Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Metab ; 36(7): 1521-1533.e5, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38718792

ABSTRACT

Diabetic retinopathy is a microvascular disease that causes blindness. Using acid sphingomyelinase knockout mice, we reported that ceramide generation is critical for diabetic retinopathy development. Here, in patients with proliferative diabetic retinopathy, we identify vitreous ceramide imbalance with pathologic long-chain C16-ceramides increasing and protective very long-chain C26-ceramides decreasing. C16-ceramides generate pro-inflammatory/pro-apoptotic ceramide-rich platforms on endothelial surfaces. To geo-localize ceramide-rich platforms, we invented a three-dimensional confocal assay and showed that retinopathy-producing cytokines TNFα and IL-1ß induce ceramide-rich platform formation on retinal endothelial cells within seconds, with volumes increasing 2-logs, yielding apoptotic death. Anti-ceramide antibodies abolish these events. Furthermore, intravitreal and systemic anti-ceramide antibodies protect from diabetic retinopathy in standardized rodent ischemia reperfusion and streptozotocin models. These data support (1) retinal endothelial ceramide as a diabetic retinopathy treatment target, (2) early-stage therapy of non-proliferative diabetic retinopathy to prevent progression, and (3) systemic diabetic retinopathy treatment; and they characterize diabetic retinopathy as a "ceramidopathy" reversible by anti-ceramide immunotherapy.


Subject(s)
Ceramides , Diabetic Retinopathy , Immunotherapy , Ceramides/metabolism , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/pathology , Diabetic Retinopathy/immunology , Animals , Humans , Mice , Endothelial Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Male , Retina/metabolism , Retina/pathology , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Rats , Apoptosis/drug effects , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/metabolism , Vitreous Body/metabolism , Female , Mice, Knockout
2.
Proc Natl Acad Sci U S A ; 120(34): e2220269120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37579172

ABSTRACT

The vascular endothelium from individual organs is functionally specialized, and it displays a unique set of accessible molecular targets. These serve as endothelial cell receptors to affinity ligands. To date, all identified vascular receptors have been proteins. Here, we show that an endothelial lung-homing peptide (CGSPGWVRC) interacts with C16-ceramide, a bioactive sphingolipid that mediates several biological functions. Upon binding to cell surfaces, CGSPGWVRC triggers ceramide-rich platform formation, activates acid sphingomyelinase and ceramide production, without the associated downstream apoptotic signaling. We also show that the lung selectivity of CGSPGWVRC homing peptide is dependent on ceramide production in vivo. Finally, we demonstrate two potential applications for this lipid vascular targeting system: i) as a bioinorganic hydrogel for pulmonary imaging and ii) as a ligand-directed lung immunization tool against COVID-19. Thus, C16-ceramide is a unique example of a lipid-based receptor system in the lung vascular endothelium targeted in vivo by circulating ligands such as CGSPGWVRC.


Subject(s)
COVID-19 , Humans , Ligands , COVID-19/metabolism , Ceramides/metabolism , Lung/metabolism , Endothelium, Vascular/metabolism , Receptors, Cell Surface/metabolism , Carrier Proteins/metabolism , Sphingomyelin Phosphodiesterase/metabolism
3.
J Cell Biol ; 219(4)2020 04 06.
Article in English | MEDLINE | ID: mdl-32328634

ABSTRACT

Diverse stresses, including reactive oxygen species (ROS), ionizing radiation, and chemotherapies, activate acid sphingomyelinase (ASMase) and generate the second messenger ceramide at plasma membranes, triggering apoptosis in specific cells, such as hematopoietic cells and endothelium. Ceramide elevation drives local bilayer reorganization into ceramide-rich platforms, macrodomains (0.5-5-µm diameter) that transmit apoptotic signals. An unresolved issue is how ASMase residing within lysosomes is released extracellularly within seconds to hydrolyze sphingomyelin preferentially enriched in outer plasma membranes. Here we show that physical damage by ionizing radiation and ROS induces full-thickness membrane disruption that allows local calcium influx, membrane lysosome fusion, and ASMase release. Further, electron microscopy reveals that plasma membrane "nanopore-like" structures (∼100-nm diameter) form rapidly due to lipid peroxidation, allowing calcium entry to initiate lysosome fusion. We posit that the extent of upstream damage to mammalian plasma membranes, calibrated by severity of nanopore-mediated local calcium influx for lysosome fusion, represents a biophysical mechanism for cell death induction.


