1
|
Wang R, Wang T, Su Y, Lin Z, Liu X, Jiao Y, Liu J, Chen E. High expression of SMPD4 promotes liver cancer and is associated with poor prognosis. BMC Res Notes 2025; 18:159. [PMID: 40211349 PMCID: PMC11987469 DOI: 10.1186/s13104-025-07212-4] [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: 11/11/2024] [Accepted: 03/27/2025] [Indexed: 04/13/2025] Open
Abstract
OBJECTIVES The expression of sphingomyelin phosphodiesterase 4 (SMPD4), a neutral sphingomyelin enzyme, is intricately associated with tumorigenesis and progression. However, its role in hepatocellular carcinoma (HCC) remains unclear. This study mainly reports the expression, prognostic value and tumor biological function of SMPD4 in HCC. METHODS The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and International Cancer Genome Consortium (ICGC) databases were utilized to investigate the expression patterns of SMPD4. Survival Analyses using the Kaplan-Meier method were conducted to assess the predictive value of SMPD4 in HCC. Immunohistochemistry method and real-time quantitative PCR were used to analyze the expression of SMPD4 in our clinical cohort. Immune infiltration analysis was performed to explore the correlation between SMPD4 expression and immune cell infiltration in HCC. Functional enrichment analysis was conducted to depict SMPD4-associated functions and pathways. Using human HCC cell lines, we studied the influence of SMPD4 in cell proliferation, invasion and migration. RESULTS We found SMPD4 was overexpressed in HCC. The Kaplan-Meier curves demonstrated that higher expression of SMPD4 was associated with worse survival in patients with HCC. Immune infiltration analysis showed that SMPD4 expression exhibited positive correlations with CD4 + T cells, Type 2 T helper cells, and negatively related to neutrophil, eosinophil, nature killer cells, macrophage, activated CD8 T cells. Functional enrichment analysis revealed that SMPD4 expression is associated with cell cycle pathways. Additionally, cell functional studies in HCC cell lines indicated that the knockdown of SMPD4 significantly inhibited cell growth, invasion and migration. CONCLUSIONS These results reveal that high SMPD4 expression is associated with poor prognosis and promotes HCC cell proliferation, invasion and migration. SMPD4 is a promising prognostic biomarker with functional significance for HCC.
Collapse
Affiliation(s)
- Rongyue Wang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
- Department of Clinical Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Ting Wang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Yanze Su
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Zhiheng Lin
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Xiaoping Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Yuanjun Jiao
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Jikui Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China.
| | - Erbao Chen
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China.
| |
Collapse
|
2
|
Zhu D, Cao L. SMPD3 as a Potential Biomarker and Therapeutic Target in Hepatocellular Carcinoma. Int J Genomics 2025; 2025:5443244. [PMID: 40226357 PMCID: PMC11991764 DOI: 10.1155/ijog/5443244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 11/30/2024] [Indexed: 04/15/2025] Open
Abstract
Background and Aims: Hepatocellular carcinoma (HCC) is a prevalent and aggressive liver cancer with high mortality rates. Sphingomyelin phosphodiesterase 3 (SMPD3) has recently been suggested to play an antitumor role in several cancers. This study is aimed at investigating the role of SMPD3 in HCC and its potential as a prognostic marker and therapeutic target. Methods: A retrospective cohort study of HCC patients was conducted using clinical data from our hospital. Survival analyses, including Kaplan-Meier and multivariate Cox regression, were performed to assess the impact of SMPD3 expression on survival. Further analyses were carried out using data from The Cancer Genome Atlas (TCGA) HCC cohort. In vitro and in vivo experiments were conducted to evaluate the effects of SMPD3 overexpression on HCC cell lines and tumor growth in mice. Results: High SMPD3 expression level was associated with improved survival in both our cohort and TCGA cohort. Multivariate Cox regression analysis confirmed high SMPD3 expression level as an independent predictor of better survival outcomes. In vitro and in vivo experiments demonstrated that SMPD3 overexpression significantly decreased HCC cell proliferation, migration, and invasion and inhibited tumor growth in a nude mouse model. Conclusions: SMPD3 plays a protective role in HCC by inhibiting tumor growth and progression. Its high expression is associated with better survival outcomes and may serve as a promising prognostic marker and potential therapeutic target in HCC. Further research into the molecular mechanisms of SMPD3's antitumor effects could lead to novel therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Lei Cao
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Bigge J, Koebbe LL, Giel AS, Bornholdt D, Buerfent B, Dasmeh P, Zink AM, Maj C, Schumacher J. Expression quantitative trait loci influence DNA damage-induced apoptosis in cancer. BMC Genomics 2024; 25:1168. [PMID: 39623312 PMCID: PMC11613471 DOI: 10.1186/s12864-024-11068-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Genomic instability and evading apoptosis are two fundamental hallmarks of cancer and closely linked to DNA damage response (DDR). By analyzing expression quantitative trait loci (eQTL) upon cell stimulation (called exposure eQTL (e2QTL)) it is possible to identify context specific gene regulatory variants and connect them to oncological diseases based on genome-wide association studies (GWAS). RESULTS We isolate CD8+ T cells from 461 healthy donors and stimulate them with high doses of 5 different carcinogens to identify regulatory mechanisms of DNA damage-induced apoptosis. Across all stimuli, we find 5,373 genes to be differentially expressed, with 85% to 99% of these genes being suppressed. While upregulated genes are specific to distinct stimuli, downregulated genes are shared across conditions but exhibit enrichment in biological processes depending on the DNA damage type. Analysis of eQTL reveals 654 regulated genes across conditions. Among them, 47 genes are significant e2QTL, representing a fraction of 4% to 5% per stimulus. To unveil disease relevant genetic variants, we compare eQTL and e2QTL with GWAS risk variants. We identify gene regulatory variants for KLF2, PIP4K2A, GPR160, RPS18, ARL17B and XBP1 that represent risk variants for oncological diseases. CONCLUSION Our study highlights the relevance of gene regulatory variants influencing DNA damage-induced apoptosis in cancer. The results provide new insights in cellular mechanisms and corresponding genes contributing to inter-individual effects in cancer development.
Collapse
Affiliation(s)
- Jessica Bigge
- Philipps University of Marburg, Center for Human Genetics, Marburg, Germany
| | - Laura L Koebbe
- Philipps University of Marburg, Center for Human Genetics, Marburg, Germany
| | - Ann-Sophie Giel
- Philipps University of Marburg, Center for Human Genetics, Marburg, Germany
| | - Dorothea Bornholdt
- Philipps University of Marburg, Center for Human Genetics, Marburg, Germany
| | - Benedikt Buerfent
- Philipps University of Marburg, Center for Human Genetics, Marburg, Germany
| | - Pouria Dasmeh
- Philipps University of Marburg, Center for Human Genetics, Marburg, Germany
| | | | - Carlo Maj
- Philipps University of Marburg, Center for Human Genetics, Marburg, Germany
| | - Johannes Schumacher
- Philipps University of Marburg, Center for Human Genetics, Marburg, Germany.
| |
Collapse
|
4
|
Butterfield ER, Obado SO, Scutts SR, Zhang W, Chait BT, Rout MP, Field MC. A lineage-specific protein network at the trypanosome nuclear envelope. Nucleus 2024; 15:2310452. [PMID: 38605598 PMCID: PMC11018031 DOI: 10.1080/19491034.2024.2310452] [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: 10/19/2023] [Accepted: 01/18/2024] [Indexed: 04/13/2024] Open
Abstract
The nuclear envelope (NE) separates translation and transcription and is the location of multiple functions, including chromatin organization and nucleocytoplasmic transport. The molecular basis for many of these functions have diverged between eukaryotic lineages. Trypanosoma brucei, a member of the early branching eukaryotic lineage Discoba, highlights many of these, including a distinct lamina and kinetochore composition. Here, we describe a cohort of proteins interacting with both the lamina and NPC, which we term lamina-associated proteins (LAPs). LAPs represent a diverse group of proteins, including two candidate NPC-anchoring pore membrane proteins (POMs) with architecture conserved with S. cerevisiae and H. sapiens, and additional peripheral components of the NPC. While many of the LAPs are Kinetoplastid specific, we also identified broadly conserved proteins, indicating an amalgam of divergence and conservation within the trypanosome NE proteome, highlighting the diversity of nuclear biology across the eukaryotes, increasing our understanding of eukaryotic and NPC evolution.
Collapse
Affiliation(s)
| | - Samson O. Obado
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
| | - Simon R. Scutts
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Wenzhu Zhang
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY, USA
| | - Brian T. Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY, USA
| | - Michael P. Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
| | - Mark C. Field
- School of Life Sciences, University of Dundee, Dundee, UK
- Biology Centre, Czech Academy of Sciences, Institute of Parasitology, České Budějovice, Czech Republic
| |
Collapse
|
5
|
Jamjoum R, Majumder S, Issleny B, Stiban J. Mysterious sphingolipids: metabolic interrelationships at the center of pathophysiology. Front Physiol 2024; 14:1229108. [PMID: 38235387 PMCID: PMC10791800 DOI: 10.3389/fphys.2023.1229108] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024] Open
Abstract
Metabolic pathways are complex and intertwined. Deficiencies in one or more enzymes in a given pathway are directly linked with genetic diseases, most of them having devastating manifestations. The metabolic pathways undertaken by sphingolipids are diverse and elaborate with ceramide species serving as the hubs of sphingolipid intermediary metabolism and function. Sphingolipids are bioactive lipids that serve a multitude of cellular functions. Being pleiotropic in function, deficiency or overproduction of certain sphingolipids is associated with many genetic and chronic diseases. In this up-to-date review article, we strive to gather recent scientific evidence about sphingolipid metabolism, its enzymes, and regulation. We shed light on the importance of sphingolipid metabolism in a variety of genetic diseases and in nervous and immune system ailments. This is a comprehensive review of the state of the field of sphingolipid biochemistry.
