pulmonary hypertension, neurodegeneration, inflammation and sickling.
Collapse
Affiliation(s)
- Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow, Scotland, UK
| | - David R Adams
- Institute of Chemical Sciences, Heriot-Watt University, Edinburgh, Scotland, UK
| | - Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow, Scotland, UK.
| |
Collapse
|
173
|
Yin X, Willinger CM, Keefe J, Liu J, Fernández-Ortiz A, Ibáñez B, Peñalvo J, Adourian A, Chen G, Corella D, Pamplona R, Portero-Otin M, Jove M, Courchesne P, van Duijn CM, Fuster V, Ordovás JM, Demirkan A, Larson MG, Levy D. Lipidomic profiling identifies signatures of metabolic risk. EBioMedicine 2019; 51:102520. [PMID: 31877415 PMCID: PMC6938899 DOI: 10.1016/j.ebiom.2019.10.046] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/19/2019] [Accepted: 10/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Metabolic syndrome (MetS), the clustering of metabolic risk factors, is associated with cardiovascular disease risk. We sought to determine if dysregulation of the lipidome may contribute to metabolic risk factors. METHODS We measured 154 circulating lipid species in 658 participants from the Framingham Heart Study (FHS) using liquid chromatography-tandem mass spectrometry and tested for associations with obesity, dysglycemia, and dyslipidemia. Independent external validation was sought in three independent cohorts. Follow-up data from the FHS were used to test for lipid metabolites associated with longitudinal changes in metabolic risk factors. RESULTS Thirty-nine lipids were associated with obesity and eight with dysglycemia in the FHS. Of 32 lipids that were available for replication for obesity and six for dyslipidemia, 28 (88%) replicated for obesity and five (83%) for dysglycemia. Four lipids were associated with longitudinal changes in body mass index and four were associated with changes in fasting blood glucose in the FHS. CONCLUSIONS We identified and replicated several novel lipid biomarkers of key metabolic traits. The lipid moieties identified in this study are involved in biological pathways of metabolic risk and can be explored for prognostic and therapeutic utility.
Collapse
Affiliation(s)
- Xiaoyan Yin
- Framingham Heart Study, Framingham, MA, United States; Department of Mathematics and School of Public Health, Boston University, Boston, MA, United States
| | - Christine M Willinger
- Framingham Heart Study, Framingham, MA, United States; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Joshua Keefe
- Framingham Heart Study, Framingham, MA, United States; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jun Liu
- Department of Epidemiology, Erasmus Medical Centre, University Medical Center Rotterdam, Rotterdam, Netherlands; Nuffield Department of Population Health, Oxford University, Oxford, UK
| | - Antonio Fernández-Ortiz
- Tufts University, Friedman School of Nutrition Science and Policy, Boston, MA, United States; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Department of Cardiology, Hospital Clinico San Carlos, Madrid, Spain; CIBERCV, Madrid, Spain
| | - Borja Ibáñez
- Tufts University, Friedman School of Nutrition Science and Policy, Boston, MA, United States; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBERCV, Madrid, Spain; Department of Cardiology, IIS-Fundación Jiménez Díaz, Madrid Spain
| | - José Peñalvo
- Tufts University, Friedman School of Nutrition Science and Policy, Boston, MA, United States
| | | | - George Chen
- Framingham Heart Study, Framingham, MA, United States; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Dolores Corella
- Department of Preventive Medicine and Public Health, Genetic and Molecular Epidemiology Unit, School of Medicine, University of Valencia, Blasco Ibañez, 15, 46010, Valencia, Spain; CIBER Obesity and Nutrition, Madrid, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Manuel Portero-Otin
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Mariona Jove
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Paul Courchesne
- Framingham Heart Study, Framingham, MA, United States; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Centre, University Medical Center Rotterdam, Rotterdam, Netherlands; Nuffield Department of Population Health, Oxford University, Oxford, UK; Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Valentín Fuster
- Tufts University, Friedman School of Nutrition Science and Policy, Boston, MA, United States; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicina at Mount Sinai School, New York, USA
| | - José M Ordovás
- Tufts University, Friedman School of Nutrition Science and Policy, Boston, MA, United States; Jean Mayer USDA-Human Nutrition Research on Aging, Tufts University, Boston, MA, United States
| | - Ayşe Demirkan
- Department of Epidemiology, Erasmus Medical Centre, University Medical Center Rotterdam, Rotterdam, Netherlands; Department of Genetics, University Medical Center Groningen, Groningen, Netherlands
| | - Martin G Larson
- Framingham Heart Study, Framingham, MA, United States; Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, United States; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
174
|
Alcántar-Fernández J, González-Maciel A, Reynoso-Robles R, Pérez Andrade ME, Hernández-Vázquez ADJ, Velázquez-Arellano A, Miranda-Ríos J. High-glucose diets induce mitochondrial dysfunction in Caenorhabditis elegans. PLoS One 2019; 14:e0226652. [PMID: 31846489 PMCID: PMC6917275 DOI: 10.1371/journal.pone.0226652] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/29/2019] [Indexed: 01/20/2023] Open
Abstract
Glucose is an important nutrient that dictates the development, fertility and lifespan of all organisms. In humans, a deficit in its homeostatic control might lead to hyperglucemia and the development of obesity and type 2 diabetes, which show a decreased ability to respond to and metabolize glucose. Previously, we have reported that high-glucose diets (HGD) induce alterations in triglyceride content, body size, progeny, and the mRNA accumulation of key regulators of carbohydrate and lipid metabolism, and longevity in Caenorhabditis elegans (PLoS ONE 13(7): e0199888). Herein, we show that increasing amounts of glucose in the diet induce the swelling of both mitochondria in germ and muscle cells. Additionally, HGD alter the enzymatic activities of the different respiratory complexes in an intricate pattern. Finally, we observed a downregulation of ceramide synthases (hyl-1 and hyl-2) and antioxidant genes (gcs-1 and gst-4), while mitophagy genes (pink-1 and dct-1) were upregulated, probably as part of a mitohormetic mechanism in response to glucose toxicity.
Collapse
Affiliation(s)
- Jonathan Alcántar-Fernández
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
- Unidad de Genética de la Nutrición, Depto. de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, UNAM e Instituto Nacional de Pediatría, Ciudad de México, México
| | - Angélica González-Maciel
- Laboratorio de Morfología Celular y Tisular, Instituto Nacional de Pediatría, Ciudad de México, México
| | - Rafael Reynoso-Robles
- Laboratorio de Morfología Celular y Tisular, Instituto Nacional de Pediatría, Ciudad de México, México
| | - Martha Elva Pérez Andrade
- Unidad de Genética de la Nutrición, Depto. de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, UNAM e Instituto Nacional de Pediatría, Ciudad de México, México
| | - Alain de J. Hernández-Vázquez
- Unidad de Genética de la Nutrición, Depto. de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, UNAM e Instituto Nacional de Pediatría, Ciudad de México, México
| | - Antonio Velázquez-Arellano
- Unidad de Genética de la Nutrición, Depto. de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, UNAM e Instituto Nacional de Pediatría, Ciudad de México, México
| | - Juan Miranda-Ríos
- Unidad de Genética de la Nutrición, Depto. de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, UNAM e Instituto Nacional de Pediatría, Ciudad de México, México
- * E-mail:
| |
Collapse
|
175
|
Dai GY, Yin J, Li KE, Chen DK, Liu Z, Bi FC, Rong C, Yao N. The Arabidopsis AtGCD3 protein is a glucosylceramidase that preferentially hydrolyzes long-acyl-chain glucosylceramides. J Biol Chem 2019; 295:717-728. [PMID: 31819005 DOI: 10.1074/jbc.ra119.011274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 12/01/2019] [Indexed: 11/06/2022] Open
Abstract
Cellular membranes contain many lipids, some of which, such as sphingolipids, have important structural and signaling functions. The common sphingolipid glucosylceramide (GlcCer) is present in plants, fungi, and animals. As a major plant sphingolipid, GlcCer is involved in the formation of lipid microdomains, and the regulation of GlcCer is key for acclimation to stress. Although the GlcCer biosynthetic pathway has been elucidated, little is known about GlcCer catabolism, and a plant GlcCer-degrading enzyme (glucosylceramidase (GCD)) has yet to be identified. Here, we identified AtGCD3, one of four Arabidopsis thaliana homologs of human nonlysosomal glucosylceramidase, as a plant GCD. We found that recombinant AtGCD3 has a low Km for the fluorescent lipid C6-NBD GlcCer and preferentially hydrolyzes long acyl-chain GlcCer purified from Arabidopsis leaves. Testing of inhibitors of mammalian glucosylceramidases revealed that a specific inhibitor of human β-glucosidase 2, N-butyldeoxynojirimycin, inhibits AtGCD3 more effectively than does a specific inhibitor of human β-glucosidase 1, conduritol β-epoxide. We also found that Glu-499 and Asp-647 in AtGCD3 are vital for GCD activity. GFP-AtGCD3 fusion proteins mainly localized to the plasma membrane or the endoplasmic reticulum membrane. No obvious growth defects or changes in sphingolipid contents were observed in gcd3 mutants. Our results indicate that AtGCD3 is a plant glucosylceramidase that participates in GlcCer catabolism by preferentially hydrolyzing long-acyl-chain GlcCers.