Subject(s)
Apoptosis , Cell Membrane/metabolism , Lysosomes/metabolism , Carbon Radioisotopes , Humans , Jurkat Cells , Reactive Oxygen Species/metabolism , Sphingomyelins/chemistry
4.
Cancer Res ; 80(5): 1219-1227, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31690670

ABSTRACT

Tissue survival responses to ionizing radiation are nonlinear with dose, rather yielding tissue-specific descending curves that impede straightforward analysis of biologic effects. Apoptotic cell death often occurs at low doses, while at clinically relevant intermediate doses, double-strand break misrepair yields mitotic death that determines outcome. As researchers frequently use a single low dose for experimentation, such strategies may inaccurately depict inherent tissue responses. Cutting edge radiobiology has adopted full dose survival profiling and devised mathematical algorithms to fit curves to observed data to generate highly reproducible numerical data that accurately define clinically relevant inherent radiosensitivities. Here, we established a protocol for irradiating organoids that delivers radiation profiles simulating the organ of origin. This technique yielded highly similar dose-survival curves of small and large intestinal crypts in vivo and their cognate organoids analyzed by the single-hit multi-target (SHMT) algorithm, outcomes reflecting the inherent radiation profile of their respective Lgr5+ stem cell populations. As this technological advance is quantitative, it will be useful for accurate evaluation of intestinal (patho)physiology and drug screening. SIGNIFICANCE: These findings establish standards for irradiating organoids that deliver radiation profiles that phenocopy the organ of origin.See related commentary by Muschel et al., p. 927.


Subject(s)
Organoids , Stem Cells , Intestines , Radiation Tolerance , Radiation, Ionizing
5.
Nat Med ; 25(10): 1607-1614, 2019 10.
Article in English | MEDLINE | ID: mdl-31591597

ABSTRACT

Rectal cancer (RC) is a challenging disease to treat that requires chemotherapy, radiation and surgery to optimize outcomes for individual patients. No accurate model of RC exists to answer fundamental research questions relevant to patients. We established a biorepository of 65 patient-derived RC organoid cultures (tumoroids) from patients with primary, metastatic or recurrent disease. RC tumoroids retained molecular features of the tumors from which they were derived, and their ex vivo responses to clinically relevant chemotherapy and radiation treatment correlated with the clinical responses noted in individual patients' tumors. Upon engraftment into murine rectal mucosa, human RC tumoroids gave rise to invasive RC followed by metastasis to lung and liver. Importantly, engrafted tumors displayed the heterogenous sensitivity to chemotherapy observed clinically. Thus, the biology and drug sensitivity of RC clinical isolates can be efficiently interrogated using an organoid-based, ex vivo platform coupled with in vivo endoluminal propagation in animals.


Subject(s)
Chemoradiotherapy , Organoids/pathology , Rectal Neoplasms/drug therapy , Rectal Neoplasms/radiotherapy , Animals , Fluorouracil/pharmacology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/radiotherapy , Liver Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Lung Neoplasms/secondary , Mice , Neoplasm Metastasis , Organoids/drug effects , Organoids/radiation effects , Rectal Neoplasms/pathology
6.
Front Immunol ; 10: 1386, 2019.
Article in English | MEDLINE | ID: mdl-31275322

ABSTRACT

Inflammatory diseases of the gastrointestinal tract are emerging as a global problem with increased evidence and prevalence in numerous countries. A dysregulated sphingolipid metabolism occurs in patients with ulcerative colitis and is discussed to contribute to its pathogenesis. In the present study, we determined the impact of acid sphingomyelinase (Asm), which catalyzes the hydrolysis of sphingomyelin to ceramide, on the course of Citrobacter (C.) rodentium-driven colitis. C. rodentium is an enteric pathogen and induces colonic inflammation very similar to the pathology in patients with ulcerative colitis. We found that mice with Asm deficiency or Asm inhibition were strongly susceptible to C. rodentium infection. These mice showed increased levels of C. rodentium in the feces and were prone to bacterial spreading to the systemic organs. In addition, mice lacking Asm activity showed an uncontrolled inflammatory Th1 and Th17 response, which was accompanied by a stronger colonic pathology compared to infected wild type mice. These findings identified Asm as an essential regulator of mucosal immunity to the enteric pathogen C. rodentium.