Collapse
Affiliation(s)
- Rama Jamjoum
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Saurav Majumder
- National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Rockville, MD, United States
| | - Batoul Issleny
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Johnny Stiban
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| |
Collapse
|
6
|
Aoki S, Watanabe K, Kato M, Konishi Y, Kubota K, Kobayashi E, Nakashima M, Saitsu H. Two novel cases of biallelic SMPD4 variants with brain structural abnormalities. Neurogenetics 2024; 25:3-11. [PMID: 37882972 DOI: 10.1007/s10048-023-00737-5] [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: 06/18/2023] [Accepted: 10/14/2023] [Indexed: 10/27/2023]
Abstract
Sphingomyelin phosphodiesterase 4 (SMPD4) encodes a member of the Mg2+-dependent, neutral sphingomyelinase family that catalyzes the hydrolysis of the phosphodiester bond of sphingomyelin to form phosphorylcholine and ceramide. Recent studies have revealed that biallelic loss-of-function variants of SMPD4 cause syndromic neurodevelopmental disorders characterized by microcephaly, congenital arthrogryposis, and structural brain anomalies. In this study, three novel loss-of-function SMPD4 variants were identified using exome sequencing (ES) in two independent patients with developmental delays, microcephaly, seizures, and brain structural abnormalities. Patient 1 had a homozygous c.740_741del, p.(Val247Glufs*21) variant and showed profound intellectual disability, hepatomegaly, a simplified gyral pattern, and a thin corpus callosum without congenital dysmorphic features. Patient 2 had a compound heterozygous nonsense c.2124_2125del, p.(Phe709*) variant and splice site c.1188+2dup variant. RNA analysis revealed that the c.1188+2dup variant caused exon 13 skipping, leading to a frameshift (p.Ala406Ser*6). In vitro transcription analysis using minigene system suggested that mRNA transcribed from mutant allele may be degraded by nonsense-mediated mRNA decay system. He exhibited diverse manifestations, including growth defects, muscle hypotonia, respiratory distress, arthrogryposis, insulin-dependent diabetes mellitus, sensorineural hearing loss, facial dysmorphism, and various brain abnormalities, including cerebral atrophy, hypomyelination, and cerebellar hypoplasia. Here, we review previous literatures and discuss the phenotypic diversity of SMPD4-related disorders.
Collapse
Affiliation(s)
- Shintaro Aoki
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
- Advanced Research Facilities & Services, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Watanabe
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kazuo Kubota
- Department of Pediatrics, Gifu University Graduate School of Medicine, Gifu, Japan
- Division of Clinical Genetics, Gifu University Hospital, Gifu, Japan
| | - Emiko Kobayashi
- Department of Pediatrics, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.
| |
Collapse
|
7
|
Kimura T, Kimura AK, Epand RM. Systematic crosstalk in plasmalogen and diacyl lipid biosynthesis for their differential yet concerted molecular functions in the cell. Prog Lipid Res 2023; 91:101234. [PMID: 37169310 DOI: 10.1016/j.plipres.2023.101234] [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: 03/03/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023]
Abstract
Plasmalogen is a major phospholipid of mammalian cell membranes. Recently it is becoming evident that the sn-1 vinyl-ether linkage in plasmalogen, contrasting to the ester linkage in the counterpart diacyl glycerophospholipid, yields differential molecular characteristics for these lipids especially related to hydrocarbon-chain order, so as to concertedly regulate biological membrane processes. A role played by NMR in gaining information in this respect, ranging from molecular to tissue levels, draws particular attention. We note here that a broad range of enzymes in de novo synthesis pathway of plasmalogen commonly constitute that of diacyl glycerophospholipid. This fact forms the basis for systematic crosstalk that not only controls a quantitative balance between these lipids, but also senses a defect causing loss of lipid in either pathway for compensation by increase of the counterpart lipid. However, this inherent counterbalancing mechanism paradoxically amplifies imbalance in differential effects of these lipids in a diseased state on membrane processes. While sharing of enzymes has been recognized, it is now possible to overview the crosstalk with growing information for specific enzymes involved. The overview provides a fundamental clue to consider cell and tissue type-dependent schemes in regulating membrane processes by plasmalogen and diacyl glycerophospholipid in health and disease.
Collapse
Affiliation(s)
- Tomohiro Kimura
- Department of Chemistry & Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506, USA.
| | - Atsuko K Kimura
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
8
|
Albi E, Mandarano M, Cataldi S, Ceccarini MR, Fiorani F, Beccari T, Sidoni A, Codini M. The Effect of Cholesterol in MCF7 Human Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24065935. [PMID: 36983016 PMCID: PMC10052157 DOI: 10.3390/ijms24065935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/14/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
In the last decade, cholesterol level has been implicated in several types of cancer, including breast cancer. In the current study, we aimed to investigate the condition of lipid depletion, hypocholesterolemia or hypercholesterolemia reproduced in vitro to analyze the response of different human breast cancer cells. Thus, MCF7 as the luminal A model, MB453 as the HER2 model and MB231 as the triple-negative model were used. No effect on cell growth and viability was detected in MB453 and MB231 cells. In MCF7 cells, hypocholesterolemia (1) reduced cell growth, and Ki67 expression; (2) increased ER/PgR expression; (3) stimulated the 3-Hydroxy-3-Methylglutaryl-CoA reductase and neutral sphingomyelinase and; (4) stimulated the expression of CDKN1A gene coding cyclin-dependent kinase inhibitor 1A protein, GADD45A coding growth arrest and DNA-damage-inducible alpha protein and, PTEN gene coding phosphatase and tensin homolog. All these effects were exacerbated by the lipid-depleted condition and reversed by the hypercholesterolemic condition. The relationship between cholesterol level and sphingomyelin metabolism was demonstrated. In summary, our data suggest that cholesterol levels should be controlled in luminal A breast cancer.
Collapse
Affiliation(s)
- Elisabetta Albi
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | - Martina Mandarano
- Division of Pathological Anatomy and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, 06126 Perugia, Italy
| | - Samuela Cataldi
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | | | - Federico Fiorani
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | - Angelo Sidoni
- Division of Pathological Anatomy and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, 06126 Perugia, Italy
| | - Michela Codini
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| |
Collapse
|
9
|
Piccoli M, Cirillo F, Ghiroldi A, Rota P, Coviello S, Tarantino A, La Rocca P, Lavota I, Creo P, Signorelli P, Pappone C, Anastasia L. Sphingolipids and Atherosclerosis: The Dual Role of Ceramide and Sphingosine-1-Phosphate. Antioxidants (Basel) 2023; 12:antiox12010143. [PMID: 36671005 PMCID: PMC9855164 DOI: 10.3390/antiox12010143] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Sphingolipids are bioactive molecules that play either pro- and anti-atherogenic roles in the formation and maturation of atherosclerotic plaques. Among SLs, ceramide and sphingosine-1-phosphate showed antithetic properties in regulating various molecular mechanisms and have emerged as novel potential targets for regulating the development of atherosclerosis. In particular, maintaining the balance of the so-called ceramide/S1P rheostat is important to prevent the occurrence of endothelial dysfunction, which is the trigger for the entire atherosclerotic process and is strongly associated with increased oxidative stress. In addition, these two sphingolipids, together with many other sphingolipid mediators, are directly involved in the progression of atherogenesis and the formation of atherosclerotic plaques by promoting the oxidation of low-density lipoproteins (LDL) and influencing the vascular smooth muscle cell phenotype. The modulation of ceramide and S1P levels may therefore allow the development of new antioxidant therapies that can prevent or at least impair the onset of atherogenesis, which would ultimately improve the quality of life of patients with coronary artery disease and significantly reduce their mortality.
Collapse
Affiliation(s)
- Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Andrea Ghiroldi
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Paola Rota
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy
| | - Simona Coviello
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Adriana Tarantino
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Faculty of Medicine and Surgery, University Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| | - Paolo La Rocca
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy
| | - Ivana Lavota
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Pasquale Creo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Paola Signorelli
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - Carlo Pappone
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Faculty of Medicine and Surgery, University Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
| | - Luigi Anastasia
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Faculty of Medicine and Surgery, University Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milan, Italy
- Correspondence: ; Tel.: +39-0226437765
| |
Collapse
|
10
|
Piët ACA, Post M, Dekkers D, Demmers JAA, Fornerod M. Proximity Ligation Mapping of Microcephaly Associated SMPD4 Shows Association with Components of the Nuclear Pore Membrane. Cells 2022; 11:cells11040674. [PMID: 35203325 PMCID: PMC8870324 DOI: 10.3390/cells11040674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
SMPD4 is a neutral sphingomyelinase implicated in a specific type of congenital microcephaly. Although not intensively studied, SMPD4 deficiency has also been found to cause cell division defects. This suggests a role for SMPD4 in cell-cycle and differentiation. In order to explore this role, we used proximity ligation to identify the partners of SMPD4 in vivo in HEK293T cells. We found that these partners localize near the endoplasmic reticulum (ER) and the nuclear membrane. Using mass spectrometry, we could identify these partners and discovered that SMPD4 is closely associated with several nucleoporins, including NUP35, a nucleoporin directly involved in pore membrane curvature and pore insertion. This suggests that SMPD4 may play a role in this process.
Collapse
Affiliation(s)
- Alexandra C. A. Piët
- Department of Cell Biology, ErasmusMC, Dr. Molewaterplein 40, 3015 GE Rotterdam, The Netherlands; (A.C.A.P.); (M.P.)
| | - Marco Post
- Department of Cell Biology, ErasmusMC, Dr. Molewaterplein 40, 3015 GE Rotterdam, The Netherlands; (A.C.A.P.); (M.P.)
| | - Dick Dekkers
- Proteomics Center, ErasmusMC, Dr. Molewaterplein 40, 3015 GE Rotterdam, The Netherlands; (D.D.); (J.A.A.D.)
| | - Jeroen A. A. Demmers
- Proteomics Center, ErasmusMC, Dr. Molewaterplein 40, 3015 GE Rotterdam, The Netherlands; (D.D.); (J.A.A.D.)
| | - Maarten Fornerod
- Department of Cell Biology, ErasmusMC, Dr. Molewaterplein 40, 3015 GE Rotterdam, The Netherlands; (A.C.A.P.); (M.P.)
- Correspondence:
| |
Collapse
|
11
|
Taniguchi M, Okazaki T. Role of ceramide/sphingomyelin (SM) balance regulated through "SM cycle" in cancer. Cell Signal 2021; 87:110119. [PMID: 34418535 DOI: 10.1016/j.cellsig.2021.110119] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
Sphingomyelin synthase (SMS), which comprises of two isozymes, SMS1 and SMS2, is the only enzyme that generates sphingomyelin (SM) by transferring phosphocholine of phosphatidylcholine to ceramide in mammals. Conversely, ceramide is generated from SM hydrolysis via sphingomyelinases (SMases), ceramide de novo synthesis, and the salvage pathway. The biosynthetic pathway for SM and ceramide content by SMS and SMase, respectively, is called "SM cycle." SM forms a SM-rich microdomain on the cell membrane to regulate signal transduction, such as proliferation/survival, migration, and inflammation. On the other hand, ceramide acts as a lipid mediator by forming a ceramide-rich platform on the membrane, and ceramide exhibits physiological actions such as cell death, cell cycle arrest, and autophagy induction. Therefore, the regulation of ceramide/SM balance by SMS and SMase is responsible for diverse cell functions not only in physiological cells but also in cancer cells. This review outlines the implications of ceramide/SM balance through "SM cycle" in cancer progression and prevention. In addition, the possible involvement of "SM cycle" is introduced in anti-cancer tumor immunity, which has become a hot topic to innovate a more effective and safer way to conquer cancer in recent years.
