1
|
Hannun YA, Merrill AH, Luberto C. The Bioactive Sphingolipid Playbook. A Primer for the Uninitiated as well as Sphingolipidologists. J Lipid Res 2025:100813. [PMID: 40254066 DOI: 10.1016/j.jlr.2025.100813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025] Open
Abstract
Sphingolipids and glycosphingolipids are among the most structurally diverse and complex compounds in the mammalian metabolome. They are well known to play important roles in biological architecture, cell-cell communication and cellular regulation, and for many biological processes, multiple sphingolipids are involved. Thus, it is not surprising that untargeted genetic/transcriptomic/pharmacologic/metabolomic screens have uncovered changes in sphingolipids and sphingolipid genes/proteins while studying physiological and pathological processes. Consequently, with increasing frequency, both targeted and untargeted mass spectrometry methodologies are being used to conduct sphingolipidomic analyses. Interpretation of such large data sets and design of follow-up experiments can be daunting for investigators with limited expertise with sphingolipids (and sometimes even for someone well-versed in sphingolipidology). Therefore, this review gives an overview of essential elements of sphingolipid structure and analysis, metabolism, functions, and roles in disease, and discusses some of the items to consider when interpreting lipidomics data and designing follow-up investigations.
Collapse
Affiliation(s)
- Yusuf A Hannun
- Departments of Biochemistry, Medicine, and the Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| | - Alfred H Merrill
- School of Biological Sciences and the Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Chiara Luberto
- Department of Physiology and Biophysics, and the Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
2
|
Chitkara S, Atilla-Gokcumen GE. Decoding ceramide function: how localization shapes cellular fate and how to study it. Trends Biochem Sci 2025; 50:356-367. [PMID: 40000311 DOI: 10.1016/j.tibs.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025]
Abstract
Recent studies emphasize that lipid synthesis, metabolism, and transport are crucial in modulating lipid function, underscoring the significance of lipid localization within the cell, in addition to their chemical structure. Ceramides stand out in this context because of their multifaceted roles in cellular processes. Here, we focus on the role of ceramides in apoptosis, senescence, and autophagy as these processes offer unique and contrasting perspectives on how ceramides function and can be intricately linked to their subcellular localization, providing critical insights into their complex biological interactions. Additionally, we highlight recent advancements in tools and techniques that have boosted our understanding of ceramide dynamics and different mechanisms of lipid functioning.
Collapse
Affiliation(s)
- Shweta Chitkara
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - G Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA.
| |
Collapse
|
3
|
Ostermeyer-Fay AG, Kanodia A, Pathak R, Hernandez-Corbacho MJ, van der Spoel AC, Hannun YA, Canals D. The steady-state level of plasma membrane ceramide is regulated by neutral sphingomyelinase 2. J Lipid Res 2025; 66:100719. [PMID: 39631562 PMCID: PMC11742583 DOI: 10.1016/j.jlr.2024.100719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
During the last 30 years, an increasing number of cellular functions have been reported to be regulated by the lipid ceramide. The diversity in the ceramide structure, leading to tens of ceramide species and the discrete distribution based on subcellular topology, could explain the wide variety of functions attributed to this bioactive lipid. One of these pools of ceramide resides in the plasma membrane, and several works have suggested that an increase in plasma membrane ceramide (PMCer) in response to stimulation leads to cell death and modulates cell adhesion and migration. However, there is a limitation in studying PMCer content in this location primarily due to the inability to quantify its mass. Our group recently developed a method to specifically quantitate PMCer. In this work, we interrogate what sphingolipid metabolizing enzymes are responsible for modulating the basal levels of plasma membrane ceramide. An in-silico prediction and experimental confirmation found an almost perfect correlation between the endogenous expression levels of neutral sphingomyelinase (nSMase2) and the amount of plasma membrane ceramide in unstimulated cells. Manipulating the expression levels of nSMase2, but not other candidate enzymes of ceramide metabolism, profoundly affected PMCer. Moreover, a physiologic induction of nSMase2 during cell confluence resulted in a nSMase2-dependent dramatic increase in PMCer. Together, these results identify nSMase2 as the primary enzyme to regulate plasma membrane ceramide.