Subject(s)
Colitis/etiology , Colitis/metabolism , Disease Susceptibility , Host-Pathogen Interactions , Sphingomyelin Phosphodiesterase/metabolism , Amitriptyline/pharmacology , Animals , Biomarkers , Citrobacter rodentium/immunology , Colitis/pathology , Disease Models, Animal , Disease Resistance/immunology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Enzyme Activation/drug effects , Host-Pathogen Interactions/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , T-Lymphocyte Subsets/metabolism
7.
Cell Signal ; 38: 134-145, 2017 10.
Article in English | MEDLINE | ID: mdl-28687494

ABSTRACT

The cancer multidrug resistance (MDR) phenotype encompasses a myriad of molecular, genetic and cellular alterations resulting from progressive oncogenic transformation and selection. Drug efflux transporters, in particular the MDR P-glycoprotein ABCB1, play an important role in MDR but cannot confer the complete phenotype alone indicating parallel alterations are prerequisite. Sphingolipids are essential constituents of lipid raft domains and directly participate in functionalization of transmembrane proteins, including providing an optimal lipid microenvironment for multidrug transporters, and are also perturbed in cancer. Here we postulate that increased sphingomyelin content, developing early in some cancers, recruits and functionalizes plasma membrane ABCB1 conferring a state of partial MDR, which is completed by glycosphingolipid disturbance and the appearance of intracellular vesicular ABCB1. In this review, the independent and interdependent roles of sphingolipid alterations and ABCB1 upregulation during the transformation process and resultant conferment of partial and complete MDR phenotypes are discussed.


Subject(s)
Drug Resistance, Multiple , Drug Resistance, Neoplasm , Neoplasms/metabolism , Sphingolipids/metabolism , Humans , Membrane Microdomains/metabolism , Phenotype , Sphingolipids/biosynthesis
8.
Biochim Biophys Acta ; 1851(11): 1482-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26253821

ABSTRACT

The survival of macrophages depends on the presence of specific cytokines that activate survival signaling events, as well as suppressing formation of apoptosis-inducing pathways. We have previously shown that macrophages deprived of macrophage colony stimulating factor (M-CSF) produce ceramide that contributes to apoptosis of these cells, a pathway that is suppressed by exposure to oxidized LDL. In this study we have examined macrophages derived from mice lacking acid sphingomyelinase (ASMase) to ask whether these events are altered due to the impaired ability of these cells to break down sphingomyelin and produce ceramide. We found that these cells do survive better than cells from wild type mice, but they still undergo cell death and some ceramide is formed. We show that the ceramide is being produced by a de novo synthetic pathway. Therefore, ceramide production in M-CSF-deprived macrophages arises from a combination of ASMase activity and de novo synthesis.


Subject(s)
Ceramides/biosynthesis , Macrophages/metabolism , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelins/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Female , Lipoproteins, LDL/pharmacology , Macrophage Colony-Stimulating Factor/deficiency , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Signal Transduction , Sphingomyelin Phosphodiesterase/deficiency
10.
Radiother Oncol ; 111(1): 88-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24794795

ABSTRACT

BACKGROUND AND PURPOSE: Single Dose Radiation Therapy (SDRT) provides remarkably high rates of control even for tumors resistant to fractionated radiotherapy. SDRT tumor control depends on acute acid sphingomyelinase-mediated endothelial cell injury and monoclonal antibodies targeting Vascular Endothelial Cell Growth Factor (VEGF) signaling radiosensitized tumor endothelium when delivered immediately prior to irradiation. Here we evaluate the ability of the oral VEGF receptor inhibitor, axitinib, to sensitize tumor endothelium and increase tumor control with SDRT. METHODS AND MATERIALS: Axitinib was added to primary cultured endothelial cells, or administered orally to Sv129/BL6 mice bearing radiosensitive MCA/129 sarcoma or radioresistant B16F1 melanoma flank tumors, followed by SDRT. Endothelial apoptosis was assessed by TUNEL assay or bis-benzamide staining. Mice with irradiated tumors were followed for 90days to evaluate the impact of axitinib on SDRT tumor control. RESULTS: Pre-treatment with axitinib increased acute endothelial cell apoptosis following SDRT in vitro, and in vivo for both MCA/129 and B16F1 tumors. Axitinib correspondingly increased SDRT tumor growth delay and complete response rate (by 40%) for both tumors. Administration precisely 1h before SDRT was critical for radiosensitization. CONCLUSIONS: Axitinib radiosensitizes tumor endothelial cells and enhances tumor cure with SDRT, which may permit dose de-escalation and significantly expand the range of clinical indications for SDRT.