Collapse
Affiliation(s)
- Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0293, Japan
| | - Toshiro Okazaki
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, 1-308 Suematsu, Nonoichi-shi, Ishikawa 921-8836, Japan; Faculty of Advanced Life Science, Graduate School of Life Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo 060-0810, Japan.
| |
Collapse
|
12
|
Silvera S, Wilkinson JA, LeBlanc PJ. Characterization of neutral sphingomyelinase activity and isoform expression in rodent skeletal muscle mitochondria. Mitochondrion 2021; 59:184-189. [PMID: 34089907 DOI: 10.1016/j.mito.2021.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/04/2021] [Accepted: 06/01/2021] [Indexed: 12/01/2022]
Abstract
Skeletal muscle is composed of fiber types that differ in mitochondrial content, antioxidant capacity, and susceptibility to apoptosis. Ceramides have been linked to oxidative stress-mediated apoptotic intracellular signalling and the enzyme neutral sphingomyelinase (nSMase) is, in part, responsible for generating these ceramides through the hydrolysis of sphingomyelin. Despite the role of ceramides in mediating apoptosis, there is a gap in the literature regarding nSMase in skeletal muscle mitochondria. This study aimed to characterize total nSMase activity and individual isoform expression in isolated subsarcolemmal (SS) mitochondria from soleus, diaphragm, plantaris, and extensor digitorum longus (EDL). Total nSMase activity did not differ between muscle types. nSMase2 content was detectable in all muscles and higher in EDL, soleus, and plantaris compared to diaphragm whereas nSMase3 was undetectable in all muscles. Finally, total nSMase activity positively correlated to nSMase2 protein content in soleus but not the other muscles. These findings suggest that nSMase associated with SS mitochondria may play a role in intracellular signalling processes involving ceramides in skeletal muscle and nSMase2 may be the key isoform, specifically in slow twitch muscle like soleus. Further studies are needed to fully elucidate the specific contribution of nSMase, along with the role of the various isoforms and mitochondrial subpopulation in generating mitochondrial ceramides in skeletal muscle, and its potential effects on mediating apoptosis.
Collapse
Affiliation(s)
- Sebastian Silvera
- Center for Bone and Muscle Health, Faculty of Applied Health Science, Brock University, Canada
| | - Jennifer A Wilkinson
- Center for Bone and Muscle Health, Faculty of Applied Health Science, Brock University, Canada
| | - Paul J LeBlanc
- Center for Bone and Muscle Health, Faculty of Applied Health Science, Brock University, Canada.
| |
Collapse
|
13
|
Xiang H, Jin S, Tan F, Xu Y, Lu Y, Wu T. Physiological functions and therapeutic applications of neutral sphingomyelinase and acid sphingomyelinase. Biomed Pharmacother 2021; 139:111610. [PMID: 33957567 DOI: 10.1016/j.biopha.2021.111610] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 11/15/2022] Open
Abstract
Sphingomyelin (SM) can be converted into ceramide (Cer) by neutral sphingomyelinase (NSM) and acid sphingomyelinase (ASM). Cer is a second messenger of lipids and can regulate cell growth and apoptosis. Increasing evidence shows that NSM and ASM play key roles in many processes, such as apoptosis, immune function and inflammation. Therefore, NSM and ASM have broad prospects in clinical treatments, especially in cancer, cardiovascular diseases (such as atherosclerosis), nervous system diseases (such as Alzheimer's disease), respiratory diseases (such as chronic obstructive pulmonary disease) and the phenotype of dwarfisms in adolescents, playing a complex regulatory role. This review focuses on the physiological functions of NSM and ASM and summarizes their roles in certain diseases and their potential applications in therapy.
Collapse
Affiliation(s)
- Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengjie Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fenglang Tan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifan Xu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
14
|
Cirillo F, Piccoli M, Ghiroldi A, Monasky MM, Rota P, La Rocca P, Tarantino A, D'Imperio S, Signorelli P, Pappone C, Anastasia L. The antithetic role of ceramide and sphingosine-1-phosphate in cardiac dysfunction. J Cell Physiol 2021; 236:4857-4873. [PMID: 33432663 DOI: 10.1002/jcp.30235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/27/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally and the number of cardiovascular patients, which is estimated to be over 30 million in 2018, represent a challenging issue for the healthcare systems worldwide. Therefore, the identification of novel molecular targets to develop new treatments is an ongoing challenge for the scientific community. In this context, sphingolipids (SLs) have been progressively recognized as potent bioactive compounds that play crucial roles in the modulation of several key biological processes, such as proliferation, differentiation, and apoptosis. Furthermore, SLs involvement in cardiac physiology and pathophysiology attracted much attention, since these molecules could be crucial in the development of CVDs. Among SLs, ceramide and sphingosine-1-phosphate (S1P) represent the most studied bioactive lipid mediators, which are characterized by opposing activities in the regulation of the fate of cardiac cells. In particular, maintaining the balance of the so-called ceramide/S1P rheostat emerged as an important novel therapeutical target to counteract CVDs. Thus, this review aims at critically summarizing the current knowledge about the antithetic roles of ceramide and S1P in cardiomyocytes dysfunctions, highlighting how the modulation of their metabolism through specific molecules, such as myriocin and FTY720, could represent a novel and interesting therapeutic approach to improve the management of CVDs.
Collapse
Affiliation(s)
- Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | - Andrea Ghiroldi
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | | | - Paola Rota
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Paolo La Rocca
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Adriana Tarantino
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy
| | - Sara D'Imperio
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy
| | - Paola Signorelli
- Department of Health Sciences, Biochemistry and Molecular Biology Laboratory, University of Milan, Milan, Italy
| | - Carlo Pappone
- Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy.,Faculty of Medicine and Surgery, University of Vita-Salute San Raffaele, Milan, Italy
| | - Luigi Anastasia
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Faculty of Medicine and Surgery, University of Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
15
|
Modulation of DNA Damage Response by Sphingolipid Signaling: An Interplay that Shapes Cell Fate. Int J Mol Sci 2020; 21:ijms21124481. [PMID: 32599736 PMCID: PMC7349968 DOI: 10.3390/ijms21124481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022] Open
Abstract
Although once considered as structural components of eukaryotic biological membranes, research in the past few decades hints at a major role of bioactive sphingolipids in mediating an array of physiological processes including cell survival, proliferation, inflammation, senescence, and death. A large body of evidence points to a fundamental role for the sphingolipid metabolic pathway in modulating the DNA damage response (DDR). The interplay between these two elements of cell signaling determines cell fate when cells are exposed to metabolic stress or ionizing radiation among other genotoxic agents. In this review, we aim to dissect the mediators of the DDR and how these interact with the different sphingolipid metabolites to mount various cellular responses.
Collapse
|
16
|
Acid and Neutral Sphingomyelinase Behavior in Radiation-Induced Liver Pyroptosis and in the Protective/Preventive Role of rMnSOD. Int J Mol Sci 2020; 21:ijms21093281. [PMID: 32384654 PMCID: PMC7247354 DOI: 10.3390/ijms21093281] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 02/08/2023] Open
Abstract
Sphingomyelins (SMs) are a class of relevant bioactive molecules that act as key modulators of different cellular processes, such as growth arrest, exosome formation, and the inflammatory response influenced by many environmental conditions, leading to pyroptosis, a form of programmed cell death due to Caspase-1 involvement. To study liver pyroptosis and hepatic SM metabolism via both lysosomal acid SMase (aSMase) and endoplasmic reticulum/nucleus neutral SMase (nSMase) during the exposure of mice to radiation and to ascertain if this process can be modulated by protective molecules, we used an experimental design (previously used by us) to evaluate the effects of both ionizing radiation and a specific protective molecule (rMnSOD) in the brain in collaboration with the Joint Institute for Nuclear Research, Dubna (Russia). As shown by the Caspase-1 immunostaining of the liver sections, the radiation resulted in the loss of the normal cell structure alongside a progressive and dose-dependent increase of the labelling, treatment, and pretreatment with rMnSOD, which had a significant protective effect on the livers. SM metabolic analyses, performed on aSMase and nSMase gene expression, as well as protein content and activity, proved that rMnSOD was able to significantly reduce radiation-induced damage by playing both a protective role via aSMase and a preventive role via nSMase.
Collapse
|
17
|
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
|
18
|
Machala M, Procházková J, Hofmanová J, Králiková L, Slavík J, Tylichová Z, Ovesná P, Kozubík A, Vondráček J. Colon Cancer and Perturbations of the Sphingolipid Metabolism. Int J Mol Sci 2019; 20:E6051. [PMID: 31801289 PMCID: PMC6929044 DOI: 10.3390/ijms20236051] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
The development and progression of colorectal cancer (CRC), a major cause of cancer-related death in the western world, is accompanied with alterations of sphingolipid (SL) composition in colon tumors. A number of enzymes involved in the SL metabolism have been found to be deregulated in human colon tumors, in experimental rodent studies, and in human colon cancer cells in vitro. Therefore, the enzymatic pathways that modulate SL levels have received a significant attention, due to their possible contribution to CRC development, or as potential therapeutic targets. Many of these enzymes are associated with an increased sphingosine-1-phosphate/ceramide ratio, which is in turn linked with increased colon cancer cell survival, proliferation and cancer progression. Nevertheless, more attention should also be paid to the more complex SLs, including specific glycosphingolipids, such as lactosylceramides, which can be also deregulated during CRC development. In this review, we focus on the potential roles of individual SLs/SL metabolism enzymes in colon cancer, as well as on the pros and cons of employing the current in vitro models of colon cancer cells for lipidomic studies investigating the SL metabolism in CRC.
Collapse
Affiliation(s)
- Miroslav Machala
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100 Brno, Czech Republic; (J.P.); (L.K.); (J.S.)
| | - Jiřina Procházková
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100 Brno, Czech Republic; (J.P.); (L.K.); (J.S.)
| | - Jiřina Hofmanová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic; (J.H.); (Z.T.); (P.O.); (A.K.); (J.V.)
| | - Lucie Králiková
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100 Brno, Czech Republic; (J.P.); (L.K.); (J.S.)
| | - Josef Slavík
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 62100 Brno, Czech Republic; (J.P.); (L.K.); (J.S.)
| | - Zuzana Tylichová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic; (J.H.); (Z.T.); (P.O.); (A.K.); (J.V.)
| | - Petra Ovesná
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic; (J.H.); (Z.T.); (P.O.); (A.K.); (J.V.)