Collapse
Affiliation(s)
| | - Abhay Kanodia
- Department of Medicine, Cancer Center at Stony Brook, Stony Brook, NY, USA; Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, USA
| | - Ranjana Pathak
- Department of Medicine, Cancer Center at Stony Brook, Stony Brook, NY, USA
| | | | - Aarnoud C van der Spoel
- The Atlantic Research Centre, Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Yusuf A Hannun
- Department of Medicine, Cancer Center at Stony Brook, Stony Brook, NY, USA; Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA; Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Daniel Canals
- Department of Medicine, Cancer Center at Stony Brook, Stony Brook, NY, USA; Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA; Biological Mass Spectrometry Center, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
4
|
Yuan Y, Wu D, Chen H, Ma Z, Peng X, Li X, Zhao C, Jiang L, Liang J, Zhang W, Dai J. Farnesol ameliorates DSS-induced IBD by regulating inflammatory cytokines, repairing the intestinal barrier, reversing the gut microbiota imbalance, and influencing fecal metabolome in C57BL/6 mice. Biomed Pharmacother 2024; 180:117518. [PMID: 39405907 DOI: 10.1016/j.biopha.2024.117518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 11/14/2024] Open
Abstract
The incidence of inflammatory bowel disease (IBD) is rising globally, increasing interest in food ingredients for its prevention and control. This study evaluated the effect of farnesol (FAR), a key component of pomelo flower volatile oil, on dextran sodium sulfate (DSS)-induced colitis in C57BL/6 mice. FAR significantly alleviated DSS-induced colitis and secondary liver injury, as shown by improved body weight, DAI, colon length, and pathology, as well as liver function and blood lipid indices. The mechanism involves FAR-mediated regulation of inflammatory cytokines, increased expression of tight junction protein genes, and decreased expression of lipid metabolism-related proteins. FAR also enhanced gut microbiota diversity, balancing harmful and probiotic bacteria. Fecal metabolome analysis indicated FAR's role in reversing metabolic disturbances related to inflammation and liver lipid metabolism. These findings support developing functional foods for IBD treatment using pomelo flower volatile oil.
Collapse
Affiliation(s)
- Ya Yuan
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Dazuo Wu
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Heping Chen
- The 3rd Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital,Chengdu 611730, PR China
| | - Zheng Ma
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Xinyue Peng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Xiaodie Li
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Chuchu Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Linping Jiang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Jinping Liang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China
| | - Weiwei Zhang
- Department of Public Health, Chengdu Medical College, Chengdu 610500, PR China.
| | - Juan Dai
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, PR China.
| |
Collapse
|
5
|
Mori T, Niki T, Uchida Y, Mukai K, Kuchitsu Y, Kishimoto T, Sakai S, Makino A, Kobayashi T, Arai H, Yokota Y, Taguchi T, Suzuki KGN. A non-toxic equinatoxin-II reveals the dynamics and distribution of sphingomyelin in the cytosolic leaflet of the plasma membrane. Sci Rep 2024; 14:16872. [PMID: 39043900 PMCID: PMC11266560 DOI: 10.1038/s41598-024-67803-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Sphingomyelin (SM) is a major sphingolipid in mammalian cells. SM is enriched in the extracellular leaflet of the plasma membrane (PM). Besides this localization, recent electron microscopic and biochemical studies suggest the presence of SM in the cytosolic leaflet of the PM. In the present study, we generated a non-toxic SM-binding variant (NT-EqtII) based on equinatoxin-II (EqtII) from the sea anemone Actinia equina, and examined the dynamics of SM in the cytosolic leaflet of living cell PMs. NT-EqtII with two point mutations (Leu26Ala and Pro81Ala) had essentially the same specificity and affinity to SM as wild-type EqtII. NT-EqtII expressed in the cytosol was recruited to the PM in various cell lines. Super-resolution microscopic observation revealed that NT-EqtII formed tiny domains that were significantly colocalized with cholesterol and N-terminal Lyn. Meanwhile, single molecule observation at high resolutions down to 1 ms revealed that all the examined lipid probes including NT-EqtII underwent apparent fast simple Brownian diffusion, exhibiting that SM and other lipids in the cytosolic leaflet rapidly moved in and out of domains. Thus, the novel SM-binding probe demonstrated the presence of the raft-like domain in the cytosolic leaflet of living cell PMs.