Collapse
Affiliation(s)
- Guang-Yi Dai
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Jian Yin
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Kai-En Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Ding-Kang Chen
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhe Liu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Fang-Cheng Bi
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Chan Rong
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Nan Yao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
176
|
Hammad SM, Hardin JR, Wilson DA, Twal WO, Nietert PJ, Oates JC. Race disparity in blood sphingolipidomics associated with lupus cardiovascular comorbidity. PLoS One 2019; 14:e0224496. [PMID: 31747417 PMCID: PMC6867606 DOI: 10.1371/journal.pone.0224496] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/15/2019] [Indexed: 11/18/2022] Open
Abstract
Systemic lupus erythematous (SLE) is a chronic multi-organ autoimmune disease. Genetic and environmental factors contribute to disease onset and severity. Sphingolipids are signaling molecules involved in regulating cell functions and have been associated with multiple genetic disease processes. African-Americans are more likely to suffer from SLE morbidity than Whites. The Medical University of South Carolina has banked plasma samples from a well-characterized lupus cohort that includes African-Americans and Whites. This study examined the influence of race on plasma sphingolipid profiles in SLE patients and association of sphingolipid levels with comorbid atherosclerosis and SLE disease activity. Mass spectrometry revealed that healthy African-Americans had higher sphingomyelin levels and lower lactosylcermide levels compared to healthy Whites. SLE patients, irrespective of race, had higher levels of ceramides, and sphingoid bases (sphingosine and dihydrosphingosine) and their phosphates compared to healthy subjects. Compared to African-American controls, African-American SLE patients had higher levels of ceramides, hexosylceramides, sphingosine and dihydrosphingosine 1-phosphate. Compared to White controls, White SLE patients exhibited higher levels of sphingoid bases and their phosphates, but lower ratios of C16:0 ceramide/sphingosine 1-phosphate and C24:1 ceramide/sphingosine 1-phosphate. White SLE patients with atherosclerosis exhibited lower levels of sphingoid bases compared to White SLE patients without atherosclerosis. In contrast, African-American SLE patients with atherosclerosis had higher levels of sphingoid bases and sphingomyelins compared to African-American SLE patients without atherosclerosis. Compared to White SLE patients with atherosclerosis, African-American SLE patients with atherosclerosis had higher levels of select sphingolipids. Plasma levels of sphingosine, C16:0 ceramide/sphingosine 1-phosphate ratio and C24:1 ceramide/sphingosine 1-phosphate ratio significantly correlated with SLEDAI in the African-American but not White SLE patients. The C16:0 ceramide/sphingosine 1-phosphate ratio in SLE patients, and levels of C18:1 and C26:1 lactosylcermides, C20:0 hexosylceramide, and sphingoid bases in SLE patients with atherosclerosis could be dependent on race. Further ethnic studies in SLE cohorts are necessary to verify use of sphingolipidomics as complementary diagnostic tool.
Collapse
Affiliation(s)
- Samar M. Hammad
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jasmyn R. Hardin
- College of Graduate Studies/Summer Undergraduate Research Program, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Dulaney A. Wilson
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Waleed O. Twal
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Paul J. Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - James C. Oates
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|
177
|
Arshad H, Alfonso JCL, Franke R, Michaelis K, Araujo L, Habib A, Zboromyrska Y, Lücke E, Strungaru E, Akmatov MK, Hatzikirou H, Meyer-Hermann M, Petersmann A, Nauck M, Brönstrup M, Bilitewski U, Abel L, Sievers J, Vila J, Illig T, Schreiber J, Pessler F. Decreased plasma phospholipid concentrations and increased acid sphingomyelinase activity are accurate biomarkers for community-acquired pneumonia. J Transl Med 2019; 17:365. [PMID: 31711507 PMCID: PMC6849224 DOI: 10.1186/s12967-019-2112-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background There continues to be a great need for better biomarkers and host-directed treatment targets for community-acquired pneumonia (CAP). Alterations in phospholipid metabolism may constitute a source of small molecule biomarkers for acute infections including CAP. Evidence from animal models of pulmonary infections and sepsis suggests that inhibiting acid sphingomyelinase (which releases ceramides from sphingomyelins) may reduce end-organ damage. Methods We measured concentrations of 105 phospholipids, 40 acylcarnitines, and 4 ceramides, as well as acid sphingomyelinase activity, in plasma from patients with CAP (n = 29, sampled on admission and 4 subsequent time points), chronic obstructive pulmonary disease exacerbation with infection (COPD, n = 13) as a clinically important disease control, and 33 age- and sex-matched controls. Results Phospholipid concentrations were greatly decreased in CAP and normalized along clinical improvement. Greatest changes were seen in phosphatidylcholines, followed by lysophosphatidylcholines, sphingomyelins and ceramides (three of which were upregulated), and were least in acylcarnitines. Changes in COPD were less pronounced, but also differed qualitatively, e.g. by increases in selected sphingomyelins. We identified highly accurate biomarkers for CAP (AUC ≤ 0.97) and COPD (AUC ≤ 0.93) vs. Controls, and moderately accurate biomarkers for CAP vs. COPD (AUC ≤ 0.83), all of which were phospholipids. Phosphatidylcholines, lysophosphatidylcholines, and sphingomyelins were also markedly decreased in S. aureus-infected human A549 and differentiated THP1 cells. Correlations with C-reactive protein and procalcitonin were predominantly negative but only of mild-to-moderate extent, suggesting that these markers reflect more than merely inflammation. Consistent with the increased ceramide concentrations, increased acid sphingomyelinase activity accurately distinguished CAP (fold change = 2.8, AUC = 0.94) and COPD (1.75, 0.88) from Controls and normalized with clinical resolution. Conclusions The results underscore the high potential of plasma phospholipids as biomarkers for CAP, begin to reveal differences in lipid dysregulation between CAP and infection-associated COPD exacerbation, and suggest that the decreases in plasma concentrations are at least partially determined by changes in host target cells. Furthermore, they provide validation in clinical blood samples of acid sphingomyelinase as a potential treatment target to improve clinical outcome of CAP.