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Poštovská 68/3, 60200 Brno, Czech Republic
| | - Alois Kozubík
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic; (J.H.); (Z.T.); (P.O.); (A.K.); (J.V.)
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic; (J.H.); (Z.T.); (P.O.); (A.K.); (J.V.)
| |
Collapse
|
19
|
Loss of SMPD4 Causes a Developmental Disorder Characterized by Microcephaly and Congenital Arthrogryposis. Am J Hum Genet 2019; 105:689-705. [PMID: 31495489 DOI: 10.1016/j.ajhg.2019.08.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023] Open
Abstract
Sphingomyelinases generate ceramide from sphingomyelin as a second messenger in intracellular signaling pathways involved in cell proliferation, differentiation, or apoptosis. Children from 12 unrelated families presented with microcephaly, simplified gyral pattern of the cortex, hypomyelination, cerebellar hypoplasia, congenital arthrogryposis, and early fetal/postnatal demise. Genomic analysis revealed bi-allelic loss-of-function variants in SMPD4, coding for the neutral sphingomyelinase-3 (nSMase-3/SMPD4). Overexpression of human Myc-tagged SMPD4 showed localization both to the outer nuclear envelope and the ER and additionally revealed interactions with several nuclear pore complex proteins by proteomics analysis. Fibroblasts from affected individuals showed ER cisternae abnormalities, suspected for increased autophagy, and were more susceptible to apoptosis under stress conditions, while treatment with siSMPD4 caused delayed cell cycle progression. Our data show that SMPD4 links homeostasis of membrane sphingolipids to cell fate by regulating the cross-talk between the ER and the outer nuclear envelope, while its loss reveals a pathogenic mechanism in microcephaly.
Collapse
|
20
|
Metabolic Nano-Machines: Extracellular Vesicles Containing Active Enzymes and Their Contribution to Liver Diseases. CURRENT PATHOBIOLOGY REPORTS 2019. [DOI: 10.1007/s40139-019-00197-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Abstract
Extracellular vesicles (EVs), and exosomes in particular, were initially considered as "garbage bags" for secretion of undesired cellular components. This view has changed considerably over the last two decades, and exosomes have now emerged as important organelles controlling cell-to-cell signaling. They are present in biological fluids and have important roles in the communication between cells in physiological and pathological processes. They are envisioned for clinical use as carriers of biomarkers, therapeutic targets, and vehicles for drug delivery. Important efforts are being made to characterize the contents of these vesicles and to understand the mechanisms that govern their biogenesis and modes of action. This chapter aims to recapitulate the place given to lipids in our understanding of exosome biology. Besides their structural role and their function as carriers, certain lipids and lipid-modifying enzymes seem to exert privileged functions in this mode of cellular communication. By extension, the use of selective "lipid inhibitors" might turn out to be interesting modulators of exosomal-based cell signaling.
Collapse
Affiliation(s)
- Antonio Luis Egea-Jimenez
- Centre de Recherche en Cancérologie de Marseille, Equipe labellisée Ligue 2018, Aix-Marseille Université, Inserm, CNRS, Institut Paoli Calmettes, Marseille, France.,Department of Human Genetics, K. U. Leuven, Leuven, Belgium
| | - Pascale Zimmermann
- Centre de Recherche en Cancérologie de Marseille, Equipe labellisée Ligue 2018, Aix-Marseille Université, Inserm, CNRS, Institut Paoli Calmettes, Marseille, France. .,Department of Human Genetics, K. U. Leuven, Leuven, Belgium.
| |
Collapse
|
22
|
Lin M, Liao W, Dong M, Zhu R, Xiao J, Sun T, Chen Z, Wu B, Jin J. Exosomal neutral sphingomyelinase 1 suppresses hepatocellular carcinoma via decreasing the ratio of sphingomyelin/ceramide. FEBS J 2018; 285:3835-3848. [PMID: 30106227 DOI: 10.1111/febs.14635] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/19/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022]
Abstract
Neutral sphingomyelinase 1 (NSMase1) mediates caspase-3 activation and apoptosis. However, the role of NSMase1, especially exosome-borne NSMase1 in hepatocellular carcinoma (HCC), remains unclear. We report that NSMase1, which converts sphingomyelin (SM) to ceramide, was significantly downregulated in HCC tissues. Low NSMase1 expression predicted poor long-term survival of HCC patients. NSMase1 downregulation in HCC resulted in increased SM and reduced ceramide (Cer) that led to an increased SM/Cer ratio. Interestingly, NSMase1 and NSMase activity were also decreased in exosomes isolated from HCC tissues and cell lines. Furthermore, NSMase activity increased in exosomes isolated from the culture medium of L02 cells transfected with pEGFP-C3-NSMase1 (NSMase1-Exo). NSMase1-Exo suppressed HCC cell growth and induced apoptosis via reduction of the SM/Cer ratio. Thus, NSMase1 in exosomes inhibits HCC growth by decreasing the SM/Cer ratio.
Collapse
Affiliation(s)
- Minglin Lin
- Department of Surgery, Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Mingjun Dong
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Rongping Zhu
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Juan Xiao
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Tian Sun
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China
| | - Zhiyong Chen
- Light of Life Biotechnology, Ltd., Changsha, China
| | - Bin Wu
- Light of Life Biotechnology, Ltd., Changsha, China
| | - Junfei Jin
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, China.,China-USA Lipids in Health and Disease Research Center, Guilin Medical University, China.,Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, China
| |
Collapse
|
23
|
Abstract
For many years, neutral sphingomyelinases (N-SMases) were long thought to be anticancer enzymes owing to their roles as key producers of ceramide linked to apoptosis, growth arrest, and the chemotherapeutic response. However, in recent years, with the cloning of multiple isoforms and with new information on their cellular roles, particularly for nSMase2, a more complex picture is emerging suggesting that N-SMases have both pro- and anticancer roles. In this chapter, we will summarize current knowledge on N-SMase expression in cancer and the roles of N-SMase activity and specific isoforms in cancer-relevant biologies. We will also discuss what we see as the major challenges ahead for research into N-SMases in cancer.
Collapse
Affiliation(s)
- Christopher J Clarke
- Department of Medicine and Cancer Center, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
24
|
Adada M, Luberto C, Canals D. Inhibitors of the sphingomyelin cycle: Sphingomyelin synthases and sphingomyelinases. Chem Phys Lipids 2016. [DOI: 10.1016/j.chemphyslip.2015.07.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
25
|
Bienias K, Fiedorowicz A, Sadowska A, Prokopiuk S, Car H. Regulation of sphingomyelin metabolism. Pharmacol Rep 2016; 68:570-81. [PMID: 26940196 DOI: 10.1016/j.pharep.2015.12.008] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 11/24/2015] [Accepted: 12/28/2015] [Indexed: 12/17/2022]
Abstract
Sphingolipids (SFs) represent a large class of lipids playing diverse functions in a vast number of physiological and pathological processes. Sphingomyelin (SM) is the most abundant SF in the cell, with ubiquitous distribution within mammalian tissues, and particularly high levels in the Central Nervous System (CNS). SM is an essential element of plasma membrane (PM) and its levels are crucial for the cell function. SM content in a cell is strictly regulated by the enzymes of SM metabolic pathways, which activities create a balance between SM synthesis and degradation. The de novo synthesis via SM synthases (SMSs) in the last step of the multi-stage process is the most important pathway of SM formation in a cell. The SM hydrolysis by sphingomyelinases (SMases) increases the concentration of ceramide (Cer), a bioactive molecule, which is involved in cellular proliferation, growth and apoptosis. By controlling the levels of SM and Cer, SMSs and SMases maintain cellular homeostasis. Enzymes of SM cycle exhibit unique properties and diverse tissue distribution. Disturbances in their activities were observed in many CNS pathologies. This review characterizes the physiological roles of SM and enzymes controlling SM levels as well as their involvement in selected pathologies of the Central Nervous System, such as ischemia/hypoxia, Alzheimer disease (AD), Parkinson disease (PD), depression, schizophrenia and Niemann Pick disease (NPD).
Collapse
Affiliation(s)
- Kamil Bienias
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland
| | - Anna Fiedorowicz
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland; Laboratory of Tumor Molecular Immunobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Anna Sadowska
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland
| | - Sławomir Prokopiuk
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Białystok, Białystok, Poland.
| |
Collapse
|
26
|
Shamseddine AA, Clarke CJ, Carroll B, Airola MV, Mohammed S, Rella A, Obeid LM, Hannun YA. P53-dependent upregulation of neutral sphingomyelinase-2: role in doxorubicin-induced growth arrest. Cell Death Dis 2015; 6:e1947. [PMID: 26512957 PMCID: PMC4632297 DOI: 10.1038/cddis.2015.268] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 12/16/2022]
Abstract
Neutral sphingomyelinase-2 (nSMase2) is a ceramide-generating enzyme that has been implicated in growth arrest, apoptosis and exosome secretion. Although previous studies have reported transcriptional upregulation of nSMase2 in response to daunorubicin, through Sp1 and Sp3 transcription factors, the role of the DNA damage pathway in regulating nSMase2 remains unclear. In this study, we show that doxorubicin induces a dose-dependent induction of nSMase2 mRNA and protein with concomitant increases in nSMase activity and ceramide levels. Upregulation of nSMase2 was dependent on ATR, Chk1 and p53, thus placing it downstream of the DNA damage pathway. Moreover, overexpression of p53 was sufficient to transcriptionally induce nSMase2, without the need for DNA damage. DNA-binding mutants as well as acetylation mutants of p53 were unable to induce nSMase2, suggesting a role of nSMase2 in growth arrest. Moreover, knockdown of nSMase2 prevented doxorubicin-induced growth arrest. Finally, p53-induced nSMase2 upregulation appears to occur via a novel transcription start site upstream of exon 3. These results identify nSMase2 as a novel p53 target gene, regulated by the DNA damage pathway to induce cell growth arrest.