Collapse
Affiliation(s)
- Toshiki Mori
- United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | - Takahiro Niki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yasunori Uchida
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Kojiro Mukai
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Yoshihiko Kuchitsu
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Takuma Kishimoto
- Division of Molecular Interaction, Institute for Genetic Medicine, Hokkaido University Graduate School of Life Science, Sapporo, Hokkaido, Japan
| | - Shota Sakai
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Asami Makino
- Lipid Biology Laboratory, RIKEN, Wako, Saitama, Japan
| | | | - Hiroyuki Arai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yasunari Yokota
- Department of EECE, Faculty of Engineering, Gifu University, Gifu, Japan
| | - Tomohiko Taguchi
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan.
| | - Kenichi G N Suzuki
- United Graduate School of Agricultural Science, Gifu University, Gifu, Japan.
- Institute for Glyco-Core Research (iGCORE), Gifu University, Gifu, Japan.
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan.
| |
Collapse
|
6
|
Coant N, Rendja K, Bellini L, Flamment M, Lherminier J, Portha B, Codogno P, Le Stunff H. Role of Sphingosine Kinase 1 in Glucolipotoxicity-Induced Early Activation of Autophagy in INS-1 Pancreatic β Cells. Cells 2024; 13:636. [PMID: 38607078 PMCID: PMC11011436 DOI: 10.3390/cells13070636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/04/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Insulin-producing pancreatic β cells play a crucial role in the regulation of glucose homeostasis, and their failure is a key event for diabetes development. Prolonged exposure to palmitate in the presence of elevated glucose levels, termed gluco-lipotoxicity, is known to induce β cell apoptosis. Autophagy has been proposed to be regulated by gluco-lipotoxicity in order to favor β cell survival. However, the role of palmitate metabolism in gluco-lipotoxcity-induced autophagy is presently unknown. We therefore treated INS-1 cells for 6 and 24 h with palmitate in the presence of low and high glucose concentrations and then monitored autophagy. Gluco-lipotoxicity induces accumulation of LC3-II levels in INS-1 at 6 h which returns to basal levels at 24 h. Using the RFP-GFP-LC3 probe, gluco-lipotoxicity increased both autophagosomes and autolysosmes structures, reflecting early stimulation of an autophagy flux. Triacsin C, a potent inhibitor of the long fatty acid acetyl-coA synthase, completely prevents LC3-II formation and recruitment to autophagosomes, suggesting that autophagic response requires palmitate metabolism. In contrast, etomoxir and bromo-palmitate, inhibitors of fatty acid mitochondrial β-oxidation, are unable to prevent gluco-lipotoxicity-induced LC3-II accumulation and recruitment to autophagosomes. Moreover, bromo-palmitate and etomoxir potentiate palmitate autophagic response. Even if gluco-lipotoxicity raised ceramide levels in INS-1 cells, ceramide synthase 4 overexpression does not potentiate LC3-II accumulation. Gluco-lipotoxicity also still stimulates an autophagic flux in the presence of an ER stress repressor. Finally, selective inhibition of sphingosine kinase 1 (SphK1) activity precludes gluco-lipotoxicity to induce LC3-II accumulation. Moreover, SphK1 overexpression potentiates autophagic flux induced by gluco-lipotxicity. Altogether, our results indicate that early activation of autophagy by gluco-lipotoxicity is mediated by SphK1, which plays a protective role in β cells.