Collapse
Affiliation(s)
- Haroon Arshad
- Research Group "Biomarkers for Infectious Diseases", TWINCORE Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Juan Carlos López Alfonso
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Raimo Franke
- Department of Chemical Biology, Helmholtz Centre for Infection Research and German Center for Infection Research (DZIF), Brunswick, Germany
| | - Katina Michaelis
- Clinic for Pneumology, Otto-von-Guericke University, Magdeburg, Germany
| | - Leonardo Araujo
- Research Group "Biomarkers for Infectious Diseases", TWINCORE Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Aamna Habib
- Research Group "Biomarkers for Infectious Diseases", TWINCORE Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Department of Chemical Biology, Helmholtz Centre for Infection Research and German Center for Infection Research (DZIF), Brunswick, Germany
| | - Yuliya Zboromyrska
- Department of Clinical Microbiology, Biomedical Diagnostic Centre (CDB), Hospital Clinic, School of Medicine, University of Barcelona, Institute of Global Health (ISGlobal), Barcelona, Spain
| | - Eva Lücke
- Clinic for Pneumology, Otto-von-Guericke University, Magdeburg, Germany
| | - Emilia Strungaru
- Clinic for Pneumology, Otto-von-Guericke University, Magdeburg, Germany
| | - Manas K Akmatov
- Research Group "Biomarkers for Infectious Diseases", TWINCORE Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany.,Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Haralampos Hatzikirou
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Astrid Petersmann
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany.,UMG-Laboratory, University Medicine Göttingen, Göttingen, Germany
| | - Matthias Nauck
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research and German Center for Infection Research (DZIF), Brunswick, Germany
| | - Ursula Bilitewski
- Department of Chemical Biology, Helmholtz Centre for Infection Research and German Center for Infection Research (DZIF), Brunswick, Germany
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France.,Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, USA
| | - Jorg Sievers
- Clinical Microbiology, GlaxoSmithKline, Collegeville, PA, USA.,Clinical Development, ViiV Healthcare, Brentford, UK
| | - Jordi Vila
- Department of Clinical Microbiology, Biomedical Diagnostic Centre (CDB), Hospital Clinic, School of Medicine, University of Barcelona, Institute of Global Health (ISGlobal), Barcelona, Spain
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Jens Schreiber
- Clinic for Pneumology, Otto-von-Guericke University, Magdeburg, Germany
| | - Frank Pessler
- Research Group "Biomarkers for Infectious Diseases", TWINCORE Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str. 7, 30625, Hannover, Germany. .,Helmholtz Centre for Infection Research, Brunswick, Germany. .,Centre for Individualised Infection Medicine, Hannover, Germany.
| |
Collapse
|
178
|
Garić D, De Sanctis JB, Shah J, Dumut DC, Radzioch D. Biochemistry of very-long-chain and long-chain ceramides in cystic fibrosis and other diseases: The importance of side chain. Prog Lipid Res 2019:100998. [PMID: 31445070 DOI: 10.1016/j.plipres.2019.100998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/01/2019] [Accepted: 03/10/2019] [Indexed: 12/18/2022]
Abstract
Ceramides, the principal building blocks of all sphingolipids, have attracted the attention of many scientists around the world interested in developing treatments for cystic fibrosis, the most common genetic disease of Caucasians. Many years of fruitful research in this field have produced some fundamentally important, yet controversial results. Here, we aimed to summarize the current knowledge on the role of long- and very-long- chain ceramides, the most abundant species of ceramides in animal cells, in cystic fibrosis and other diseases. We also aim to explain the importance of the length of their side chain in the context of stability of transmembrane proteins through a concise synthesis of their biophysical chemistry, cell biology, and physiology. This review also addresses several remaining riddles in this field. Finally, we discuss the technical challenges associated with the analysis and quantification of ceramides. We provide the evaluation of the antibodies used for ceramide quantification and we demonstrate their lack of specificity. Results and discussion presented here will be of interest to anyone studying these enigmatic lipids.
Collapse
Affiliation(s)
- Dušan Garić
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Juan B De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Juhi Shah
- Department of Pharmacology and Experimental Therapeutics, McGill University, Montreal, QC, Canada
| | - Daciana Catalina Dumut
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Danuta Radzioch
- Department of Human Genetics, McGill University, Montreal, QC, Canada; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic; Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
179
|
Chao HC, Lee TH, Chiang CS, Yang SY, Kuo CH, Tang SC. Sphingolipidomics Investigation of the Temporal Dynamics after Ischemic Brain Injury. J Proteome Res 2019; 18:3470-3478. [PMID: 31310127 DOI: 10.1021/acs.jproteome.9b00370] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Sphingolipids (SPLs) have been proposed as potential therapeutic targets for strokes, but no reports have ever profiled the changes of the entire range of SPLs after a stroke. This study applied sphingolipidomic methods to investigate the temporal and individual changes in the sphingolipidome including the effect of atorvastatin after ischemic brain injury. We conducted sphingolipidomic profiling of mouse brain tissue by liquid chromatography-electrospray ionization tandem mass spectrometry at 3 h and 24 h after 1 h of middle cerebral artery occlusion (MCAO), and SPL levels were compared with those of the Sham control group. At 3 h post-MCAO, ceramides (Cers) exhibited an increase in levels of long-chain Cers but a decrease in very-long-chain Cers. Moreover, sphingosine, the precursor of sphingosine-1-phosphate (S1P), decreased and S1P increased at 3 h after MCAO. In contrast to 3 h, both long-chain and very-long-chain Cers showed an increased trend at 24 h post-MCAO. Most important, the administration of atorvastatin improved the neurological function of the mice and significantly reversed the SPL changes resulting from the ischemic injury. Furthermore, we used plasma samples from nonstroke control and stroke patients at time points of 72 h after a stroke, and found a similar trend of Cers as in the MCAO model. This study successfully elucidated the overall effect of ischemic injury on SPL metabolism with and without atorvastatin treatment. The network of SPL components that change upon ischemic damage may provide novel therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Hsi-Chun Chao
- School of Pharmacy, College of Medicine , National Taiwan University , Taipei 100 , Taiwan.,The Metabolomics Core Laboratory, Center of Genomic Medicine , National Taiwan University , Taipei 100 , Taiwan
| | - Tsung-Heng Lee
- School of Pharmacy, College of Medicine , National Taiwan University , Taipei 100 , Taiwan.,The Metabolomics Core Laboratory, Center of Genomic Medicine , National Taiwan University , Taipei 100 , Taiwan
| | - Chien-Sung Chiang
- Stroke Center and Department of Neurology , National Taiwan University Hospital , Taipei 100 , Taiwan
| | - Sin-Yu Yang
- Stroke Center and Department of Neurology , National Taiwan University Hospital , Taipei 100 , Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine , National Taiwan University , Taipei 100 , Taiwan.,The Metabolomics Core Laboratory, Center of Genomic Medicine , National Taiwan University , Taipei 100 , Taiwan.,Department of Pharmacy , National Taiwan University Hospital , Taipei 100 , Taiwan
| | - Sung-Chun Tang
- Stroke Center and Department of Neurology , National Taiwan University Hospital , Taipei 100 , Taiwan
| |
Collapse
|
180
|
Lai M, La Rocca V, Amato R, Freer G, Pistello M. Sphingolipid/Ceramide Pathways and Autophagy in the Onset and Progression of Melanoma: Novel Therapeutic Targets and Opportunities. Int J Mol Sci 2019; 20:ijms20143436. [PMID: 31336922 PMCID: PMC6678284 DOI: 10.3390/ijms20143436] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Melanoma is a malignant tumor deriving from neoplastic transformation of melanocytes. The incidence of melanoma has increased dramatically over the last 50 years. It accounts for most cases of skin cancer deaths. Early diagnosis leads to remission in 90% of cases of melanoma; conversely, for melanoma at more advanced stages, prognosis becomes more unfavorable also because dvanced melanoma is often resistant to pharmacological and radiological therapies due to genetic plasticity, presence of cancer stem cells that regenerate the tumor, and efficient elimination of drugs. This review illustrates the role of autophagy in tumor progression and resistance to therapy, focusing on molecular targets for future drugs.
Collapse
Affiliation(s)
- Michele Lai
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Veronica La Rocca
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Rachele Amato
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Giulia Freer
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy
| | - Mauro Pistello
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy.
- Virology Unit, Pisa University Hospital, 56127 Pisa, Italy.
| |
Collapse
|
181
|
Sakamoto W, Canals D, Salamone S, Allopenna J, Clarke CJ, Snider J, Obeid LM, Hannun YA. Probing compartment-specific sphingolipids with targeted bacterial sphingomyelinases and ceramidases. J Lipid Res 2019; 60:1841-1850. [PMID: 31243119 DOI: 10.1194/jlr.m094722] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/11/2019] [Indexed: 12/20/2022] Open
Abstract
Sphingolipids contribute to the regulation of cell and tissue homeostasis, and disorders of sphingolipid metabolism lead to diseases such as inflammation, stroke, diabetes, and cancer. Sphingolipid metabolic pathways involve an array of enzymes that reside in specific subcellular organelles, resulting in the formation of many diverse sphingolipids with distinct molecular species based on the diversity of the ceramide (Cer) structure. In order to probe compartment-specific metabolism of sphingolipids in this study, we analyzed the Cer and SM species preferentially produced in the inner plasma membrane (PM), Golgi apparatus, ER, mitochondria, nucleus, and cytoplasm by using compartmentally targeted bacterial SMases and ceramidases. The results showed that the length of the acyl chain of Cer becomes longer according to the progress of Cer from synthesis in the ER to the Golgi apparatus, then to the PM. These findings suggest that each organelle shows different properties of SM-derived Cers consistent with its emerging distinct functions in vitro and in vivo.