Subject(s)
Imidazoles/pharmacology , Indazoles/pharmacology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/radiotherapy , Animals , Apoptosis/radiation effects , Axitinib , Endothelial Cells/drug effects , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Protein Kinase Inhibitors/pharmacology , Radiation Tolerance/drug effects , Radiotherapy Dosage , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Signal Transduction/drug effects
11.
Cancer Cell ; 24(3): 279-81, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-24029224

ABSTRACT

Cancer cells display lysosome hypertrophy, secreting lysosomal hydrolases for tumor progression. Hypertrophy renders lysosomes fragile, increasing lysosomal membrane permeabilization (LMP) tendency. In this issue of Cancer Cell, Petersen and colleagues show that lysosomal sphingomyelin content determines LMP and cationic drugs displace acid sphingomyelinase from lysosomal membranes, increasing tumor LMP and death.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Enzyme Inhibitors/pharmacology , Lysosomes/metabolism , Sphingolipids/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Animals , Female , Humans
12.
Radiat Res ; 176(4): 434-46, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21631289

ABSTRACT

The sphingomyelin pathway involves the enzymatic cleavage of sphingomyelin to produce ceramide, a second messenger that serves as a key mediator in the rapid apoptotic response to various cell stressors. Low-linear energy transfer (LET) γ radiation can initiate this pathway, independent of DNA damage, via the cell membrane. Whether short-ranged, high-LET α particles, which are of interest as potent environmental carcinogens, radiotherapies and potential components of dirty bombs, can act through this mechanism to signal apoptosis is unknown. Here we show that irradiation of Jurkat cells with α particles emitted by the ²²5Ac-DOTA-anti-CD3 IgG antibody construct results in dose-dependent apoptosis. This apoptosis was significantly reduced by pretreating cells with cholesterol-depleting nystatin, a reagent known to inhibit ceramide signaling by interfering with membrane raft coalescence and ceramide-rich platform generation. The effects of nystatin on α-particle-induced apoptosis were related to disruption of the ceramide pathway and not to microdosimetry alterations, because similar results were obtained after external irradiation of the cells with a broad beam of collimated α particles using a planar ²4¹Am source. External irradiation allowed for more precise control of the dosimetry and geometry of the irradiation, independent of antibody binding or cell internalization kinetics. Mechanistically consistent with these findings, Jurkat cells rapidly increased membrane concentrations of ceramide after external irradiation with an average of five α-particle traversals per cell. These data indicate that α particles can activate the sphingomyelin pathway to induce apoptosis.


Subject(s)
Alpha Particles/adverse effects , Apoptosis/radiation effects , Sphingomyelins/metabolism , Actinium/adverse effects , Apoptosis/drug effects , Humans , Immunoglobulin G/metabolism , Isotope Labeling , Jurkat Cells , Nystatin/pharmacology , Radiometry
13.
FEBS Lett ; 581(27): 5289-94, 2007 Nov 13.
Article in English | MEDLINE | ID: mdl-17977534

ABSTRACT

Ceramide is a key metabolite in the pathway of sphingolipid biosynthesis. In mammals, ceramide is synthesized by N-acylation of a sphingoid long-chain base by a family of ceramide synthases (CerS), each of which displays a high specificity towards acyl CoAs of different chain lengths. We now optimize a previously-described assay for measuring CerS activity for use upon over-expression of mammalian CerS, and using these conditions, establish the K(m) value of each CerS towards sphinganine. Remarkably, the K(m) values towards sphinganine are all similar, ranging from 2 to 5microM, even for CerS proteins that are able to use more than one acyl CoA for ceramide synthesis (i.e. CerS4). The availability of this assay will permit further accurate characterization of the kinetic parameters of mammalian CerS proteins.