Collapse
Affiliation(s)
- Nicolas Coant
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
- Department of Pathology and Stony Brook Cancer Center, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA
| | - Karima Rendja
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
| | - Lara Bellini
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
| | - Mélissa Flamment
- Inserm, UMR-S 872, Centre de Recherche des Cordeliers, 75006 Paris, France
| | - Jeannine Lherminier
- INRA, UMR1347 Agroécologie, ERL CNRS 6300, Plateforme DImaCell, Centre de Microscopie INRA/Université de Bourgogne, 21065 Dijon, France
| | - Bernard Portha
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
| | - Patrice Codogno
- INSERM U1151-CNRS UMR 8253, Institut Necker Enfants-Malades, University Paris Descartes, 75006 Paris, France
| | - Hervé Le Stunff
- Unité BFA, Université Paris Cité, CNRS UMR 8251, 75006 Paris, France; (N.C.); (B.P.)
- CNRS UMR 9197, Institut des Neurosciences Paris-Saclay, Saclay, University Paris, 91400 Saclay, France
| |
Collapse
|
7
|
Canals D, Hannun YA. Biological function, topology, and quantification of plasma membrane Ceramide. Adv Biol Regul 2024; 91:101009. [PMID: 38128364 PMCID: PMC11829740 DOI: 10.1016/j.jbior.2023.101009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Over the past 30 years, a growing body of evidence has revealed the regulatory role of the lipid ceramide in various cellular functions. The structural diversity of ceramide, resulting in numerous species, and its distinct distribution within subcellular compartments may account for its wide range of functions. However, our ability to study the potential role of ceramide in specific subcellular membranes has been limited. Several works have shown mitochondrial, Golgi, and plasma membrane ceramide to mediate signaling pathways independently. These results have started to shift the focus on ceramide signaling research toward specific membrane pools. Nonetheless, the challenge arises from the substantial intracellular ceramide content, hindering efforts to quantify its presence in particular membranes. Recently, we have developed the first method capable of detecting and quantifying ceramide in the plasma membrane, leading to unexpected results such as detecting different pools of ceramide responding to drug concentration or time. This review summarizes the historical context that defined the idea of pools of ceramide, the studies on plasma membrane ceramide as a bioactive entity, and the tools available for its study, especially the new method to detect and, for the first time, quantify plasma membrane ceramide. We believe this method will open new avenues for researching sphingolipid signaling and metabolism.
Collapse
Affiliation(s)
- Daniel Canals
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
8
|
El-Amouri S, Karakashian A, Bieberich E, Nikolova-Karakashian M. Regulated translocation of neutral sphingomyelinase-2 to the plasma membrane drives insulin resistance in steatotic hepatocytes. J Lipid Res 2023; 64:100435. [PMID: 37640282 PMCID: PMC10550728 DOI: 10.1016/j.jlr.2023.100435] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023] Open
Abstract
Obesity-associated diabetes is linked to the accumulation of ceramide in various organs, including the liver. The exact mechanisms by which ceramide contributes to diabetic pathology are unclear, but one proposed scenario is that ceramide accumulation may inhibit insulin signaling pathways. It is unknown however whether the excess ceramide is generated proximal to the insulin receptor, that is, at the plasma membrane (PM), where it could affect the insulin signaling pathway directly, or the onset of insulin resistance is due to ceramide-induced mitochondrial dysfunction and/or lipotoxicity. Using hepatic cell lines and primary cultures, gain- and loss- of function approach, and state-of-the art lipid imaging, this study shows that PM-associated neutral sphingomyelinase 2 (nSMase2) regulates ceramide homeostasis in fat-loaded hepatocytes and drives the onset of insulin resistance. Our results provide evidence of a regulated translocation of nSMase2 to the PM which leads to local generation of ceramide and insulin resistance in cells treated with palmitic acid (PAL), a type of fat commonly found in diabetogenic diets. Oleic acid, which also causes accumulation of lipid droplets, does not induce nSMase2 translocation and insulin resistance. Experiments using the acyl-biotin exchange method to quantify protein palmitoylation show that cellular PAL abundance regulates the rate of nSMase2 palmitoylation. Furthermore, while inhibition of nSMase2 with GW4869 prevents PAL-induced insulin resistance, the overexpression of wild type nSMase2 but not palmitoylation-defective mutant protein potentiates the suppressive effect of PAL on insulin signaling. Overall, this study identifies nSMase2 as a novel component of the mechanism of insulin resistance onset in fat-loaded hepatocytes, that is, cell-autonomous and driven by PAL.