Collapse
Affiliation(s)
- Wataru Sakamoto
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY.,Ono Pharmaceutical Company, Ltd. Oncology Research Laboratories, Osaka, Japan
| | - Daniel Canals
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY
| | - Silvia Salamone
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY
| | - Janet Allopenna
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY
| | - Christopher J Clarke
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY
| | - Justin Snider
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY
| | - Lina M Obeid
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY.,Northport Veterans Affairs Medical Center, Northport, NY
| | - Yusuf A Hannun
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY .,Departments of Biochemistry, Pharmacology, and Pathology, Stony Brook University, Stony Brook, NY
| |
Collapse
|
182
|
Simón MV, Prado Spalm FH, Vera MS, Rotstein NP. Sphingolipids as Emerging Mediators in Retina Degeneration. Front Cell Neurosci 2019; 13:246. [PMID: 31244608 PMCID: PMC6581011 DOI: 10.3389/fncel.2019.00246] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022] Open
Abstract
The sphingolipids ceramide (Cer), sphingosine-1-phosphate (S1P), sphingosine (Sph), and ceramide-1-phosphate (C1P) are key signaling molecules that regulate major cellular functions. Their roles in the retina have gained increasing attention during the last decade since they emerge as mediators of proliferation, survival, migration, neovascularization, inflammation and death in retina cells. As exacerbation of these processes is central to retina degenerative diseases, they appear as crucial players in their progression. This review analyzes the functions of these sphingolipids in retina cell types and their possible pathological roles. Cer appears as a key arbitrator in diverse retinal pathologies; it promotes inflammation in endothelial and retina pigment epithelium (RPE) cells and its increase is a common feature in photoreceptor death in vitro and in animal models of retina degeneration; noteworthy, inhibiting Cer synthesis preserves photoreceptor viability and functionality. In turn, S1P acts as a double edge sword in the retina. It is essential for retina development, promoting the survival of photoreceptors and ganglion cells and regulating proliferation and differentiation of photoreceptor progenitors. However, S1P has also deleterious effects, stimulating migration of Müller glial cells, angiogenesis and fibrosis, contributing to the inflammatory scenario of proliferative retinopathies and age related macular degeneration (AMD). C1P, as S1P, promotes photoreceptor survival and differentiation. Collectively, the expanding role for these sphingolipids in the regulation of critical processes in retina cell types and in their dysregulation in retina degenerations makes them attractive targets for treating these diseases.
Collapse
Affiliation(s)
- M Victoria Simón
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| | - Facundo H Prado Spalm
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| | - Marcela S Vera
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| | - Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| |
Collapse
|
183
|
Bennett MK, Wallington-Beddoe CT, Pitson SM. Sphingolipids and the unfolded protein response. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1483-1494. [PMID: 31176037 DOI: 10.1016/j.bbalip.2019.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/29/2019] [Accepted: 06/01/2019] [Indexed: 12/17/2022]
Abstract
The unfolded protein response (UPR) is a response by the endoplasmic reticulum to stress, classically caused by any disruption to cell homeostasis that results in an accumulation in unfolded proteins. However, there is an increasing body of research demonstrating that the UPR can also be activated by changes in lipid homeostasis, including changes in sphingolipid metabolism. Sphingolipids are a family of bioactive lipids with important roles in both the formation and integrity of cellular membranes, and regulation of key cellular processes, including cell proliferation and apoptosis. Bi-directional interactions between sphingolipids and the UPR have now been observed in a range of diseases, including cancer, diabetes and liver disease. Determining how these two key cellular components influence each other could play an important role in deciphering the causes of these diseases and potentially reveal new therapeutic approaches.
Collapse
Affiliation(s)
- Melissa K Bennett
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5001, Australia
| | - Craig T Wallington-Beddoe
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5001, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia; Flinders Medical Centre, Bedford Park, SA 5042, Australia; College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5001, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
184
|
Inherited monogenic defects of ceramide metabolism: Molecular bases and diagnoses. Clin Chim Acta 2019; 495:457-466. [PMID: 31128082 DOI: 10.1016/j.cca.2019.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023]
Abstract
Ceramides are membrane lipids implicated in the regulation of numerous biological functions. Recent evidence suggests that specific subsets of molecular species of ceramide may play distinct physiological roles. The importance of this family of molecules in vertebrates is witnessed by the deleterious consequences of genetic alterations in ceramide metabolism. This brief review summarizes the clinical presentation of human disorders due to the deficiency of enzymes involved either in the biosynthesis or the degradation of ceramides. Information on the possible underlying pathophysiological mechanisms is also provided, based on knowledge gathered from animal models of these inherited rare conditions. When appropriate, tools for chemical and molecular diagnosis of these disorders and therapeutic options are also presented.
Collapse
|
185
|
Castro K, Ntranos A, Amatruda M, Petracca M, Kosa P, Chen EY, Morstein J, Trauner D, Watson CT, Kiebish MA, Bielekova B, Inglese M, Katz Sand I, Casaccia P. Body Mass Index in Multiple Sclerosis modulates ceramide-induced DNA methylation and disease course. EBioMedicine 2019; 43:392-410. [PMID: 30981648 PMCID: PMC6557766 DOI: 10.1016/j.ebiom.2019.03.087] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/24/2019] [Accepted: 03/29/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Multiple Sclerosis (MS) results from genetic predisposition and environmental variables, including elevated Body Mass Index (BMI) in early life. This study addresses the effect of BMI on the epigenome of monocytes and disease course in MS. METHODS Fifty-four therapy-naive Relapsing Remitting (RR) MS patients with high and normal BMI received clinical and MRI evaluation. Blood samples were immunophenotyped, and processed for unbiased plasma lipidomic profiling and genome-wide DNA methylation analysis of circulating monocytes. The main findings at baseline were validated in an independent cohort of 91 therapy-naïve RRMS patients. Disease course was evaluated by a two-year longitudinal follow up and mechanistic hypotheses tested in human cell cultures and in animal models of MS. FINDINGS Higher monocytic counts and plasma ceramides, and hypermethylation of genes involved in negative regulation of cell proliferation were detected in the high BMI group of MS patients compared to normal BMI. Ceramide treatment of monocytic cell cultures increased proliferation in a dose-dependent manner and was prevented by DNA methylation inhibitors. The high BMI group of MS patients showed a negative correlation between monocytic counts and brain volume. Those subjects at a two-year follow-up showed increased T1 lesion load, increased disease activity, and worsened clinical disability. Lastly, the relationship between body weight, monocytic infiltration, DNA methylation and disease course was validated in mouse models of MS. INTERPRETATION High BMI negatively impacts disease course in Multiple Sclerosis by modulating monocyte cell number through ceramide-induced DNA methylation of anti-proliferative genes. FUND: This work was supported by funds from the Friedman Brain Institute, NIH, and Multiple Sclerosis Society.
