Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell ; 177(6): 1536-1552.e23, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150623

ABSTRACT

Ectopic lipid deposition and altered mitochondrial dynamics contribute to the development of obesity and insulin resistance. However, the mechanistic link between these processes remained unclear. Here we demonstrate that the C16:0 sphingolipid synthesizing ceramide synthases, CerS5 and CerS6, affect distinct sphingolipid pools and that abrogation of CerS6 but not of CerS5 protects from obesity and insulin resistance. We identify proteins that specifically interact with C16:0 sphingolipids derived from CerS5 or CerS6. Here, only CerS6-derived C16:0 sphingolipids bind the mitochondrial fission factor (Mff). CerS6 and Mff deficiency protect from fatty acid-induced mitochondrial fragmentation in vitro, and the two proteins genetically interact in vivo in obesity-induced mitochondrial fragmentation and development of insulin resistance. Our experiments reveal an unprecedented specificity of sphingolipid signaling depending on specific synthesizing enzymes, provide a mechanistic link between hepatic lipid deposition and mitochondrial fragmentation in obesity, and define the CerS6-derived sphingolipid/Mff interaction as a therapeutic target for metabolic diseases.


Subject(s)
Membrane Proteins/metabolism , Obesity/metabolism , Sphingolipids/metabolism , Sphingosine N-Acyltransferase/metabolism , Animals , Apoptosis , Cell Line , HeLa Cells , Humans , Insulin Resistance/physiology , Liver/metabolism , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Obesity/physiopathology , Sphingolipids/physiology , Sphingosine N-Acyltransferase/physiology
2.
Adv Exp Med Biol ; 1159: 49-63, 2019.
Article in English | MEDLINE | ID: mdl-31502199

ABSTRACT

The majority of enzymes in the sphingolipid (SL) biosynthetic pathway have been identified over the past couple of decades. Despite significant work, and despite their crucial and central roles in SL synthesis, significant information is still lacking concerning the enzymes that catalyze the N-acylation of sphingoid long chain bases, namely the ceramide synthases (CerS), a family of six mammalian genes originally named longevity assurance (Lass) genes. Each of these six endoplasmic reticulum (ER) membrane-bound enzymes utilizes a relatively restricted sub-set of fatty acyl-CoAs for N-acylation, but are far more promiscuous about the use of long chain bases. The reason that mammals and other species have multiple CerS, generating a specific subset of ceramides, is not yet known, but implies an important role for ceramides containing specific fatty acids in cell physiology. In this brief chapter, we will stroll down the CerS lane and discuss what is known, and what is not known, about this important enzyme family.


Subject(s)
Ceramides/biosynthesis , Endoplasmic Reticulum/enzymology , Sphingosine N-Acyltransferase/physiology , Animals , Fatty Acids/chemistry , Sphingolipids
3.
Hum Mol Genet ; 21(3): 586-608, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22038835

ABSTRACT

The stratum corneum as the outermost epidermal layer protects against exsiccation and infection. Both the underlying cornified envelope (CE) and the intercellular lipid matrix contribute essentially to these two main protective barriers. Epidermis-unique ceramides with ultra-long-chain acyl moities (ULC-Cers) are key components of extracellular lipid lamellae (ELL) and are bound to CE proteins, thereby contributing to the cornified lipid envelope (CLE). Here, we identified human and mouse ceramide synthase 3 (CerS3), among CerS1-6, to be exclusively required for the ULC-Cer synthesis in vitro and of mouse CerS3 in vivo. Deficiency of CerS3 in mice results in complete loss of ULC-Cers (≥C26), lack of continuous ELL and a non-functional CLE. Consequently, newborn mutant mice die shortly after birth from transepidermal water loss. Mutant skin is prone to Candida albicans infection highlighting ULC-Cers to be pivotal for both barrier functions. Persistent periderm, hyperkeratosis and deficient cornification are hallmarks of mutant skin demonstrating loss of Cers to trigger a keratinocyte maturation arrest at an embryonic pre-barrier stage.