Collapse
Affiliation(s)
- S El-Amouri
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - A Karakashian
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - E Bieberich
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - M Nikolova-Karakashian
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
9
|
KRAS Affects the Lipid Composition by Regulating Mitochondrial Functions and MAPK Activation in Bovine Mammary Epithelial Cells. Animals (Basel) 2022; 12:ani12223070. [PMID: 36428301 PMCID: PMC9686882 DOI: 10.3390/ani12223070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS), or guanosine triphosphatase KRAS, is a proto-oncogene that encodes the small guanosine triphosphatase transductor protein. Previous studies have found that KRAS can promote cytokine secretion, cell chemotaxis, and survival. However, its effects on milk fat synthesis in bovine mammary epithelial cells are unclear. In this study, the effects of KRAS inhibition on cell metabolism, autophagy, oxidative stress, endoplasmic reticulum stress, mitochondrial function, and lipid composition as well as the potential mechanisms were detected in an immortalized dairy cow mammary epithelial cell line (MAC-T). The results showed that inhibition of KRAS changed the lipid composition (especially the triglyceride level), mitochondrial functions, autophagy, and endoplasmic reticulum stress in cells. Moreover, KRAS inhibition regulated the levels of the mammalian target of rapamycin and mitogen-activated protein kinase (extracellular regulated protein kinases, c-Jun N-terminal kinases, p38) activation. These results indicated that regulation of KRAS would affect the synthesis and composition of milk fat. These results are also helpful for exploring the synthesis and secretion of milk fat at the molecular level and provide a theoretical basis for improving the percentage of fat in milk and the yield of milk from cows.
Collapse
|
10
|
Ca 2+-activated sphingomyelin scrambling and turnover mediate ESCRT-independent lysosomal repair. Nat Commun 2022; 13:1875. [PMID: 35388011 PMCID: PMC8986845 DOI: 10.1038/s41467-022-29481-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/11/2022] [Indexed: 12/14/2022] Open
Abstract
Lysosomes are vital organelles vulnerable to injuries from diverse materials. Failure to repair or sequester damaged lysosomes poses a threat to cell viability. Here we report that cells exploit a sphingomyelin-based lysosomal repair pathway that operates independently of ESCRT to reverse potentially lethal membrane damage. Various conditions perturbing organelle integrity trigger a rapid calcium-activated scrambling and cytosolic exposure of sphingomyelin. Subsequent metabolic conversion of sphingomyelin by neutral sphingomyelinases on the cytosolic surface of injured lysosomes promotes their repair, also when ESCRT function is compromised. Conversely, blocking turnover of cytosolic sphingomyelin renders cells more sensitive to lysosome-damaging drugs. Our data indicate that calcium-activated scramblases, sphingomyelin, and neutral sphingomyelinases are core components of a previously unrecognized membrane restoration pathway by which cells preserve the functional integrity of lysosomes.