Collapse
Affiliation(s)
- Kamilah Castro
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, NY, New York, United States of America
| | - Achilles Ntranos
- Department of Neurology, Icahn School of Medicine at Mount Sinai, NY, New York, United States of America
| | - Mario Amatruda
- Advanced Science Research Center at The Graduate Center of The City University of New York and Inter-Institutional Center for Glial Biology at Icahn School of Medicine New York, New York, United States of America
| | - Maria Petracca
- Department of Neurology, Icahn School of Medicine at Mount Sinai, NY, New York, United States of America
| | - Peter Kosa
- Neuroimmunological Disease Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Emily Y Chen
- BERG, LLC. Framingham, MA, United States of America
| | - Johannes Morstein
- Department of Chemistry, New York University, NY, New York, United States of America
| | - Dirk Trauner
- Department of Chemistry, New York University, NY, New York, United States of America
| | - Corey T Watson
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, United States of America
| | | | - Bibiana Bielekova
- Neuroimmunological Disease Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Matilde Inglese
- Department of Neurology, Icahn School of Medicine at Mount Sinai, NY, New York, United States of America
| | - Ilana Katz Sand
- Department of Neurology, Icahn School of Medicine at Mount Sinai, NY, New York, United States of America
| | - Patrizia Casaccia
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, NY, New York, United States of America; Advanced Science Research Center at The Graduate Center of The City University of New York and Inter-Institutional Center for Glial Biology at Icahn School of Medicine New York, New York, United States of America.
| |
Collapse
|
186
|
Zhou DR, Eid R, Miller KA, Boucher E, Mandato CA, Greenwood MT. Intracellular second messengers mediate stress inducible hormesis and Programmed Cell Death: A review. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:773-792. [PMID: 30716408 DOI: 10.1016/j.bbamcr.2019.01.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022]
|
187
|
Differential lipid composition and regulation along the hippocampal longitudinal axis. Transl Psychiatry 2019; 9:144. [PMID: 31028243 PMCID: PMC6486574 DOI: 10.1038/s41398-019-0478-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/02/2019] [Indexed: 01/16/2023] Open
Abstract
Lipids are major constituents of the brain largely implicated in physiological and pathological processes. The hippocampus is a complex brain structure involved in learning, memory and emotional responses, and its functioning is also affected in various disorders. Despite conserved intrinsic circuitry, behavioral and anatomical studies suggest the existence of a structural and functional gradient along the hippocampal longitudinal axis. Here, we used an unbiased mass spectrometry approach to characterize the lipid composition of distinct hippocampal subregions. In addition, we evaluated the susceptibility of each area to lipid modulation by corticosterone (CORT), an important mediator of the effects of stress. We confirmed a great similarity between hippocampal subregions relatively to other brain areas. Moreover, we observed a continuous molecular gradient along the longitudinal axis of the hippocampus, with the dorsal and ventral extremities differing significantly from each other, particularly in the relative abundance of sphingolipids and phospholipids. Also, whereas chronic CORT exposure led to remodeling of triacylglycerol and phosphatidylinositol species in both hippocampal poles, our study suggests that the ventral hippocampus is more sensitive to CORT-induced changes, with regional modulation of ceramide, dihydrosphingomyelin and phosphatidic acid. Thus, our results confirm a multipartite molecular view of dorsal-ventral hippocampal axis and emphasize lipid metabolites as candidate effectors of glucocorticoid signaling, mediating regional susceptibility to neurological disorders associated with stress.
Collapse
|
188
|
Möuts A, Yamamoto T, Nyholm TKM, Murata M, Slotte JP. Nonlamellar-Phase-Promoting Colipids Enhance Segregation of Palmitoyl Ceramide in Fluid Bilayers. Biophys J 2019; 116:1507-1515. [PMID: 30940348 DOI: 10.1016/j.bpj.2019.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 11/15/2022] Open
Abstract
Ceramide is an important intermediate in sphingolipid homeostasis. We examined how colipids, with negative intrinsic curvature and which may induce curvature stress in the bilayers, affected the segregation of palmitoyl ceramide (PCer). Such colipids include 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and tetra-linoleoyl cardiolipin (CL). In 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) bilayers, PCer formed ordered, gel-like domains at concentrations above 10 mol% at 23°C, as evidenced by the change in the average lifetime of the trans-parinaric acid emission. When POPE or DOPE were included in the DOPC bilayer (at 20:80 or 40:60 POPE or DOPE to DOPC, by mol), the lateral segregation of PCer was facilitated in a concentration-dependent manner, and less PCer was required for the formation of the ordered ceramide-rich domains. Inclusion of CL in the DOPE bilayer (at 10:90 or 20:80 CL to PC, by mol) also caused a similar facilitation of the lateral segregation of PCer. The PCer-rich domains formed in the presence of POPE, DOPE, or CL in DOPC bilayers were slightly more thermostable (by 2-10°C) when compared to PCer-rich domains in DOPC-only bilayers. Nonlamellar phases were not present in bilayers in which the effects of POPE or DOPE on PCer segregation were the largest, as verified by 31P NMR. When palmitoyl sphingomyelin was added to the different bilayer compositions at 5 mol%, relative to the phospholipids, PCer segregated into gel domains at lower concentrations (2-3 mol% PCer), and the effect of POPE on PCer segregation was eliminated. We suggest that the effects of POPE, DOPE, and CL on PCer segregation was in part influenced by their effects on membrane curvature stress and in part because of unfavorable interactions with PCer due to their unsaturated acyl chains. These lipids are abundant in mitochondrial membranes and are likely to affect functional properties of saturated ceramides in them.
Collapse
Affiliation(s)
- Anna Möuts
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Tomoya Yamamoto
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan
| | - Thomas K M Nyholm
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Michio Murata
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan; Japan Science and Technology Agency, ERATO, Lipid Active Structure Project, Toyonaka, Osaka, Japan
| | - J Peter Slotte
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
| |
Collapse
|
189
|
Moro K, Nagahashi M, Gabriel E, Takabe K, Wakai T. Clinical application of ceramide in cancer treatment. Breast Cancer 2019; 26:407-415. [PMID: 30963461 DOI: 10.1007/s12282-019-00953-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/04/2019] [Indexed: 12/15/2022]
Abstract
Development of innovative strategies for cancer treatment is a pressing public health issue. Despite recent advances, the mechanisms of cancer progression and the resistance to cancer treatment have not been fully elucidated. Sphingolipids, including ceramide and sphingoshin-1-phosphate, are bioactive mediators that regulate cancer cell death and survival through the dynamic balance of what has been termed the 'sphingolipid rheostat'. Specifically, ceramide, which acts as the central hub of sphingolipid metabolism, is generated via three major pathways by many stressors, including anti-cancer treatments, environmental stresses, and cytokines. We have previously shown in breast cancer patients that elevated ceramide correlated with less aggressive cancer phenotypes, leading to a prognostic impact. Recent studies showed that ceramide have the possibility of becoming the reinforcing agent of cancer treatment as well as other roles such as nanoparticles and diagnostic biomarker. We review ceramide as one of the key molecules to investigate in overcoming resistance to current drug therapies and in becoming one of the newest cancer treatments.
Collapse
Affiliation(s)
- Kazuki Moro
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan.
| | | | - Kazuaki Takabe
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan.,Division of Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, the State University of New York, Buffalo, NY, USA
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| |
Collapse
|
190
|
Shi XX, Zhang H, Chen M, Zhang YD, Zhu MF, Zhang MJ, Li FQ, Wratten S, Zhou WW, Mao C, Zhu ZR. Two sphingomyelin synthase homologues regulate body weight and sphingomyelin synthesis in female brown planthopper, N. lugens (Stål). INSECT MOLECULAR BIOLOGY 2019; 28:253-263. [PMID: 30375099 DOI: 10.1111/imb.12549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Although sphingomyelins known to be are lipid constituents of the plasma membrane in vertebrates, much remains obscure about the metabolism of sphingomyelins in insects. With ultra performance liquid chromatography-time-of-flight-tandem mass spectrometry analysis, we revealed for the first time that sphingomyelins are abundant in Nilaparvata lugens (Stål), the brown planthopper (BPH), and their biosynthesis is carried out by sphingomyelin synthase-like protein 2 (SMSL2), which is homologous to sphingomyelin synthase-related protein (SMSr). Unlike other insect species, high concentrations of sphingomyelins rather than ceramide phosphoethanolamines exist in the BPH. Two putative genes, which are homologous to SMSr, are named Nilaparvata lugens SMS-like 1 (NlSMSL1) and 2 (NlSMSL2). Knockdowns of both NlSMSL2 and NlSMSL1 were conducted but only the first decreased concentrations of sphingomyelins in the BPH, indicating that NlSMSL2 plays a role in the biosynthesis of sphingomyelins. Real-time quantitative PCR analysis revealed both NlSMSL1 and NlSMSL2 are highly expressed in BPH adults, with NlSMSL1 specifically highly expressed in reproductive organs (ovaries and testes) whereas NlSMSL2 was highly expressed in the malpighian tubules. The knockdown of NlSMSL1 or NlSMSL2 increased BPH female body weight but not that of males, suggesting sex-specific roles for SMSLs in influencing BPH body weight. The results suggest that NlSMSL2 catalyses the synthesis of sphingomyelins and maintains female BPH body weight through alteration of sphingolipid content.