Collapse
Affiliation(s)
- A A Shamseddine
- Department of Medicine, Stony Brook University, Health Science Center, Stony Brook, NY 11794-8430, USA
| | - C J Clarke
- Department of Medicine, Stony Brook University, Health Science Center, Stony Brook, NY 11794-8430, USA
| | - B Carroll
- Department of Medicine, Stony Brook University, Health Science Center, Stony Brook, NY 11794-8430, USA
| | - M V Airola
- Department of Medicine, Stony Brook University, Health Science Center, Stony Brook, NY 11794-8430, USA
| | - S Mohammed
- Department of Medicine, Stony Brook University, Health Science Center, Stony Brook, NY 11794-8430, USA
| | - A Rella
- Stony Brook University Cancer Center, Stony Brook, NY 11794-8430, USA
| | - L M Obeid
- Department of Medicine, Stony Brook University, Health Science Center, Stony Brook, NY 11794-8430, USA
- Department of Microbiology and Immunology at Stony Brook University, Stony Brook, NY 11794-8430, USA
| | - Y A Hannun
- Department of Medicine, Stony Brook University, Health Science Center, Stony Brook, NY 11794-8430, USA
- Stony Brook University Cancer Center, Stony Brook, NY 11794-8430, USA
- The Northport Veterans Affairs Hospital, Northport, NY 11768, USA
| |
Collapse
|
27
|
Makoukji J, Raad M, Genadry K, El-Sitt S, Makhoul NJ, Saad Aldin E, Nohra E, Jabbour M, Sangaralingam A, Chelala C, Habib RH, Boulos F, Tfayli A, Boustany RM. Association between CLN3 (Neuronal Ceroid Lipofuscinosis, CLN3 Type) Gene Expression and Clinical Characteristics of Breast Cancer Patients. Front Oncol 2015; 5:215. [PMID: 26528430 PMCID: PMC4601263 DOI: 10.3389/fonc.2015.00215] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/22/2015] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most common cancer in women worldwide. Elucidation of underlying biology and molecular pathways is necessary for improving therapeutic options and clinical outcomes. CLN3 protein (CLN3p), deficient in neurodegenerative CLN3 disease is anti-apoptotic, and defects in the CLN3 gene cause accelerated apoptosis of neurons in CLN3 disease and up-regulation of ceramide. Dysregulated apoptotic pathways are often implicated in the development of the oncogenic phenotype. Predictably, CLN3 mRNA expression and CLN3 protein were up-regulated in a number of human and murine breast cancer-cell lines. Here, we determine CLN3 expression in non-tumor vs. tumor samples from fresh and formalin-fixed/paraffin-embedded (FFPE) breast tissue and analyze the association between CLN3 overexpression and different clinicopathological characteristics of breast cancer patients. Additionally, gene expression of 28 enzymes involved in sphingolipid metabolism was determined. CLN3 mRNA is overexpressed in tumor vs. non-tumor breast tissue from FFPE and fresh samples, as well as in mouse MCF7 breast cancer compared to MCF10A normal cells. Of the clinicopathological characteristics of tumor grade, age, menopause status, estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2), only absence of HER2 expression correlated with CLN3 overexpression. Sphingolipid genes for ceramide synthases 2 and 6 (CerS2; CerS6), delta(4)-desaturase sphingolipid 2 (DEGS2), and acidic sphingomyelinase (SMPD1) displayed higher expression levels in breast cancer vs. control tissue, whereas ceramide galactosyltransferase (UGT8) was underexpressed in breast cancer samples. CLN3 may be a novel molecular target for cancer drug discovery with the goal of modulation of ceramide pathways.
Collapse
Affiliation(s)
- Joelle Makoukji
- Department of Biochemistry and Molecular Genetics, American University of Beirut Medical Center , Beirut , Lebanon
| | - Mohamad Raad
- Department of Biochemistry and Molecular Genetics, American University of Beirut Medical Center , Beirut , Lebanon
| | - Katia Genadry
- Department of Biochemistry and Molecular Genetics, American University of Beirut Medical Center , Beirut , Lebanon
| | - Sally El-Sitt
- Department of Biochemistry and Molecular Genetics, American University of Beirut Medical Center , Beirut , Lebanon
| | - Nadine J Makhoul
- Department of Biochemistry and Molecular Genetics, American University of Beirut Medical Center , Beirut , Lebanon
| | - Ehab Saad Aldin
- Department of Radiology, University of Iowa Hospitals and Clinics , Iowa City, IA , USA
| | - Eden Nohra
- Department of Internal Medicine, American University of Beirut Medical Center , Beirut , Lebanon
| | - Mark Jabbour
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center , Beirut , Lebanon
| | - Ajanthah Sangaralingam
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London , London , UK
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London , London , UK
| | - Robert H Habib
- Outcomes Research Unit, American University of Beirut Medical Center , Beirut , Lebanon
| | - Fouad Boulos
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center , Beirut , Lebanon
| | - Arafat Tfayli
- Department of Internal Medicine, American University of Beirut Medical Center , Beirut , Lebanon
| | - Rose-Mary Boustany
- Department of Biochemistry and Molecular Genetics, American University of Beirut Medical Center , Beirut , Lebanon ; Neurogenetics Program, Division of Pediatric Neurology, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center , Beirut , Lebanon
| |
Collapse
|
28
|
Hwang SY, Kim TH, Lee HH. Neutral sphingomyelinase and breast cancer research. J Menopausal Med 2015; 21:24-7. [PMID: 26046034 PMCID: PMC4452810 DOI: 10.6118/jmm.2015.21.1.24] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 03/24/2015] [Accepted: 04/01/2015] [Indexed: 01/09/2023] Open
Abstract
Our understanding of the functions of neutral sphingomyelinase (N-SMase) signaling has advanced over the past decade. In this review, we focus on the roles and regulation of N-SMase 1, N-SMase 2, N-SMase 3, an enzyme that generates the bioactive lipid ceramide through the hydrolysis of the membrane lipid sphingomyelin. A large body of work has now implicated N-SMase 2 in a diverse set of cellular functions, physiological processes, and disease pathologies. We focus on different aspects of this enzyme's regulation from transcriptional, post-translational, and biochemical. Furthermore, we expected N-SMase involvement in cellular processes including inflammatory signaling, cell growth, apoptosis, and tumor necrosis factor which in turn play important roles in pathologies such as cancer metastasis, variable disease, and other organ system disorders. Lastly, we examine avenues where targeted N-SMase inhibition may be clinically beneficial in disease scenarios.
Collapse
Affiliation(s)
- Sun-Yong Hwang
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang University, Asan, Korea
| | - Tae-Hee Kim
- Department of Obstetrics and Gynecology, Soonchunhyang University, College of Medicine, Bucheon, Korea
| | - Hae-Hyeog Lee
- Department of Obstetrics and Gynecology, Soonchunhyang University, College of Medicine, Bucheon, Korea
| |
Collapse
|
29
|
Rajagopalan V, Canals D, Luberto C, Snider J, Voelkel-Johnson C, Obeid LM, Hannun YA. Critical determinants of mitochondria-associated neutral sphingomyelinase (MA-nSMase) for mitochondrial localization. Biochim Biophys Acta Gen Subj 2014; 1850:628-39. [PMID: 25484313 DOI: 10.1016/j.bbagen.2014.11.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/07/2014] [Accepted: 11/25/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND A novel murine mitochondria-associated neutral sphingomyelinase (MA-nSMase) has been recently cloned and partially characterized. The subcellular localization of the enzyme was found to be predominant in mitochondria. In this work, the determinants of mitochondrial localization and its topology were investigated. METHODS MA-nSMase mutants lacking consecutive regions and fusion proteins of GFP with truncated MA-nSMase regions were constructed and expressed in MCF-7 cells. Its localization was analyzed using confocal microscopy and sub-cellular fractionation methods. The sub-mitochondrial localization of MA-nSMase was determined using protease protection assay on isolated mitochondria. RESULTS The results initially showed that a putative mitochondrial localization signal (MLS), homologous to an MLS in the zebra-fish mitochondrial SMase is not necessary for the mitochondrial localization of the murine MA-nSMase. Evidence is provided to the presence of two regions in MA-nSMase that are sufficient for mitochondrial localization: a signal sequence (amino acids 24-56) that is responsible for the mitochondrial localization and an additional 'signal-anchor' sequence (amino acids 77-99) that anchors the protein to the mitochondrial membrane. This protein is topologically located in the outer mitochondrial membrane where both the C and N-termini remain exposed to the cytosol. CONCLUSIONS MA-nSMase is a membrane anchored protein with a MLS and a signal-anchor sequence at its N-terminal to localize it to the outer mitochondrial membrane. GENERAL SIGNIFICANCE Mitochondrial sphingolipids have been reported to play a critical role in cellular viability. This study opens a new window to investigate their cellular functions, and to define novel therapeutic targets.
Collapse
Affiliation(s)
- Vinodh Rajagopalan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173, Ashley Avenue, Charleston, SC 29425, USA
| | - Daniel Canals
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Chiara Luberto
- Stony Brook Cancer Center and the Department of Physiology and Biophysics, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Justin Snider
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Christina Voelkel-Johnson
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173, Ashley Avenue, Charleston, SC 29425, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Yusuf A Hannun
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA.
| |
Collapse
|
30
|
Carroll B, Donaldson JC, Obeid L. Sphingolipids in the DNA damage response. Adv Biol Regul 2014; 58:38-52. [PMID: 25434743 DOI: 10.1016/j.jbior.2014.11.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 12/16/2022]
Abstract
Recently, sphingolipid metabolizing enzymes have emerged as important targets of many chemotherapeutics and DNA damaging agents and therefore play significant roles in mediating the physiological response of the cell to DNA damage. In this review we will highlight points of connection between the DNA damage response (DDR) and sphingolipid metabolism; specifically how certain sphingolipid enzymes are regulated in response to DNA damage and how the bioactive lipids produced by these enzymes affect cell fate.