Subject(s)
Skin Physiological Phenomena , Sphingosine N-Acyltransferase/physiology , Animals , Animals, Newborn , Candida albicans/physiology , Cell Membrane/ultrastructure , Ceramides/analysis , Ceramides/chemistry , Ceramides/metabolism , Epidermal Cells , Epidermis/embryology , Epidermis/enzymology , Fatty Acids/metabolism , Genes, Lethal , HEK293 Cells , HeLa Cells , Humans , Keratinocytes/cytology , Mice , Skin/microbiology , Sphingosine N-Acyltransferase/deficiency , Sphingosine N-Acyltransferase/genetics , Water Loss, Insensible
4.
J Immunol ; 188(11): 5723-33, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22544924

ABSTRACT

Ceramides are mediators of apoptosis and inflammatory processes. In an animal model of multiple sclerosis (MS), the experimental autoimmune encephalomyelitis (EAE) model, we observed a significant elevation of C(16:0)-Cer in the lumbar spinal cord of EAE mice. This was caused by a transiently increased expression of ceramide synthase (CerS) 6 in monocytes/macrophages and astroglia. Notably, this corresponds to the clinical finding that C(16:0)-Cer levels were increased 1.9-fold in cerebrospinal fluid of MS patients. NO and TNF-α secreted by IFN-γ-activated macrophages play an essential role in the development of MS. In murine peritoneal and mouse-derived RAW 264.7 macrophages, IFN-γ-mediated expression of inducible NO synthase (iNOS)/TNF-α and NO/TNF-α release depends on upregulation of CerS6/C(16:0)-Cer. Downregulation of CerS6 by RNA interference or endogenous upregulation of C(16:0)-Cer mediated by palmitic acid in RAW 264.7 macrophages led to a significant reduction or increase in NO/TNF-α release, respectively. EAE/IFN-γ knockout mice showed a significant delay in disease onset accompanied by a significantly less pronounced increase in CerS6/C(16:0)-Cer, iNOS, and TNF-α compared with EAE/wild-type mice. Treatment of EAE mice with l-cycloserine prevented the increase in C(16:0)-Cer and iNOS/TNF-α expression and caused a remission of the disease. In conclusion, CerS6 plays a critical role in the onset of MS, most likely by regulating NO and TNF-α synthesis. CerS6 may represent a new target for the inhibition of inflammatory processes promoting MS development.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Membrane Proteins/physiology , Sphingosine N-Acyltransferase/physiology , Adult , Aged , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/physiology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/cerebrospinal fluid , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nitric Oxide/metabolism , Sphingosine N-Acyltransferase/biosynthesis , Sphingosine N-Acyltransferase/cerebrospinal fluid , Young Adult
5.
Chin J Cancer ; 29(7): 697-702, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20591223

ABSTRACT

TMSG-1 is a newly discovered tumor metastasis suppressor gene, which plays important roles in promoting apoptosis and inhibiting invasion and metastasis of tumor cells. The inhibitory function of TMSG-1 in tumor cells may be related to vacuolar H+-ATPase and ceramide, but the underlying mechanism remains unknown. Studies on TMSG-1 are limited worldwide, and only a research group in Shanghai and our group have recently studied on it. As a new research field, the function of TMSG-1 remains to be explored. This review discusses the discovery of TMSG-1, structure of its encoded protein, its roles and possible mechanism in inhibiting tumor invasion and metastasis.


Subject(s)
Ceramides/pharmacology , Membrane Proteins/metabolism , Neoplasms , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Ceramides/chemical synthesis , Enzyme Activation , Humans , Membrane Proteins/physiology , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Sphingosine N-Acyltransferase/physiology , Tumor Suppressor Proteins/physiology
6.
J Invest Dermatol ; 140(10): 1927-1937.e5, 2020 10.
Article in English | MEDLINE | ID: mdl-32092351