Collapse
Affiliation(s)
- X-X Shi
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
- Institute of Insect Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - H Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M Chen
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - Y-D Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M-F Zhu
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M-J Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - F-Q Li
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - S Wratten
- Bio-Protection Research Centre, Lincoln University, Lincoln, New Zealand
| | - W-W Zhou
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - C Mao
- Department of Medicine and Stony Brook Cancer Center, The State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Z-R Zhu
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| |
Collapse
|
191
|
Casasampere M, Bielsa N, Riba D, Bassas L, Xu R, Mao C, Fabriàs G, Abad JL, Delgado A, Casas J. New fluorogenic probes for neutral and alkaline ceramidases. J Lipid Res 2019; 60:1174-1181. [PMID: 30926626 DOI: 10.1194/jlr.d092759] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
New fluorogenic ceramidase substrates derived from the N-acyl modification of our previously reported probes (RBM14) are reported. While none of the new probes were superior to the known RBM14C12 as acid ceramidase substrates, the corresponding nervonic acid amide (RBM14C24:1) is an efficient and selective substrate for the recombinant human neutral ceramidase, both in cell lysates and in intact cells. A second generation of substrates, incorporating the natural 2-(N-acylamino)-1,3-diol-4-ene framework (compounds RBM15) is also reported. Among them, the corresponding fatty acyl amides with an unsaturated N-acyl chain can be used as substrates to determine alkaline ceramidase (ACER)1 and ACER2 activities. In particular, compound RBM15C18:1 has emerged as the best fluorogenic probe reported so far to measure ACER1 and ACER2 activities in a 96-well plate format.
Collapse
Affiliation(s)
- Mireia Casasampere
- Spanish National Research Council (CSIC), Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, Research Unit on Bioactive Molecules (RUBAM), 08034 Barcelona, Spain.,Faculty of Pharmacy and Food Sciences Department of Pharmacology, Toxicology, and Medicinal Chemistry, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), University of Barcelona, 08028 Barcelona, Spain
| | - Núria Bielsa
- Spanish National Research Council (CSIC), Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, Research Unit on Bioactive Molecules (RUBAM), 08034 Barcelona, Spain.,Faculty of Pharmacy and Food Sciences Department of Pharmacology, Toxicology, and Medicinal Chemistry, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), University of Barcelona, 08028 Barcelona, Spain
| | - Daniel Riba
- Spanish National Research Council (CSIC), Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, Research Unit on Bioactive Molecules (RUBAM), 08034 Barcelona, Spain
| | - Laura Bassas
- Spanish National Research Council (CSIC), Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, Research Unit on Bioactive Molecules (RUBAM), 08034 Barcelona, Spain
| | - Ruijuan Xu
- Department of Medicine State University of New York at Stony Brook, Stony Brook, NY 11794-8155
| | - Cungui Mao
- Department of Medicine State University of New York at Stony Brook, Stony Brook, NY 11794-8155
| | - Gemma Fabriàs
- Spanish National Research Council (CSIC), Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, Research Unit on Bioactive Molecules (RUBAM), 08034 Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBEREHD), 28029 Madrid, Spain
| | - José-Luis Abad
- Spanish National Research Council (CSIC), Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, Research Unit on Bioactive Molecules (RUBAM), 08034 Barcelona, Spain
| | - Antonio Delgado
- Spanish National Research Council (CSIC), Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, Research Unit on Bioactive Molecules (RUBAM), 08034 Barcelona, Spain .,Faculty of Pharmacy and Food Sciences Department of Pharmacology, Toxicology, and Medicinal Chemistry, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), University of Barcelona, 08028 Barcelona, Spain
| | - Josefina Casas
- Spanish National Research Council (CSIC), Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, Research Unit on Bioactive Molecules (RUBAM), 08034 Barcelona, Spain .,Centro de Investigación Biomédica en Red (CIBEREHD), 28029 Madrid, Spain
| |
Collapse
|
192
|
Hu SJ, Jiang SS, Zhang J, Luo D, Yu B, Yang LY, Zhong HH, Yang MW, Liu LY, Hong FF, Yang SL. Effects of apoptosis on liver aging. World J Clin Cases 2019; 7:691-704. [PMID: 30968034 PMCID: PMC6448073 DOI: 10.12998/wjcc.v7.i6.691] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/10/2019] [Accepted: 01/26/2019] [Indexed: 02/05/2023] Open
Abstract
As an irreversible and perennial process, aging is accompanied by functional and morphological declines in organs. Generally, aging liver exhibits a decline in volume and hepatic blood flow. Even with a preeminent regenerative capacity to restore its functions after liver cell loss, its biosynthesis and metabolism abilities decline, and these are difficult to restore to previous standards. Apoptosis is a programmed death process via intrinsic and extrinsic pathways, in which Bcl-2 family proteins and apoptosis-related genes, such as p21 and p53, are involved. Apoptosis inflicts both favorable and adverse influences on liver aging. Apoptosis eliminates transformed abnormal cells but promotes age-related liver diseases, such as nonalcoholic fatty liver disease, liver fibrosis, cirrhosis, and liver cancer. We summarize the roles of apoptosis in liver aging and age-related liver diseases.
Collapse
Affiliation(s)
- Shao-Jie Hu
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Sha-Sha Jiang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Jin Zhang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Dan Luo
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Bo Yu
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Liang-Yan Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Hua-Hua Zhong
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Mei-Wen Yang
- Department of Nurse, Nanchang University Hospital, Nanchang 330006, Jiangxi Province, China
| | - Li-Yu Liu
- Department of Nurse, Nanchang University Hospital, Nanchang 330006, Jiangxi Province, China
| | - Fen-Fang Hong
- Experimental Teaching Center, Nanchang University, Nanchang 330031, Jiangxi Province, China
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
193
|
Preuss C, Jelenik T, Bódis K, Müssig K, Burkart V, Szendroedi J, Roden M, Markgraf DF. A New Targeted Lipidomics Approach Reveals Lipid Droplets in Liver, Muscle and Heart as a Repository for Diacylglycerol and Ceramide Species in Non-Alcoholic Fatty Liver. Cells 2019; 8:cells8030277. [PMID: 30909521 PMCID: PMC6468791 DOI: 10.3390/cells8030277] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is frequently associated with excessive accumulation of lipids in ectopic tissue and presents a major risk factor for type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD). Diacylglycerols (DAGs) and ceramides (CERs) were identified as key players in lipid-induced insulin resistance, typical for such diseases. Recent results suggest that the subcellular distribution of these lipids affects their lipotoxic properties. However, the subcellular dynamics of these lipids and the role of lipid droplets (LDs) as a potential storage site is not understood. Here, we developed a liquid chromatography triple quadrupole mass spectrometry (LC-MS/MS)-method for the rapid and simultaneous quantification of DAG and CER species in tissue sample fractions. The assay is characterized by excellent recovery of analytes, limit of quantification, accuracy and precision. We established a fractionation protocol that allows the separation of subcellular tissue fractions. This method was subsequently tested to measure the concentration of DAGs and CERs in subcellular fractions of human muscle and several mouse tissues. In a mouse model of NAFLD, application of this method revealed a prominent role for LDs as repository for lipotoxic DAG and CER species. In conclusion, the new method proved as a valuable tool to analyse the subcellular dynamics of lipotoxins, related to the pathogenesis of insulin resistance, T2D and NAFLD.
Collapse
Affiliation(s)
- Christina Preuss
- Institute for Clinical Diabetology, German Diabetes Center, c/o Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), München, D-85764 Neuherberg, Germany.
| | - Tomas Jelenik
- Institute for Clinical Diabetology, German Diabetes Center, c/o Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), München, D-85764 Neuherberg, Germany.
| | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, c/o Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), München, D-85764 Neuherberg, Germany.
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center, c/o Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), München, D-85764 Neuherberg, Germany.
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, D-40225, Düsseldorf, Germany.
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, c/o Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), München, D-85764 Neuherberg, Germany.
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, c/o Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), München, D-85764 Neuherberg, Germany.
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, D-40225, Düsseldorf, Germany.
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, c/o Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), München, D-85764 Neuherberg, Germany.