Collapse
Affiliation(s)
- Brittany Carroll
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jane Catalina Donaldson
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Lina Obeid
- Northport VA Medical Center, Northport, NY 11768, USA; Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
31
|
Moylan JS, Smith JD, Wolf Horrell EM, McLean JB, Deevska GM, Bonnell MR, Nikolova-Karakashian MN, Reid MB. Neutral sphingomyelinase-3 mediates TNF-stimulated oxidant activity in skeletal muscle. Redox Biol 2014; 2:910-20. [PMID: 25180167 PMCID: PMC4143815 DOI: 10.1016/j.redox.2014.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 11/26/2022] Open
Abstract
Aims Sphingolipid and oxidant signaling affect glucose uptake, atrophy, and force production of skeletal muscle similarly and both are stimulated by tumor necrosis factor (TNF), suggesting a connection between systems. Sphingolipid signaling is initiated by neutral sphingomyelinase (nSMase), a family of agonist-activated effector enzymes. Northern blot analyses suggest that nSMase3 may be a striated muscle-specific nSMase. The present study tested the hypothesis that nSMase3 protein is expressed in skeletal muscle and functions to regulate TNF-stimulated oxidant production. Results We demonstrate constitutive nSMase activity in skeletal muscles of healthy mice and humans and in differentiated C2C12 myotubes. nSMase3 (Smpd4 gene) mRNA is highly expressed in muscle. An nSMase3 protein doublet (88 and 85 kD) is derived from alternative mRNA splicing of exon 11. The proteins partition differently. The full-length 88 kD isoform (nSMase3a) fractionates with membrane proteins that are resistant to detergent extraction; the 85 kD isoform lacking exon 11 (nSMase3b) is more readily extracted and fractionates with detergent soluble membrane proteins; neither variant is detected in the cytosol. By immunofluorescence microscopy, nSMase3 resides in both internal and sarcolemmal membranes. Finally, myotube nSMase activity and cytosolic oxidant activity are stimulated by TNF. Both if these responses are inhibited by nSMase3 knockdown. Innovation These findings identify nSMase3 as an intermediate that links TNF receptor activation, sphingolipid signaling, and skeletal muscle oxidant production. Conclusion Our data show that nSMase3 acts as a signaling nSMase in skeletal muscle that is essential for TNF-stimulated oxidant activity. First measures of endogenous nSMase3 protein in muscle. Detection of nSMase3 splice variant proteins. Identification of a functional role for nSMase3 in redox signaling. Identification of an intermediate in TNF/redox signaling.
Collapse
Affiliation(s)
- Jennifer S Moylan
- Department of Physiology, University of Kentucky, Lexington, KY, USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Jeffrey D Smith
- Department of Physiology, University of Kentucky, Lexington, KY, USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Erin M Wolf Horrell
- Department of Physiology, University of Kentucky, Lexington, KY, USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA ; Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Julie B McLean
- Department of Physiology, University of Kentucky, Lexington, KY, USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Gergana M Deevska
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Mark R Bonnell
- Department of Surgery, University of Kentucky, Lexington, KY, USA
| | | | - Michael B Reid
- Department of Physiology, University of Kentucky, Lexington, KY, USA ; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
32
|
REVILL KATE, WANG TIM, LACHENMAYER ANJA, KOJIMA KENSUKE, HARRINGTON ANDREW, LI JINYU, HOSHIDA YUJIN, LLOVET JOSEPM, POWERS SCOTT. Genome-wide methylation analysis and epigenetic unmasking identify tumor suppressor genes in hepatocellular carcinoma. Gastroenterology 2013; 145:1424-35.e1-25. [PMID: 24012984 PMCID: PMC3892430 DOI: 10.1053/j.gastro.2013.08.055] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 07/30/2013] [Accepted: 08/29/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Epigenetic silencing of tumor suppressor genes contributes to the pathogenesis of hepatocellular carcinoma (HCC). To identify clinically relevant tumor suppressor genes silenced by DNA methylation in HCC, we integrated DNA methylation data from human primary HCC samples with data on up-regulation of gene expression after epigenetic unmasking. METHODS We performed genome-wide methylation analysis of 71 human HCC samples using the Illumina HumanBeadchip27K array; data were combined with those from microarray analysis of gene re-expression in 4 liver cancer cell lines after their exposure to reagents that reverse DNA methylation (epigenetic unmasking). RESULTS Based on DNA methylation in primary HCC and gene re-expression in cell lines after epigenetic unmasking, we identified 13 candidate tumor suppressor genes. Subsequent validation led us to focus on functionally characterizing 2 candidates, sphingomyelin phosphodiesterase 3 (SMPD3) and neurofilament, heavy polypeptide (NEFH), which we found to behave as tumor suppressor genes in HCC. Overexpression of SMPD3 and NEFH by stable transfection of inducible constructs into an HCC cell line reduced cell proliferation by 50% and 20%, respectively (SMPD3, P = .003 and NEFH, P = .003). Conversely, knocking down expression of these genes with small hairpin RNA promoted cell invasion and migration in vitro (SMPD3, P = .0001 and NEFH, P = .022), and increased their ability to form tumors after subcutaneous injection or orthotopic transplantation into mice, confirming their role as tumor suppressor genes in HCC. Low levels of SMPD3 were associated with early recurrence of HCC after curative surgery in an independent patient cohort (P = .001; hazard ratio = 3.22; 95% confidence interval: 1.6-6.5 in multivariate analysis). CONCLUSIONS Integrative genomic analysis identified SMPD3 and NEFH as tumor suppressor genes in HCC. We provide evidence that SMPD3 is a potent tumor suppressor gene that could affect tumor aggressiveness; a reduced level of SMPD3 is an independent prognostic factor for early recurrence of HCC.
Collapse
Affiliation(s)
- KATE REVILL
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, New York,Mount Sinai Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - TIM WANG
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, New York,Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - ANJA LACHENMAYER
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York,Department of General, Visceral, and Pediatric Surgery, University Hospital Düsseldorf, Düsseldorf, Germany
| | - KENSUKE KOJIMA
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - ANDREW HARRINGTON
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - JINYU LI
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, New York
| | - YUJIN HOSHIDA
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - JOSEP M. LLOVET
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York,HCC Translational Research Laboratory, Barcelona-Clinic Liver Cancer Group, Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), CIBERehd, Liver Unit, Hospital Clinic, University of Barcelona, Catalonia, Spain,Institucio Catalana de Recerca i Estudis Avancats (ICREA), Catalonia, Spain
| | - SCOTT POWERS
- Cancer Genome Center, Cold Spring Harbor Laboratory, Woodbury, New York
| |
Collapse
|
33
|
Abstract
Sphingolipid-metabolizing enzymes are becoming targets for chemotherapeutic development with an increasing interest in the recent years. In this chapter we introduce the sphingolipid family of lipids, and the role of individual species in cell homeostasis. We also discuss their roles in several rare diseases and overall, in cancer transformation. We follow the biosynthesis pathway of the sphingolipid tree, focusing on the enzymes in order to understand how using small molecule inhibitors makes it possible to modulate cancer progression. Finally, we describe the most used and historically significant inhibitors employed in cancer research, their relationships to sphingolipid metabolism, and some promising results found in this field.
Collapse
Affiliation(s)
- Daniel Canals
- Department of Medicine, University of Stony Brook, Stony Brook, New York 11794
| | - Yusuf A. Hannun
- Health Science Center, Stony Brook University, 100 Nicolls Road, L-4, 178, Stony Brook, NY 11794, USA
| |
Collapse
|
34
|
Mühle C, Reichel M, Gulbins E, Kornhuber J. Sphingolipids in psychiatric disorders and pain syndromes. Handb Exp Pharmacol 2013:431-456. [PMID: 23563670 DOI: 10.1007/978-3-7091-1511-4_22] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Despite the high prevalence and devastating impact of psychiatric disorders, little is known about their etiopathology. In this review, we provide an overview on the participation of sphingolipids and enzymes responsible for their metabolism in mechanisms underlying psychiatric disorders. We focus on the pathway from sphingomyelin to proapoptotic ceramide and the subsequent metabolism of ceramide to sphingosine, which is in turn phosphorylated to yield anti-apoptotic sphingosine-1-phosphate (S1P).The sphingomyelinase/ceramide system has been linked to effects of reactive oxygen species and proinflammatory cytokines in the central nervous system as well as to synaptic transmission. Compared to ubiquitously expressed acid sphingomyelinase, acid and neutral ceramidase and neutral sphingomyelinase are highly active in brain regions. Depressed patients show elevated plasma ceramide levels and increased activities of acid sphingomyelinase which is functionally inhibited by many anti-depressive drugs. Exposure to alcohol is associated with an activation of acid and neutral sphingomyelinase observed in cell culture, mouse models and in alcohol-dependent patients and with increased concentrations of ceramide in various organs.Levels of sphingomyelin and ceramide are altered in erythrocytes and post-mortem brain tissues of schizophrenic patients in addition to changes in expression patterns for serine palmitoyltransferase and acid ceramidase leading to impaired myelination. After induction of anxiety-like behavior in animal models, higher serum levels of S1P were reported to lead to neurodegeneration. Correspondingly, S1P infusion appeared to increase anxiety-like behavior. Significantly upregulated levels of the endogenous ceramide catabolite N,N-dimethylsphingosine were observed in rat models of allodynia. Conversely, rats injected intrathecally with N,N-dimethylsphingosine developed mechanical allodynia. Moreover, S1P has been implicated in spinal nociceptive processing.The increasing interest in lipidomics and improved analytical methods led to growing insight into the connection between psychiatric and neurological disorders and sphingolipid metabolism and may once provide new targets and strategies for therapeutic intervention.
Collapse
Affiliation(s)
- C Mühle
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen, Erlangen, Germany
| | | | | | | |
Collapse
|
35
|
Abstract
Sphingolipids are an important class of lipid molecules that play fundamental roles in our cells and body. Beyond a structural role, it is now clearly established that sphingolipids serve as bioactive signaling molecules to regulate diverse processes including inflammatory signaling, cell death, proliferation, and pain sensing. Sphingolipid metabolites have been implicated in the onset and progression of various diseases including cancer, lung disease, diabetes, and lysosomal storage disorders. Here we review sphingolipid metabolism to introduce basic concepts as well as emerging complexities in sphingolipid function gained from modern technological advances and detailed cell and animal studies. Furthermore, we discuss the family of neutral sphingomyelinases (N-SMases), which generate ceramide through the hydrolysis of sphingomyelin and are key enzymes in sphingolipid metabolism. Four mammalian N-SMase enzymes have now been identified. Most prominent is nSMase2 with established roles in bone mineralization, exosome formation, and cellular stress responses. Function for the other N-SMases has been more enigmatic and is an area of active investigation. The known properties and potential role(s) of each enzyme are discussed to help guide future studies.
Collapse
|
36
|
Abstract
Chemotherapy is frequently used to treat primary or metastatic cancers, but intrinsic or acquired drug resistance limits its efficiency. Sphingolipids are important regulators of various cellular processes including proliferation, apoptosis, differentiation, angiogenesis, stress, and inflammatory responses which are linked to various aspects of cancer, like tumor growth, neoangiogenesis, and response to chemotherapy. Ceramide, the central molecule of sphingolipid metabolism, generally mediates antiproliferative and proapoptotic functions, whereas sphingosine-1-phosphate and other derivatives have opposing effects. Among the variety of enzymes that control ceramide generation, acid or neutral sphingomyelinases and ceramide synthases are important targets to allow killing of cancer cells by chemotherapeutic drugs. On the contrary, glucosylceramide synthase, ceramidase, and sphingosine kinase are other targets driving cancer cell resistance to chemotherapy. This chapter focuses on ceramide-based mechanisms leading to cancer therapy sensitization or resistance which could have some impacts on the development of novel cancer therapeutic strategies.