Subject(s)
Oxidoreductases/metabolism , Sphingosine/analogs & derivatives , Animals , Base Sequence , COS Cells , Cell Line , Chlorocebus aethiops , DNA Primers/genetics , Humans , Kinetics , Mice , Oxidoreductases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity , Sphingosine/metabolism , Substrate Specificity , Transfection
14.
Mitochondrion ; 6(3): 118-25, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16713754

ABSTRACT

Recent evidence suggests that the ability of ceramides to induce apoptosis is due to a direct action on mitochondria. Mitochondria are known to contain enzymes responsible for ceramide synthesis and hydrolysis and mitochondrial ceramide levels have been shown to be elevated prior to the mitochondrial phase of apoptosis. Ceramides have been reported to induce the release of intermembrane space proteins from mitochondria, which has been linked to their ability to form large channels in membranes. The aim of this study was to determine if the membrane concentration of ceramide required for the formation of protein permeable channels is within the range that is present in mitochondria during the induction phase of apoptosis. Only a very small percentage of the ceramide actually inserts into the mitochondrial membranes. The permeability of the mitochondrial outer membrane correlates directly with the level of ceramide in the membrane. Importantly, the concentration of ceramide at which significant channel formation occurs is consistent with the level of mitochondrial ceramide that occurs during the induction phase of apoptosis (4 pmol ceramide/nanomole phospholipid). Similar results were obtained with short- and long-chain ceramide. Ceramide channel formation is specific to mitochondrial membranes in that no channel formation occurs in the plasma membranes of erythrocytes even at concentrations 20 times higher than those required for channel formation in mitochondrial outer membranes. Thus, ceramide channels are good candidates for the pathway by which proapoptotic proteins are released from mitochondria during the induction phase of apoptosis.


Subject(s)
Ceramides/metabolism , Erythrocytes/metabolism , Intracellular Membranes/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Membranes/metabolism , Animals , Apoptosis , Dose-Response Relationship, Drug , Hydrolysis , Male , Phospholipids/chemistry , Rats , Time Factors
15.
FASEB J ; 19(7): 860-2, 2005 May.
Article in English | MEDLINE | ID: mdl-15728664

ABSTRACT

An age-dependent acceleration of apoptosis occurs in female germ cells (oocytes), and this requires communication between the oocyte and its surrounding somatic (cumulus) cells. Here we show in aged mice that ceramide is translocated from cumulus cells into the adjacent oocyte and induces germ cell apoptosis that can be prevented by sphingosine-1-phosphate. Trafficking of ceramide requires gap junction-dependent communication between the cumulus cells and the oocyte as well as intact lipid rafts. Further, the occurrence of the elevated incidence of apoptosis in oocytes of aged females is concomitant with an enhanced sensitivity of the oocyte to a spike in cytosolic ceramide levels, as well as increased bax mRNA and Bax protein levels. Thus, the force driving the age-related increase in female germ cell death is multifactorial, but changes in the intercellular trafficking of ceramide, along with hypersensitivity of oocytes to ceramide, are key factors in this process.


Subject(s)
Aging/physiology , Apoptosis/physiology , Ceramides/physiology , Oocytes/physiology , Animals , Apoptosis/drug effects , Biological Transport , Cell Communication , Cells, Cultured , Ceramides/analysis , Ceramides/deficiency , Ceramides/pharmacology , Cytochromes c/metabolism , Cytosol/chemistry , Female , Filipin/pharmacology , Gap Junctions/drug effects , Gap Junctions/physiology , Glycyrrhetinic Acid/pharmacology , Lysophospholipids/pharmacology , Membrane Microdomains/drug effects , Membrane Microdomains/physiology , Mice , Mice, Inbred ICR , Mice, Knockout , Oocytes/chemistry , Oocytes/ultrastructure , Ovarian Follicle/cytology , Ovarian Follicle/physiology , RNA, Messenger/analysis , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , bcl-2-Associated X Protein/analysis , bcl-2-Associated X Protein/genetics
16.
Radiat Res ; 162(6): 711-28, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15548121