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, D-40225, Düsseldorf, Germany.
| | - Daniel F Markgraf
- Institute for Clinical Diabetology, German Diabetes Center, c/o Auf'm Hennekamp 65, D-40225 Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), München, D-85764 Neuherberg, Germany.
| |
Collapse
|
194
|
Garić D, De Sanctis JB, Shah J, Dumut DC, Radzioch D. Biochemistry of very-long-chain and long-chain ceramides in cystic fibrosis and other diseases: The importance of side chain. Prog Lipid Res 2019; 74:130-144. [PMID: 30876862 DOI: 10.1016/j.plipres.2019.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/01/2019] [Accepted: 03/10/2019] [Indexed: 12/19/2022]
Abstract
Ceramides, the principal building blocks of all sphingolipids, have attracted the attention of many scientists around the world interested in developing treatments for cystic fibrosis, the most common genetic disease of Caucasians. Many years of fruitful research in this field have produced some fundamentally important, yet controversial results. Here, we aimed to summarize the current knowledge on the role of long- and very-long- chain ceramides, the most abundant species of ceramides in animal cells, in cystic fibrosis and other diseases. We also aim to explain the importance of the length of their side chain in the context of stability of transmembrane proteins through a concise synthesis of their biophysical chemistry, cell biology, and physiology. This review also addresses several remaining riddles in this field. Finally, we discuss the technical challenges associated with the analysis and quantification of ceramides. We provide the evaluation of the antibodies used for ceramide quantification and we demonstrate their lack of specificity. Results and discussion presented here will be of interest to anyone studying these enigmatic lipids.
Collapse
Affiliation(s)
- Dušan Garić
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Juan B De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Juhi Shah
- Department of Pharmacology and Experimental Therapeutics, McGill University, Montreal, QC, Canada
| | - Daciana Catalina Dumut
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Danuta Radzioch
- Department of Human Genetics, McGill University, Montreal, QC, Canada; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic; Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
195
|
Elevated peritoneal fluid ceramides in human endometriosis-associated infertility and their effects on mouse oocyte maturation. Fertil Steril 2019; 110:767-777.e5. [PMID: 30196975 DOI: 10.1016/j.fertnstert.2018.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/02/2018] [Accepted: 05/02/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To characterize the peritoneal fluid (PF) sphingolipid profile in endometriosis-associated infertility (EAI), and to assess the plausible functional role(s) of ceramides in oocyte maturation potential. DESIGN Retrospective case-control study and in vitro mouse oocyte study. SETTING University-affiliated hospital and university laboratory. SUBJECTS Twenty-seven infertile patients diagnosed with endometriosis and 20 infertile patients who did not have endometriosis; BALB/c female mice. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) PF sphingolipid concentrations. Number of metaphase II (MII) mouse oocytes. RESULT(S) Liquid chromatography-tandem mass spectrometry revealed 11 significantly elevated PF sphingolipids in infertile women with severe endometriosis compared with infertile women without endometriosis (change >50%, false discovery rate ≤10%). Logistic regression analysis identified three very-long-chain ceramides potentially associated with EAI. Functional studies revealed that very-long-chain ceramides may compromise or induce murine MII oocyte maturation. The oocyte maturation effects induced by the very long-chain ceramides were triggered by alterations in mitochondrial superoxide production in a concentration-dependent manner. Scavenging of mitochondrial superoxide reversed the maturation effects of C24:0 ceramide. CONCLUSION(S) EAI is associated with accumulation of PF very-long-chain ceramides. Mouse studies demonstrated how ceramides affect MII oocyte maturation, mediating through mitochondrial superoxide. These results provide an opportunity for direct functional readout of pathophysiology in EAI, and future therapies targeted at this sphingolipid metabolism may be harnessed for improved oocyte maturation.
Collapse
|
196
|
Kim MJ, Jeon S, Burbulla LF, Krainc D. Acid ceramidase inhibition ameliorates α-synuclein accumulation upon loss of GBA1 function. Hum Mol Genet 2019; 27:1972-1988. [PMID: 29579237 DOI: 10.1093/hmg/ddy105] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/19/2018] [Indexed: 11/14/2022] Open
Abstract
GBA1 encodes the lysosomal enzyme β-glucocerebrosidase (GCase) which converts glucosylceramide into ceramide and glucose. Mutations in GBA1 lead to Gaucher's disease and are a major risk factor for Parkinson's disease (PD) and Dementia with Lewy bodies (DLB), synucleinopathies characterized by accumulation of intracellular α-synuclein. In this study, we examined whether decreased ceramide that is observed in GCase-deficient cells contributes to α-synuclein accumulation. We demonstrated that deficiency of GCase leads to a reduction of C18-ceramide species and altered intracellular localization of Rab8a, a small GTPase implicated in secretory autophagy, that contributed to impaired secretion of α-synuclein and accumulation of intracellular α-synuclein. This secretory defect was rescued by exogenous C18-ceramide or chemical inhibition of lysosomal enzyme acid ceramidase that converts lysosomal ceramide into sphingosine. Inhibition of acid ceramidase by carmofur resulted in increased ceramide levels and decreased glucosylsphingosine levels in GCase-deficient cells, and also reduced oxidized α-synuclein and levels of ubiquitinated proteins in GBA1-PD patient-derived dopaminergic neurons. Together, these results suggest that decreased ceramide generation via the catabolic lysosomal salvage pathway in GCase mutant cells contributes to α-synuclein accumulation, potentially due to impaired secretory autophagy. We thus propose that acid ceramidase inhibition which restores ceramide levels may be a potential therapeutic strategy to target synucleinopathies linked to GBA1 mutations including PD and DLB.
Collapse
Affiliation(s)
- Myung Jong Kim
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sohee Jeon
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lena F Burbulla
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dimitri Krainc
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
197
|
Pant DC, Dorboz I, Schluter A, Fourcade S, Launay N, Joya J, Aguilera-Albesa S, Yoldi ME, Casasnovas C, Willis MJ, Ruiz M, Ville D, Lesca G, Siquier-Pernet K, Desguerre I, Yan H, Wang J, Burmeister M, Brady L, Tarnopolsky M, Cornet C, Rubbini D, Terriente J, James KN, Musaev D, Zaki MS, Patterson MC, Lanpher BC, Klee EW, Pinto E Vairo F, Wohler E, Sobreira NLDM, Cohen JS, Maroofian R, Galehdari H, Mazaheri N, Shariati G, Colleaux L, Rodriguez D, Gleeson JG, Pujades C, Fatemi A, Boespflug-Tanguy O, Pujol A. Loss of the sphingolipid desaturase DEGS1 causes hypomyelinating leukodystrophy. J Clin Invest 2019; 129:1240-1256. [PMID: 30620337 DOI: 10.1172/jci123959] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Abstract
Sphingolipid imbalance is the culprit in a variety of neurological diseases, some affecting the myelin sheath. We have used whole-exome sequencing in patients with undetermined leukoencephalopathies to uncover the endoplasmic reticulum lipid desaturase DEGS1 as the causative gene in 19 patients from 13 unrelated families. Shared features among the cases include severe motor arrest, early nystagmus, dystonia, spasticity, and profound failure to thrive. MRI showed hypomyelination, thinning of the corpus callosum, and progressive thalamic and cerebellar atrophy, suggesting a critical role of DEGS1 in myelin development and maintenance. This enzyme converts dihydroceramide (DhCer) into ceramide (Cer) in the final step of the de novo biosynthesis pathway. We detected a marked increase of the substrate DhCer and DhCer/Cer ratios in patients' fibroblasts and muscle. Further, we used a knockdown approach for disease modeling in Danio rerio, followed by a preclinical test with the first-line treatment for multiple sclerosis, fingolimod (FTY720, Gilenya). The enzymatic inhibition of Cer synthase by fingolimod, 1 step prior to DEGS1 in the pathway, reduced the critical DhCer/Cer imbalance and the severe locomotor disability, increasing the number of myelinating oligodendrocytes in a zebrafish model. These proof-of-concept results pave the way to clinical translation.