Collapse
|
37
|
An J, Shi J, He Q, Lui K, Liu Y, Huang Y, Sheikh MS. CHCM1/CHCHD6, novel mitochondrial protein linked to regulation of mitofilin and mitochondrial cristae morphology. J Biol Chem 2012; 287:7411-7426. [PMID: 22228767 PMCID: PMC3293568 DOI: 10.1074/jbc.m111.277103] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 01/04/2012] [Indexed: 11/06/2022] Open
Abstract
The structural integrity of mitochondrial cristae is crucial for mitochondrial functions; however, the molecular events controlling the structural integrity and biogenesis of mitochondrial cristae remain to be fully elucidated. Here, we report the functional characterization of a novel mitochondrial protein named CHCM1 (coiled coil helix cristae morphology 1)/CHCHD6. CHCM1/CHCHD6 harbors a coiled coil helix-coiled coil helix domain at its C-terminal end and predominantly localizes to mitochondrial inner membrane. CHCM1/CHCHD6 knockdown causes severe defects in mitochondrial cristae morphology. The mitochondrial cristae in CHCM1/CHCHD6-deficient cells become hollow with loss of structural definitions and reduction in electron-dense matrix. CHCM1/CHCHD6 depletion also leads to reductions in cell growth, ATP production, and oxygen consumption. CHCM1/CHCHD6 through its C-terminal end strongly and directly interacts with the mitochondrial inner membrane protein mitofilin, which is known to also control mitochondrial cristae morphology. CHCM1/CHCHD6 also interacts with other mitofilin-associated proteins, including DISC1 and CHCHD3. Knockdown of CHCM1/CHCHD6 reduces mitofilin protein levels; conversely, mitofilin knockdown leads to reduction in CHCM1 levels, suggesting coordinate regulation between these proteins. Our results further indicate that genotoxic anticancer drugs that induce DNA damage down-regulate CHCM1/CHCHD6 expression in multiple human cancer cells, whereas mitochondrial respiratory chain inhibitors do not affect CHCM1/CHCHD6 levels. CHCM1/CHCHD6 knockdown in human cancer cells enhances chemosensitivity to genotoxic anticancer drugs, whereas its overexpression increases resistance. Collectively, our results indicate that CHCM1/CHCHD6 is linked to regulation of mitochondrial cristae morphology, cell growth, ATP production, and oxygen consumption and highlight its potential as a possible target for cancer therapeutics.
Collapse
Affiliation(s)
- Jie An
- From the Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York 13210
| | - Jingxue Shi
- From the Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York 13210
| | - Qin He
- From the Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York 13210
| | - Ki Lui
- From the Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York 13210
| | - Yuxin Liu
- From the Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York 13210
| | - Ying Huang
- From the Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York 13210
| | - M. Saeed Sheikh
- From the Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York 13210
| |
Collapse
|
38
|
The Roles of Neutral Sphingomyelinases in Neurological Pathologies. Neurochem Res 2012; 37:1137-49. [DOI: 10.1007/s11064-011-0692-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 11/25/2011] [Accepted: 12/29/2011] [Indexed: 12/14/2022]
|
39
|
Neutral sphingomyelinase 2 (nSMase2) is the primary neutral sphingomyelinase isoform activated by tumour necrosis factor-α in MCF-7 cells. Biochem J 2011; 435:381-90. [PMID: 21303347 DOI: 10.1042/bj20101752] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Activation of N-SMase (neutral sphingomyelinase) is an established part of the response of cytokines such as TNF (tumour necrosis factor)-α. However, it remains unclear which of the currently cloned N-SMase isoforms (nSMase1, nSMase2 and nSMase3) are responsible for this activity. In MCF-7 cells, we found that TNF-α induces late, but not early, increases in N-SMase activity, and that nSMase2 is the primary isoform activated, most likely through post-transcriptional mechanisms. Surprisingly, overexpression of tagged or untagged nSMase3 in multiple cell lines had no significant effect on in vitro N-SMase activity. Moreover, only overexpression of nSMase2, but not nSMase1 or nSMase3, had significant effects on cellular sphingolipid levels, increasing ceramide and decreasing sphingomyelin. Additionally, only siRNA (small interfering RNA) knockdown of nSMase1 significantly decreased basal in vitro N-SMase activity of MCF-7 cells, whereas nSMase2 but not nSMase3 siRNA inhibited TNF-α-induced activity. Taken together, these results identify nSMase2 as the major TNF-α-responsive N-SMase in MCF-7 cells. Moreover, the results suggest that nSMase3 may not possess in vitro N-SMase activity and does not affect cellular sphingolipid levels in the cell lines evaluated. On the other hand, nSMase1 contributes to in vitro N-SMase activity, but does not affect cellular sphingolipids much.
Collapse
|
40
|
Henry B, Möller C, Dimanche-Boitrel MT, Gulbins E, Becker KA. Targeting the ceramide system in cancer. Cancer Lett 2011; 332:286-94. [PMID: 21862212 DOI: 10.1016/j.canlet.2011.07.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 06/30/2011] [Accepted: 07/08/2011] [Indexed: 12/20/2022]
Abstract
Sphingolipids, in particular ceramide, have been described as important components of cellular signalling pathways. Ceramide can be produced via multiple mechanisms including through the hydrolysis of sphingomyelin by acid and neutral sphingomyelinase or by a de novo synthesis pathway. Recent studies have identified sphingomyelinases and ceramide synthases as important targets for γ-irradiation and chemotherapeutic drugs. Likewise, common cancer treatment modalities, such as γ-irradiation and many chemotherapeutic agents, induce cell death via the generation of ceramide. This suggests that the manipulation of ceramide production and metabolism could offer promising means for the enhancement of anti-tumor therapies. The focus of this mini-review will be to discuss contemporary evidence suggesting that ceramide forming pathways and ceramide itself are important targets for the treatment of tumors and the development of novel tumor treatment strategies.
Collapse
Affiliation(s)
- Brian Henry
- Dept. of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45 122 Essen, Germany
| | | | | | | | | |
Collapse
|
41
|
Abstract
Evidence has consistently indicated that activation of sphingomyelinases and/or ceramide synthases and the resulting accumulation of ceramide mediate cellular responses to stressors such as lipopolysaccharide, interleukin 1beta, tumor necrosis factor alpha, serum deprivation, irradiation and various antitumor treatments. Recent studies had identified the genes encoding most of the enzymes responsible for the generation of ceramide and ongoing research is aimed at characterizing their individual functions in cellular response to stress. This chapter discusses the seminal and more recent discoveries in regards to the pathways responsible for the accumulation of ceramide during stress and the mechanisms by which ceramide affects cell functions. The former group includes the roles of neutral sphingomyelinase 2, serine palmitoyltransferase, ceramide synthases, as well as the secretory and endosomal/lysosomal forms of acid sphingomyelinase. The latter summarizes the mechanisms by which ceramide activate its direct targets, PKCzeta, PP2A and cathepsin D. The ability of ceramide to affect membrane organization is discussed in the light of its relevance to cell signaling. Emerging evidence to support the previously assumed notion that ceramide acts in a strictly structure-specific manner are also included. These findings are described in the context of several physiological and pathophysiological conditions, namely septic shock, obesity-induced insulin resistance, aging and apoptosis of tumor cells in response to radiation and chemotherapy.
Collapse
|
42
|
Remodeling of sphingolipids by plasma membrane associated enzymes. Neurochem Res 2010; 36:1636-44. [PMID: 21181265 DOI: 10.1007/s11064-010-0360-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2010] [Indexed: 12/16/2022]
Abstract
The sphingolipid plasma membrane content and pattern is the result of several processes, among which the main, in term of quantity, are: neo-biosynthesis in endoplasmic reticulum and Golgi apparatus, membrane turnover with final catabolism in lysosomes and membrane shedding. In addition to this, past and recent data suggest that the head group of sphingolipids can be opportunely modified at the plasma membrane level, probably inside specific membrane lipid domains, by the action of enzymes involved in the sphingolipids metabolism, working directly at the cell surface. The number of membrane enzymes, hydrolases and transferases, acting on membrane sphingolipids is growing very rapidly. In this report we describe some properties of these enzymes.
Collapse
|
43
|
Milhas D, Clarke CJ, Idkowiak-Baldys J, Canals D, Hannun YA. Anterograde and retrograde transport of neutral sphingomyelinase-2 between the Golgi and the plasma membrane. Biochim Biophys Acta Mol Cell Biol Lipids 2010; 1801:1361-74. [PMID: 20713176 DOI: 10.1016/j.bbalip.2010.08.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/30/2010] [Accepted: 08/02/2010] [Indexed: 02/06/2023]
Abstract
The activation of neutral sphingomyelinase-2 (nSMase2) and consequent ceramide production are implicated in many stress-induced signaling pathways. Trafficking of nSMase2 from the Golgi compartment to the plasma membrane (PM) in response to signaling stimuli has been described. However, the precise mechanisms of transport remain unknown. This study aimed to investigate the trafficking of nSMase2 between the Golgi and the PM. We show here that V5-nSMase2 localizes at the PM and Golgi in MCF-7 cells and confirm relocalization of nSMase2 to the PM at confluence. Although cycloheximide (CHX) treatment partially inhibited the Golgi localization of GFP-nSMase2, recovery of GFP-nSMase2 to an intracellular compartment was still observed after photobleaching. Moreover, in the presence of CHX, GFP- and V5-nSMase2 co-localized with endosomal/recycling markers. In HEK293 cells, activation of either protein kinase C-alpha or betaII, with the phorbol ester PMA led to relocalization of both wild-type and inactive nSMase2 to the pericentrion, a PKC-dependent subset of recycling endosomes. Finally, inhibition of nSMase2 endocytosis by K+depletion reduced the intracellular pool of nSMase2 and increased nSMase2 activity resulting in elevated ceramide levels. Altogether, these results suggest that nSMase2 traffics from the Golgi to the PM as a membrane protein en route to the cell surface and recycles back to the Golgi through the endosomal/recycling compartment. Moreover, the recycling of nSMase2 from the PM is important for its catalytic regulation.