Collapse
|
11
|
Zhang B, Zeng M, Wang Y, Li M, Wu Y, Xu R, Zhang Q, Jia J, Huang Y, Zheng X, Feng W. Oleic acid alleviates LPS-induced acute kidney injury by restraining inflammation and oxidative stress via the Ras/MAPKs/PPAR-γ signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153818. [PMID: 34798521 DOI: 10.1016/j.phymed.2021.153818] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/27/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Rehmannia Glutinosa Libosch. is applied for the treatment of renal and inflammatory-related diseases, and oleic acid (OA) is a compound isolated from Rehmannia Glutinosa Libosch.. Unfortunately, the pharmacological activity of OA on LPS treated acute kidney injury (AKI) has not been investigated. AIMS The research is aiming to probe the activities of OA on LPS-induced AKI. METHODS Information of OA effect on AKI were from network pharmacology. H&E staining, creatinine (CRE) and urea nitrogen (UN) were performed to evaluate the activities of OA on kidney function. Inflammatory factors in serum were measured by cytometric bead array. Increased ratio of reactive oxygen species (ROS) in kidney and immune cells in the peripheral blood were determined by flow cytometry (FCM). PPAR-γ, MAPK and apoptotic signaling pathways were measured by Western blot. Then, a metabolomics approach was utilized to investigate OA's response to AKI. The role of salirasib (FTS, Ras inhibitor) in OA acted on ROS, Ca2+, MMP and Ras signaling pathway in LPS treated NRK-52e cells were investigated by FCM and In-cell western. RESULTS It is proved that OA effetively ameliorated renal function, alleviated inflammatory response and oxidative stress, and transformed apoptotic, MAPK and PPAR-γ signaling pathways in mice with AKI, regulated phenylalanine metabolism, purine metabolism, sphingolipid metabolism, taurine and hypotaurine metabolism, moreover, the role of OA in injury of NRK-52e was blocked by FTS. CONCLUSION In a word, OA could alleviate AKI by restraining inflammation and oxidative stress via regulating the Ras/MAPKs/PPAR-γ signaling pathway, phenylalanine metabolism, purine metabolism, sphingolipid metabolism and taurine and hypotaurine metabolism, which might be a useful strategy for treating AKI.
Collapse
Affiliation(s)
- Beibei Zhang
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengnan Zeng
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yangyang Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Meng Li
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yuanyuan Wu
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Ruiqi Xu
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Qinqin Zhang
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Jufang Jia
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yanjie Huang
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Xiaoke Zheng
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| | - Weisheng Feng
- 156 Jinshui East Road, Henan University of Chinese Medicine, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| |
Collapse
|
12
|
Canals D, Clarke CJ. Compartmentalization of Sphingolipid metabolism: Implications for signaling and therapy. Pharmacol Ther 2021; 232:108005. [PMID: 34582834 DOI: 10.1016/j.pharmthera.2021.108005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022]
Abstract
Sphingolipids (SLs) are a family of bioactive lipids implicated in a variety of cellular processes, and whose levels are controlled by an interlinked network of enzymes. While the spatial distribution of SL metabolism throughout the cell has been understood for some time, the implications of this for SL signaling and biological outcomes have only recently begun to be fully explored. In this review, we outline the compartmentalization of SL metabolism and describe advances in tools for investigating and probing compartment-specific SL functions. We also briefly discuss the implications of SL compartmentalization for cell signaling and therapeutic approaches to targeting the SL network.