ABSTRACT

Epidermal barrier dysfunction is associated with a wide range of highly prevalent inflammatory skin diseases. However, the molecular processes that drive epidermal barrier maintenance are still largely unknown. Here, using quantitative proteomics, lipidomics, and mouse genetics, we characterize epidermal barrier maintenance versus a newly established barrier and functionally identify differential ceramide synthase 4 protein expression as one key difference. We show that epidermal loss of ceramide synthase 4 first disturbs epidermal lipid metabolism and adult epidermal barrier function, ultimately resulting in chronic skin barrier disease characterized by acanthosis, hyperkeratosis, and immune cell accumulation. Importantly, prolonged barrier dysfunction induced by loss of ceramide synthase 4 induced a barrier repair response that largely recapitulates molecular programs of barrier establishment. Collectively, this study provides an unbiased temporal proteomic characterization of barrier maintenance and disturbed homeostasis and shows that lipid homeostasis is essential to maintain adult skin barrier function to prevent disease.


Subject(s)
Homeostasis/physiology , Skin/metabolism , Sphingosine N-Acyltransferase/physiology , Animals , Epidermis/metabolism , Lipid Metabolism , Mice , Mice, Inbred C57BL , Morphogenesis , Proteomics
7.
Sci Adv ; 5(9): eaax1978, 2019 09.
Article in English | MEDLINE | ID: mdl-31535025

ABSTRACT

How lipid metabolism is regulated at the outer mitochondrial membrane (OMM) for transducing stress signaling remains largely unknown. We show here that this process is controlled by trafficking of ceramide synthase 1 (CerS1) from the endoplasmic reticulum (ER) to the OMM by a previously uncharacterized p17, which is now renamed protein that mediates ER-mitochondria trafficking (PERMIT). Data revealed that p17/PERMIT associates with newly translated CerS1 on the ER surface to mediate its trafficking to the OMM. Cellular stress induces Drp1 nitrosylation/activation, releasing p17/PERMIT to retrieve CerS1 for its OMM trafficking, resulting in mitochondrial ceramide generation, mitophagy and cell death. In vivo, CRISPR-Cas9-dependent genetic ablation of p17/PERMIT prevents acute stress-mediated CerS1 trafficking to OMM, attenuating mitophagy in p17/PERMIT-/- mice, compared to controls, in various metabolically active tissues, including brain, muscle, and pancreas. Thus, these data have implications in diseases associated with accumulation of damaged mitochondria such as cancer and/or neurodegeneration.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/physiology , Mitochondria/pathology , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/physiology , Mitophagy , Sphingosine N-Acyltransferase/physiology , Stress, Physiological , Animals , CRISPR-Cas Systems , Ceramides/metabolism , Endoplasmic Reticulum/pathology , Humans , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Protein Transport
8.
Zhonghua Bing Li Xue Za Zhi ; 36(10): 672-6, 2007 Oct.
Article in Zh | MEDLINE | ID: mdl-18194600

ABSTRACT

OBJECTIVE: To investigate the effects of tumor metastasis suppressor gene 1 (TMSG-1) overexpression on the proliferation, invasion and apoptosis of breast cancer cells and to determine possible correlations of TMSG-1 and metastasis of breast cancer. METHODS: Full-length human TMSG-1 coding sequences were cloned into plasmid pcDNA3.0-FLAG. The recombinant plasmids constructs were transfeced into MDA-MB-231, a highly malignant breast cancer cell line. Parental, vector-only stable transfectant and TMSG-1 stable transfectant clones were tested by MTT, soft agar colony formation and Boyden chamber assays. At twenty-four hours and forty-eight hours post transient transfection, double staining with Annexin-V-FITC and PI were employed to distinguish apoptotic cells from living cells by flow cytometry analysis. RESULTS: Three TMSG-1 overexpression clones were selected. Compared with the control cells, TMSG-1 overexpression MDA-MB-231 cells showed strong inhibition of proliferation and decreased clonogenicity in soft agar (P<0.05). Transfection of TMSG-1 into MDA-MB-231 cells significantly suppressed the cell invasion ability in vitro (decreased numbers of cells trespassing the matrigel in three experiments: 72.3+/-8.1, 85.0+/-4.2, and 73.5+/-7.8) in comparison with nave cells without transfection (187.5+/-2.1) and cells transfected with the control vector (162.3+/-6.8) (P<0.01). Transient transfection of TMSG-1 into MDA-MB-231 cells could promote cell apoptosis at 24 and 48 hours after transfection (P<0.05). CONCLUSIONS: TMSG-1 protein may have multiple functions in the regulation of proliferation, invasion and apoptosis of metastatic breast cancer cells, likely as a metastasis suppressor gene.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Cell Proliferation , Membrane Proteins/metabolism , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Neoplasm Invasiveness , Plasmids , Recombinant Proteins/metabolism , Sphingosine N-Acyltransferase/genetics , Sphingosine N-Acyltransferase/physiology , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
9.
J Clin Invest ; 126(1): 254-65, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26650179