Collapse
Affiliation(s)
- Devesh C Pant
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Imen Dorboz
- INSERM UMR 1141, DHU PROTECT, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Agatha Schluter
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Stéphane Fourcade
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Nathalie Launay
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Javier Joya
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Sergio Aguilera-Albesa
- Pediatric Neurology Unit, Department of Pediatrics, Navarra Health Service, Navarrabiomed, Pamplona, Spain
| | - Maria Eugenia Yoldi
- Pediatric Neurology Unit, Department of Pediatrics, Navarra Health Service, Navarrabiomed, Pamplona, Spain
| | - Carlos Casasnovas
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain.,Neuromuscular Unit, Neurology Department, Hospital Universitari de Bellvitge, c/Feixa Llarga s/n, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mary J Willis
- Department of Pediatrics, Naval Medical Center San Diego, San Diego, California, USA
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Dorothée Ville
- Department of Neuropediatrics, Lyon University Hospital, Lyon, France
| | - Gaetan Lesca
- Department of Medical Genetics, Lyon University Hospital and GENDEV team CNRS UMR 5292, INSERM U1028, CRNL, and University Claude Bernard Lyon 1, Lyon, France
| | - Karine Siquier-Pernet
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Developmental Brain Disorders Laboratory, INSERM UMR 1163, Paris, France
| | - Isabelle Desguerre
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Developmental Brain Disorders Laboratory, INSERM UMR 1163, Paris, France
| | - Huifang Yan
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Molecular & Behavioral Neuroscience Institute, and
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Margit Burmeister
- Molecular & Behavioral Neuroscience Institute, and.,Departments of Computational Medicine & Bioinformatics, Psychiatry and Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Lauren Brady
- Department of Pediatrics (Neuromuscular and Neurometabolics), McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Mark Tarnopolsky
- Department of Pediatrics (Neuromuscular and Neurometabolics), McMaster Children's Hospital, Hamilton, Ontario, Canada
| | | | | | | | - Kiely N James
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, California, USA
| | - Damir Musaev
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, California, USA
| | - Maha S Zaki
- Human Genetics and Genome Research Division, Clinical Genetics Department, National Research Centre, Cairo, Egypt
| | - Marc C Patterson
- Departments of Neurology and Pediatrics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Eric W Klee
- Department of Clinical Genomics and.,Center for Individualized Medicine, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Filippo Pinto E Vairo
- Department of Clinical Genomics and.,Center for Individualized Medicine, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth Wohler
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nara Lygia de M Sobreira
- McKusick-Nathans Institute of Genetic Medicine, and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julie S Cohen
- Moser Center for Leukodystrophies at the Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Reza Maroofian
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St. George's, University of London, London, United Kingdom
| | - Hamid Galehdari
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Neda Mazaheri
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz, Iran
| | - Gholamreza Shariati
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz, Iran.,Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Laurence Colleaux
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Developmental Brain Disorders Laboratory, INSERM UMR 1163, Paris, France
| | - Diana Rodriguez
- APHP, Department of Neuropediatrics, National Reference Center for Neurogenetic Disorders, Hôpital Armand-Trousseau, GHUEP, Paris, France.,GRC ConCer-LD, Sorbonne Universités, UPMC Université, Paris, France
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, California, USA
| | - Cristina Pujades
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ali Fatemi
- Moser Center for Leukodystrophies at the Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Odile Boespflug-Tanguy
- INSERM UMR 1141, DHU PROTECT, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,Assistance Publique des Hopitaux de Paris (APHP), Reference Center for Leukodystrophies and Rare Leukoencephalopathies (LEUKOFRANCE), Hôpital Robert Debré, Paris, France
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain.,Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| |
Collapse
|
198
|
Al Sazzad MA, Möuts A, Palacios-Ortega J, Lin KL, Nyholm TKM, Slotte JP. Natural Ceramides and Lysophospholipids Cosegregate in Fluid Phosphatidylcholine Bilayers. Biophys J 2019; 116:1105-1114. [PMID: 30795873 DOI: 10.1016/j.bpj.2019.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/24/2019] [Accepted: 02/05/2019] [Indexed: 01/11/2023] Open
Abstract
The mode of interactions between palmitoyl lysophosphatidylcholine (palmitoyl lyso-PC) or other lysophospholipids (lyso-PLs) and palmitoyl ceramide (PCer) or other ceramide analogs in dioleoylphosphatidylcholine (DOPC) bilayers has been examined. PCer is known to segregate laterally into a ceramide-rich phase at concentrations that depend on the nature of the ceramides and the co-phospholipids. In DOPC bilayers, PCer forms a ceramide-rich phase at concentrations above 10 mol%. In the presence of 20 mol% palmitoyl lyso-PC in the DOPC bilayer, the lateral segregation of PCer was markedly facilitated (segregation at lower PCer concentrations). The thermostability of the PCer-rich phase in the presence of palmitoyl lyso-PC was also increased compared to that in the absence of palmitoyl lyso-PC. Other saturated lyso-PLs (e.g., palmitoyl lyso-phosphatidylethanolamine and lyso-sphingomyelin) also facilitated the lateral segregation of PCer in a similar manner as palmitoyl lyso-PC. When examined in the DOPC bilayer, it appeared that the association between palmitoyl lyso-PC and PCer was equimolar in nature. It is proposed that the interaction of PCer with lyso-PLs was driven by the need of ceramide to obtain a large-headgroup co-lipid, and saturated lyso-PLs were preferred co-lipids over DOPC because of the nature of their acyl chain. Structural analogs of PCer (1- or 3-deoxy-PCer) were also associated with palmitoyl lyso-PC, similarly to PCer, suggesting that the ceramide/lyso-PL interaction was not sensitive to structural alterations in the ceramide molecule. Binary complexes containing palmitoyl lyso-PC and ceramide were prepared, and these had a bilayer structure as ascertained by transmission electron microscopy. It is concluded that ceramides and lyso-PLs associated with each other both in binary bilayers and in ternary systems based on the DOPC bilayers. This association may have biological relevance under conditions in which both sphingomyelinases and phospholipase A2 enzymes are activated, such as during inflammatory processes.
Collapse
Affiliation(s)
- Md Abdullah Al Sazzad
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Anna Möuts
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Juan Palacios-Ortega
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Departamento de Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | - Kai-Lan Lin
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Thomas K M Nyholm
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - J Peter Slotte
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
| |
Collapse
|
199
|
Retention time bracketing for targeted sphingolipidomics by liquid chromatography-tandem mass spectrometry. Bioanalysis 2019; 11:185-201. [PMID: 30661375 DOI: 10.4155/bio-2018-0036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Aim: In complex biological matrixes, many sphingolipids are present with multiple reaction monitoring traces or lack of standard for verification, potentially leading to inaccurate identification and quantitation. Results/methodology: Based on these retention times of available standards, we devised a retention time bracketing approach to identify and predict sphingolipids of the same homologous series. Excellent concordance of predicted and observed retention times (<0.1 min) of sphingolipids were demonstrated. We also showed that many odd- and/or short-chain sphingolipids, commonly used as internal standards, are present in biological matrices including human serum, peritoneal fluid and cells. Conclusion: A retention time table, and a list of appropriate standards are presented, which are expected to be useful resources in targeted sphingolipidomics.
Collapse
|
200
|
Emergence of membrane sphingolipids as a potential therapeutic target. Biochimie 2019; 158:257-264. [PMID: 30703477 DOI: 10.1016/j.biochi.2019.01.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/23/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Though sphingolipids are ubiquitously present in eukaryotic cells, but until the last decade, they were merely considered as a structural component of the plasma membrane with limited function. However, over the last decade, numerous functions have been ascribed to sphingolipids after the seminal discoveries on the bioactivities of several sphingolipids. SCOPE OF REVIEW Sphingolipids are now well-recognized signals for fundamental cellular processes. Here we discussed about the advent of several sphingolipids components as potential therapeutic target for both human and plants. MAJOR CONCLUSIONS Sphingolipid contents and/or sphingolipid-metabolizing enzyme expression/activity often get impaired during pathophysiological conditions, and hence manipulation of this signaling pathway may be beneficial in disease diagnosis, and the plasma concentrations can serve as an important prognostic and diagnostic marker for the disease. GENERAL SIGNIFICANCE Sphingolipids are emerging as a goldmine for new therapeutic drug targets with promising new applications (cosmeceutical and nutraceutical), thereby opening new avenues for pharmaceuticals and nutraceutical industries.
Collapse
|