Collapse
Affiliation(s)
- Daniel Canals
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| | - Christopher J Clarke
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
13
|
Schneider-Schaulies S, Schumacher F, Wigger D, Schöl M, Waghmare T, Schlegel J, Seibel J, Kleuser B. Sphingolipids: Effectors and Achilles Heals in Viral Infections? Cells 2021; 10:cells10092175. [PMID: 34571822 PMCID: PMC8466362 DOI: 10.3390/cells10092175] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022] Open
Abstract
As viruses are obligatory intracellular parasites, any step during their life cycle strictly depends on successful interaction with their particular host cells. In particular, their interaction with cellular membranes is of crucial importance for most steps in the viral replication cycle. Such interactions are initiated by uptake of viral particles and subsequent trafficking to intracellular compartments to access their replication compartments which provide a spatially confined environment concentrating viral and cellular components, and subsequently, employ cellular membranes for assembly and exit of viral progeny. The ability of viruses to actively modulate lipid composition such as sphingolipids (SLs) is essential for successful completion of the viral life cycle. In addition to their structural and biophysical properties of cellular membranes, some sphingolipid (SL) species are bioactive and as such, take part in cellular signaling processes involved in regulating viral replication. It is especially due to the progress made in tools to study accumulation and dynamics of SLs, which visualize their compartmentalization and identify interaction partners at a cellular level, as well as the availability of genetic knockout systems, that the role of particular SL species in the viral replication process can be analyzed and, most importantly, be explored as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sibylle Schneider-Schaulies
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Würzburg, Germany; (S.S.-S.); (M.S.); (T.W.)
| | - Fabian Schumacher
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany; (F.S.); (D.W.)
| | - Dominik Wigger
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany; (F.S.); (D.W.)
| | - Marie Schöl
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Würzburg, Germany; (S.S.-S.); (M.S.); (T.W.)
| | - Trushnal Waghmare
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Würzburg, Germany; (S.S.-S.); (M.S.); (T.W.)
| | - Jan Schlegel
- Department for Biotechnology and Biophysics, University of Wuerzburg, 97074 Würzburg, Germany;
| | - Jürgen Seibel
- Department for Organic Chemistry, University of Wuerzburg, 97074 Würzburg, Germany;
| | - Burkhard Kleuser
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany; (F.S.); (D.W.)
- Correspondence: ; Tel.: +49-30-8386-9823
| |
Collapse
|
14
|
Canals D, Salamone S, Santacreu BJ, Aguilar D, Hernandez-Corbacho MJ, Ostermeyer-Fay AG, Greene M, Nemeth E, Haley JD, Obeid LM, Hannun YA. The doxorubicin-induced cell motility network is under the control of the ceramide-activated protein phosphatase 1 alpha. FASEB J 2021; 35:e21396. [PMID: 33583073 PMCID: PMC8220868 DOI: 10.1096/fj.202002427r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 12/20/2022]
Abstract
We have recently reported that a specific pool of ceramide, located in the plasma membrane, mediated the effects of sublethal doses of the chemotherapeutic compound doxorubicin on enhancing cancer cell migration. We identified neutral sphingomyelinase 2 (nSMase2) as the enzyme responsible to generate this bioactive pool of ceramide. In this work, we explored the role of members of the protein phosphatases 1 family (PP1), and we identified protein phosphatase 1 alpha isoform (PP1 alpha) as the specific PP1 isoform to mediate this phenotype. Using a bioinformatics approach, we build a functional interaction network based on phosphoproteomics data on plasma membrane ceramide. This led to the identification of several ceramide-PP1 alpha downstream substrates. Studies on phospho mutants of ezrin (T567) and Scrib (S1378/S1508) demonstrated that their dephosphorylation is sufficient to enhance cell migration. In summary, we identified a mechanism where reduced doses of doxorubicin result in the dysregulation of cytoskeletal proteins and enhanced cell migration. This mechanism could explain the reported effects of doxorubicin worsening cancer metastasis in animal models.