ABSTRACT

Sphingolipids make up a family of molecules associated with an array of biological functions, including cell death and migration. Sphingolipids are often altered in cancer, though how these alterations lead to tumor formation and progression is largely unknown. Here, we analyzed non-small-cell lung cancer (NSCLC) specimens and cell lines and determined that ceramide synthase 6 (CERS6) is markedly overexpressed compared with controls. Elevated CERS6 expression was due in part to reduction of microRNA-101 (miR-101) and was associated with increased invasion and poor prognosis. CERS6 knockdown in NSCLC cells altered the ceramide profile, resulting in decreased cell migration and invasion in vitro, and decreased the frequency of RAC1-positive lamellipodia formation while CERS6 overexpression promoted it. In murine models, CERS6 knockdown in transplanted NSCLC cells attenuated lung metastasis. Furthermore, combined treatment with l-α-dimyristoylphosphatidylcholine liposome and the glucosylceramide synthase inhibitor D-PDMP induced cell death in association with ceramide accumulation and promoted cancer cell apoptosis and tumor regression in murine models. Together, these results indicate that CERS6-dependent ceramide synthesis and maintenance of ceramide in the cellular membrane are essential for lamellipodia formation and metastasis. Moreover, these results suggest that targeting this homeostasis has potential as a therapeutic strategy for CERS6-overexpressing NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Membrane Proteins/physiology , Sphingosine N-Acyltransferase/physiology , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Ceramides/metabolism , Dimyristoylphosphatidylcholine/pharmacology , Humans , Lung Neoplasms/drug therapy , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , MicroRNAs/physiology , Neoplasm Metastasis , Phenotype , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics
10.
Oncogene ; 32(13): 1682-90, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-22580606

ABSTRACT

A main obstacle to overcome during the treatment of tumors is drug resistance to chemotherapy; emerging studies indicate that a key factor contributing to this problem is the acidic tumor microenvironment. Here, we found that LASS2 expression was significantly lower in drug-resistant Michigan Cancer Foundation-7/adriamycin (MCF-7/ADR) human breast cancer cells than the drug-sensitive MCF-7 cells, and low expression of LASS2 was associated with poor prognosis in patients with breast cancer. Our results showed that the overexpression of LASS2 in MCF-7/ADR cells increased the chemosensitivity to multiple chemotherapeutic agents, including doxorubicin (Dox), whereas LASS2 knockdown in MCF-7 cells decreased the chemosensitivity. Cell-cycle analysis revealed a corresponding increase in apoptosis in the LASS2-overexpressing cells following Dox exposure, showing that the overexpression of LASS2 increased the susceptibility to Dox cytotoxicity. This effect was mediated by a significant increase in pHe (extracellular pH) and lysosomal pH, and more Dox entered the cells and stayed in the nuclei of cells. In nude mice, the combination of LASS2 overexpression and Dox significantly inhibited the growth of xenografts. Our findings suggest that LASS2 is involved in chemotherapeutic outcomes and low LASS2 expression may predict chemoresistance.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Drug Resistance, Neoplasm/genetics , Membrane Proteins/physiology , Sphingosine N-Acyltransferase/physiology , Tumor Microenvironment , Tumor Suppressor Proteins/physiology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Acids/antagonists & inhibitors , Acids/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma/diagnosis , Carcinoma/drug therapy , Carcinoma/pathology , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Hydrogen-Ion Concentration/drug effects , Membrane Proteins/genetics , Mice , Mice, Nude , Prognosis , Sphingosine N-Acyltransferase/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Suppressor Proteins/genetics , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Xenograft Model Antitumor Assays
11.
Cell Signal ; 22(9): 1300-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20406683