Collapse
Affiliation(s)
- Delphine Milhas
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
44
|
Mendoza-Macías CL, Barrios-Ceballos MP, Anaya-Velázquez F, Nakada-Tsukui K, Nozaki T, Padilla-Vaca F. Entamoeba histolytica: Molecular cloning and characterization of a novel neutral sphingomyelinase. Exp Parasitol 2010; 125:279-85. [DOI: 10.1016/j.exppara.2010.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 01/28/2010] [Accepted: 02/01/2010] [Indexed: 01/21/2023]
|
45
|
Wu BX, Clarke CJ, Hannun YA. Mammalian neutral sphingomyelinases: regulation and roles in cell signaling responses. Neuromolecular Med 2010; 12:320-30. [PMID: 20552297 DOI: 10.1007/s12017-010-8120-z] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 06/02/2010] [Indexed: 12/22/2022]
Abstract
Ceramide, a bioactive lipid, has been extensively studied and identified as an essential bioactive molecule in mediating cellular signaling pathways. Sphingomyelinase (SMase), (EC 3.1.4.12) catalyzes the cleavage of the phosphodiester bond in sphingomyelin (SM) to form ceramide and phosphocholine. In mammals, three Mg(2+)-dependent neutral SMases termed nSMase1, nSMase2 and nSMase3 have been identified. Among the three enzymes, nSMase2 is the most studied and has been implicated in multiple physiological responses including cell growth arrest, apoptosis, development and inflammation. In this review, we summarize recent findings for the cloned nSMases and discuss the insights for their roles in regulation ceramide metabolism and cellular signaling pathway.
Collapse
Affiliation(s)
- Bill X Wu
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 29425, USA
| | | | | |
Collapse
|
46
|
Wu BX, Rajagopalan V, Roddy PL, Clarke CJ, Hannun YA. Identification and characterization of murine mitochondria-associated neutral sphingomyelinase (MA-nSMase), the mammalian sphingomyelin phosphodiesterase 5. J Biol Chem 2010; 285:17993-8002. [PMID: 20378533 DOI: 10.1074/jbc.m110.102988] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sphingolipids play important roles in regulating cellular responses. Although mitochondria contain sphingolipids, direct regulation of their levels in mitochondria or mitochondria-associated membranes is mostly unclear. Neutral SMase (N-SMase) isoforms, which catalyze hydrolysis of sphingomyelin (SM) to ceramide and phosphocholine, have been found in the mitochondria of yeast and zebrafish, yet their existence in mammalian mitochondria remains unknown. Here, we have identified and cloned a cDNA based on nSMase homologous sequences. This cDNA encodes a novel protein of 483 amino acids that displays significant homology to nSMase2 and possesses the same catalytic core residues as members of the extended N-SMase family. A transiently expressed V5-tagged protein co-localized with both mitochondria and endoplasmic reticulum markers in MCF-7 and HEK293 cells; accordingly, the enzyme is referred to as mitochondria-associated nSMase (MA-nSMase). MA-nSMase was highly expressed in testis, pancreas, epididymis, and brain. MA-nSMase had an absolute requirement for cations such as Mg(2+) and Mn(2+) and activation by the anionic phospholipids, especially phosphatidylserine and the mitochondrial cardiolipin. Importantly, overexpression of MA-nSMase in HEK293 cells significantly increased in vitro N-SMase activity and also modulated the levels of SM and ceramide, indicating that the identified cDNA encodes a functional SMase. Thus, these studies identify and characterize, for the first time, a mammalian MA-nSMase. The characterization of MA-nSMase described here will contribute to our understanding of pathways regulated by sphingolipid metabolites, particularly with reference to the mitochondria and associated organelles.
Collapse
Affiliation(s)
- Bill X Wu
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
47
|
He Q, Shi J, Sun H, An J, Huang Y, Sheikh MS. Characterization of Human Homeodomain-interacting Protein Kinase 4 (HIPK4) as a Unique Member of the HIPK Family. MOLECULAR AND CELLULAR PHARMACOLOGY 2010; 2:61-68. [PMID: 20508833 PMCID: PMC2876313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Homeodomain-interacting protein kinases including HIPK1, HIPK2 and HIPK3 are serine/threonine kinases that form a family of highly conserved kinases. HIPKs are involved in diverse cellular functions including regulation cell death, survival, proliferation and differentiation. Here we report the characterization of a human HIPK4 that we identified in a proteomic screen during our efforts to unravel novel markers linked to cell death and survival. Human HIPK4 protein is composed of 616 residues with predicted molecular mass of 69.425 kDa and harbors a serine/threonine protein kinase catalytic domain at its N-terminal end. In the in vitro kinase assay, HIPK4 exhibits kinase activity and mutation of the conserved lysine 40 or aspartic acid 136 residue in its catalytic domain inactivates its kinase function. Human HIPK4 harbors multiple putative serine/threonine- and tyrosine-specific phsophorylation sites and also contains four high probability sumoylation sites, findings that suggest its function to be modulated by post-translational modifications. HIPK4 has been so named in the database because of its sequence homology to HIPK1, 2 and 3 predominantly within its catalytic domain. However, HIPK4 is smaller in size than the known HIPKs and has additional distinct features suggesting it to be a unique member of the HIPK family. Further functional characterization of HIPK4 is needed and will prove valuable to ascertain whether it performs distinct functions or share overlapping functions with other HIPKs.
Collapse
Affiliation(s)
- Qin He
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York
| | | | | | | | | | | |
Collapse
|
48
|
Gault CR, Obeid LM, Hannun YA. An overview of sphingolipid metabolism: from synthesis to breakdown. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 688:1-23. [PMID: 20919643 DOI: 10.1007/978-1-4419-6741-1_1] [Citation(s) in RCA: 770] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sphingolipids constitute a class of lipids defined by their eighteen carbon amino-alcohol backbones which are synthesized in the ER from nonsphingolipid precursors. Modification of this basic structure is what gives rise to the vast family of sphingolipids that play significant roles in membrane biology and provide many bioactive metabolites that regulate cell function. Despite the diversity of structure and function of sphingolipids, their creation and destruction are governed by common synthetic and catabolic pathways. In this regard, sphingolipid metabolism can be imagined as an array of interconnected networks that diverge from a single common entry point and converge into a single common breakdown pathway. In their simplest forms, sphingosine, phytosphingosine and dihydrosphingosine serve as the backbones upon which further complexity is achieved. For example, phosphorylation of the C1 hydroxyl group yields the final breakdown products and/or the important signaling molecules sphingosine-1-phosphate, phytosphingosine-1-phosphate and dihydrosphingosine-1-phosphate, respectively. On the other hand, acylation of sphingosine, phytosphingosine, or dihydrosphingosine with one of several possible acyl CoA molecules through the action of distinct ceramide synthases produces the molecules defined as ceramide, phytoceramide, or dihydroceramide. Ceramide, due to the differing acyl CoAs that can be used to produce it, is technically a class of molecules rather than a single molecule and therefore may have different biological functions depending on the acyl chain it is composed of. At the apex of complexity is the group of lipids known as glycosphingolipids (GSL) which contain dozens of different sphingolipid species differing by both the order and type of sugar residues attached to their headgroups. Since these molecules are produced from ceramide precursors, they too may have differences in their acyl chain composition, revealing an additional layer of variation. The glycosphingolipids are divided broadly into two categories: glucosphingolipids and galactosphingolipids. The glucosphingolipids depend initially on the enzyme glucosylceramide synthase (GCS) which attaches glucose as the first residue to the C1 hydroxyl position. Galactosphingolipids, on the other hand, are generated from galactosylceramide synthase (GalCerS), an evolutionarily dissimilar enzyme from GCS. Glycosphingolipids are further divided based upon further modification by various glycosyltransferases which increases the potential variation in lipid species by several fold. Far more abundant are the sphingomyelin species which are produced in parallel with glycosphingolipids, however they are defined by a phosphocholine headgroup rather than the addition of sugar residues. Although sphingomyelin species all share a common headgroup, they too are produced from a variety of ceramide species and therefore can have differing acyl chains attached to their C-2 amino groups. Whether or not the differing acyl chain lengths in SMs dictate unique functions or important biophysical distinctions has not yet been established. Understanding the function of all the existing glycosphingolipids and sphingomyelin species will be a major undertaking in the future since the tools to study and measure these species are only beginning to be developed (see Fig 1 for an illustrated depiction of the various sphingolipid structures). The simple sphingolipids serve both as the precursors and the breakdown products of the more complex ones. Importantly, in recent decades, these simple sphingolipids have gained attention for having significant signaling and regulatory roles within cells. In addition, many tools have emerged to measure the levels of simple sphingolipids and therefore have become the focus of even more intense study in recent years. With this thought in mind, this chapter will pay tribute to the complex sphingolipids, but focus on the regulation of simple sphingolipid metabolism.
Collapse
Affiliation(s)
- Christopher R Gault
- Department of Biochemistry, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | |
Collapse
|
49
|
Ito H, Murakami M, Furuhata A, Gao S, Yoshida K, Sobue S, Hagiwara K, Takagi A, Kojima T, Suzuki M, Banno Y, Tanaka K, Tamiya-Koizumi K, Kyogashima M, Nozawa Y, Murate T. Transcriptional regulation of neutral sphingomyelinase 2 gene expression of a human breast cancer cell line, MCF-7, induced by the anti-cancer drug, daunorubicin. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2009; 1789:681-90. [DOI: 10.1016/j.bbagrm.2009.08.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2009] [Revised: 08/08/2009] [Accepted: 08/10/2009] [Indexed: 11/29/2022]
|
50
|
Milhas D, Clarke CJ, Hannun YA. Sphingomyelin metabolism at the plasma membrane: implications for bioactive sphingolipids. FEBS Lett 2009; 584:1887-94. [PMID: 19857494 DOI: 10.1016/j.febslet.2009.10.058] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 10/19/2009] [Accepted: 10/20/2009] [Indexed: 12/24/2022]
Abstract
The plasma membrane (PM) is a major resource for production of bioactive lipids and contains a large proportion of the cellular sphingomyelin (SM) content. Consequently, the regulation of SM levels at the PM by enzymes such as sphingomyelinase (SMase) and SM synthase 2 (SMS2) can have profound effects - both on biophysical properties of the membrane, but also on cellular signaling. Over the past 20 years, there has been considerable research into the physiological and cellular functions associated with regulation of SM levels, notably with regards to the production of ceramide. In this review, we will summarize this research with particular focus on the SMases and SMS2. We will outline what biological functions are associated with SM metabolism/production at the PM, and discuss what we believe are major challenges that need to be addressed in future studies.
Collapse
Affiliation(s)
- Delphine Milhas
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|