Collapse
Affiliation(s)
- Daniel Canals
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Silvia Salamone
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Bruno Jaime Santacreu
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
- Facultad de Farmacia y Bioquimica, Catedra de Biologia Celular y Molecular, Buenos Aires, Argentina
| | - Daniel Aguilar
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Barcelona, Catalunya, Spain
| | | | | | - Meaghan Greene
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Erika Nemeth
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - John D. Haley
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
- Department of Pathology, Stony Brook University, NY, USA
| | - Lina M. Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
- Northport VA Hospital, Northport, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Yusuf A. Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
- Department of Biochemistry, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
15
|
Li L, Wang H, Jones JW. Sphingolipid metabolism as a marker of hepatotoxicity in drug-induced liver injury. Prostaglandins Other Lipid Mediat 2020; 151:106484. [PMID: 33007444 PMCID: PMC7669681 DOI: 10.1016/j.prostaglandins.2020.106484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/09/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Drug-induced liver injury (DILI) has a substantial impact on human health and is a major monetary burden on the drug development process. Presently, there is a lack of robust and analytically validated markers for predicting and early diagnosis of DILI. Sphingolipid metabolism and subsequent disruption of sphingolipid homeostasis has been documented to play a key role contributing to hepatocellular death and subsequent liver injury. A more comprehensive understanding of sphingolipid metabolism in response to liver toxicity has great potential to gain mechanistic insight into hepatotoxicity and define molecular markers that are responsible for hepatocyte dysfunction. Here, we present an analytical platform that provides multidimensional mass spectrometry-based datasets for comprehensive structure characterization of sphingolipids extracted from human primary hepatocytes (HPH) exposed to toxic levels of acetaminophen (APAP). Sphingolipid metabolism as measured by characterization of individual sphingolipid structure was sensitive to APAP toxicity displaying a concentration-dependent response. A number of sphingolipid structures were differentially expressed across varying APAP exposures highlighting the unique role sphingolipid metabolism has in response to hepatotoxicity and its potential use as a molecular marker in DILI.
Collapse
Affiliation(s)
- Linhao Li
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States
| | - Hongbing Wang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States
| | - Jace W Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States.
| |
Collapse
|
16
|
Li CY, Niu M, Liu YL, Tang JF, Chen W, Qian G, Zhang MY, Shi YF, Lin JZ, Li XJ, Li RS, Xiao XH, Li GH, Wang JB. Screening for Susceptibility-Related Factors and Biomarkers of Xianling Gubao Capsule-Induced Liver Injury. Front Pharmacol 2020; 11:810. [PMID: 32547402 PMCID: PMC7274038 DOI: 10.3389/fphar.2020.00810] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
Although increasing reports from the literature on herbal-related hepatotoxicity, the identification of susceptibility-related factors and biomarkers remains challenging due to idiosyncratic drug-induced liver injury (IDILI). As a well-known Chinese medicine prescription, Xianling Gubao Capsule (XLGB) has attracted great attention due to reports of potential liver toxicity. But the mechanism behind it is difficult to determine. In this paper, we found that XLGB-induced liver injury belongs to IDILI through the analysis of clinical liver injury cases. In toxicological experiment assessment, co-exposure to XLGB and non-toxic dose of lipopolysaccharide (LPS) could cause evident liver injury as manifested by significantly increased plasma alanine aminotransferase activity and obvious liver histological damage. However, it failed to induce observable liver injury in normal rats, suggesting that mild immune stress may be a susceptibility factor for XLGB-induced idiosyncratic liver injury. Furthermore, plasma cytokines were determined and 15 cytokines (such as IL-1β, IFN-γ, and MIP-2α etc) were acquired by receiver operating characteristic (ROC) curves analysis. The expression of these 15 cytokines in LPS group was significantly up-regulated in contrast to the normal group. Meanwhile, the metabolomics profile showed that mild immune stress caused metabolic reprogramming, including sphingolipid metabolism, phenylalanine metabolism, and glycerophospholipid metabolism. 8 potential biomarkers (such as sphinganine, glycerophosphoethanolamine, and phenylalanine etc.) were identified by correlation analysis. Therefore, these results suggested that intracellular metabolism and immune changes induced by mild immune stress may be important susceptibility mechanisms for XLGB IDILI.
Collapse
Affiliation(s)
- Chun-Yu Li
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ya-Lei Liu
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jin-Fa Tang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Wei Chen
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Geng Qian
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Yu Zhang
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ya-Fei Shi
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun-Zhi Lin
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xing-Jie Li
- Research Center for Clinical and Translational Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Rui-Sheng Li
- Research Center for Clinical and Translational Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guo-Hui Li
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|