ABSTRACT

The role of ceramide neo-genesis in cellular stress response signaling is gaining increasing attention with recent progress in elucidating the novel roles and biochemical properties of the ceramide synthase (CerS) enzymes. Selective tissue and subcellular distribution of the six mammalian CerS isoforms, combined with distinct fatty acyl chain length substrate preferences, implicate differential functions of specific ceramide species in cellular signaling. We report here that ionizing radiation (IR) induces de novo synthesis of ceramide to influence HeLa cell apoptosis by specifically activating CerS isoforms 2, 5, and 6 that generate opposing anti- and pro-apoptotic ceramides in mitochondrial membranes. Overexpression of CerS2 resulted in partial protection from IR-induced apoptosis whereas overexpression of CerS5 increased apoptosis in HeLa cells. Knockdown studies determined that CerS2 is responsible for all observable IR-induced C(24:0) CerS activity, and while CerS5 and CerS6 each confer approximately 50% of the C(16:0) CerS baseline synthetic activity, both are required for IR-induced activity. Additionally, co-immunoprecipitation studies suggest that CerS2, 5, and 6 might exist as heterocomplexes in HeLa cells, providing further insight into the regulation of CerS proteins. These data add to the growing body of evidence demonstrating interplay among the CerS proteins in a stress stimulus-, cell type- and subcellular compartment-specific manner.


Subject(s)
Apoptosis , Membrane Proteins/metabolism , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Acyl Coenzyme A/metabolism , Ceramides/biosynthesis , HeLa Cells , Humans , Isoenzymes/metabolism , Membrane Proteins/physiology , Mitochondria/metabolism , Mitochondria/radiation effects , Palmitoyl Coenzyme A/metabolism , Radiation, Ionizing , Sphingosine N-Acyltransferase/physiology , Tumor Suppressor Proteins/physiology
12.
Biochem Pharmacol ; 80(11): 1632-40, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20735991

ABSTRACT

Ceramides serve as bioactive molecules with important roles in cell proliferation and apoptosis. Ceramides (Cer) with different N-acyl side chains (C(14:0)-Cer-C(26:0)-Cer) possess distinctive roles in cell signaling and are differentially expressed in HCT-116 colon cancer cells. Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, exhibiting antiproliferative effects, activates the sphingolipid pathway. To elucidate the mechanism, HCT-116 cells were treated with 50µM celecoxib leading to a significant increase of C(16:0)-Cer. Interestingly, 50µM celecoxib resulted in a 2.8-fold increase of ceramide synthase (CerS) activity as measured by a cell-based activity assay. siRNA against several CerSs revealed that CerS6 was predominantly responsible for the increase of C(16:0)-Cer in HCT-116 cells. Moreover, the silencing of CerS6 partially protected HCT-116 cells from the toxic effects induced by celecoxib. Treatment of cells with celecoxib and fumonisin B1 (inhibitor of CerSs) or myriocin (inhibitor of l-serine palmitoyl transferase) or desipramine (inhibitor of acid sphingomyelinase and acid ceramidase) revealed that the increase of C(16:0)-Cer results predominantly from activation of the salvage pathway. Using the nude mouse model we demonstrated that celecoxib induces also in vivo a significant increase of C(16:0)-Cer in stomach, small intestine and tumor tissue. In conclusion, celecoxib causes a specific increase of C(16:0)-Cer by activating CerS6 and the salvage pathway, which contribute to the toxic effects of celecoxib.


Subject(s)
Ceramides/biosynthesis , Membrane Proteins/metabolism , Pyrazoles/pharmacology , Sphingosine N-Acyltransferase/metabolism , Sulfonamides/pharmacology , Animals , Celecoxib , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , HCT116 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Nude , Pyrazoles/toxicity , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/physiology , Sulfonamides/toxicity , Tissue Distribution/drug effects , Tissue Distribution/physiology
SELECTION OF CITATIONS
SEARCH DETAIL