ABSTRACT

To develop approaches to prophylaxis/protection, mitigation and treatment of radiation injuries, appropriate models are needed that integrate the complex events that occur in the radiation-exposed organism. While the spectrum of agents in clinical use or preclinical development is limited, new research findings promise improvements in survival after whole-body irradiation and reductions in the risk of adverse effects of radiotherapy. Approaches include agents that act on the initial radiochemical events, agents that prevent or reduce progression of radiation damage, and agents that facilitate recovery from radiation injuries. While the mechanisms of action for most of the agents with known efficacy are yet to be fully determined, many seem to be operating at the tissue, organ or whole animal level as well as the cellular level. Thus research on prophylaxis/protection, mitigation and treatment of radiation injuries will require studies in whole animal models. Discovery, development and delivery of effective radiation modulators will also require collaboration among researchers in diverse fields such as radiation biology, inflammation, physiology, toxicology, immunology, tissue injury, drug development and radiation oncology. Additional investment in training more scientists in radiation biology and in the research portfolio addressing radiological and nuclear terrorism would benefit the general population in case of a radiological terrorism event or a large-scale accidental event as well as benefit patients treated with radiation.


Subject(s)
Radiation Injuries/prevention & control , Animals , Central Nervous System/radiation effects , Gastrointestinal Tract/radiation effects , Hematopoietic System/radiation effects , Humans , Kidney/radiation effects , Lung/radiation effects , Radiation Injuries/drug therapy , Skin/radiation effects , Zebrafish
17.
Cancer Lett ; 206(2): 169-80, 2004 Apr 08.
Article in English | MEDLINE | ID: mdl-15013522

ABSTRACT

Sphingolipids, which include ceramides and sphingosine, are essential structural components of cell membranes that also have messenger functions that regulate the proliferation, survival, and death of cells. Exogenous application of ceramide is cytotoxic, and exposure of cells to radiation or chemotherapy is associated with increased ceramide levels due to enhanced de novo synthesis, catabolism of sphingomyelin, or both. Ceramide can be metabolized to less toxic forms by glycosylation, acylation, or by catabolism to sphingosine, which is then phosphorylated to the anti-apoptotic sphingosine 1-phosphate. Glucosylceramide synthase overexpression has been shown to enhance resistance to doxorubicin, suggesting that inhibition of ceramide metabolism or catabolism might enhance cancer chemotherapy. Several anticancer agents, including the cytotoxic retinoid, fenretinide (4-HPR), have been shown to act, at least in part, by increasing tumor cell ceramide via de novo synthesis. Combinations of 4-HPR and modulators of ceramide action and/or metabolism demonstrated increased anti-tumor activity in pre-clinical models with minimal toxicity for non-malignant cells, and were effective in a p53-independent manner against tumor cell lines resistant to standard cytotoxic agents. Phase I trials of ceramide metabolism inhibitors in combination with 4-HPR and with other cytotoxic agents are in development. Thus, pharmacological manipulation of sphingolipid metabolism to enhance tumor cell ceramide is being realized and offers a novel approach to cancer chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Ceramides/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans
18.
J Biol Chem ; 278(39): 37169-74, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-12855704

ABSTRACT

Ceramide is known to induce structural rearrangements in membrane bilayers, including the formation of ceramide-rich and -poor domains and the efflux of aqueous solutes. This report describes a novel effect of ceramide, namely the induction of transbilayer lipid movements. This effect was demonstrated in both model (large unilamellar vesicles) and cell (erythrocyte ghost) membranes in which ceramide generation took place in situ through the action of an externally added sphingomyelinase. Two different novel assays were developed to detect transbilayer lipid movement. One of the assays required the preparation of vesicles containing a ganglioside only in the outer monolayer and entrapped neuraminidase. Sphingomyelinase activity induced ganglioside hydrolysis under conditions in which no neuraminidase was released from the vesicles. The second assay involved the preparation of liposomes or erythrocyte ghosts labeled with a fluorescent energy donor in their inner leaflets. Sphingomyelin hydrolysis was accompanied by fluorescence energy transfer to an impermeable acceptor in the outer aqueous medium. Ceramide-induced transbilayer lipid movement is explained in terms of another well known property of ceramide, namely the facilitation of lamellar to non-lamellar lipid-phase transitions. Thus, sphingomyelinase generates ceramide on one side of the membrane; ceramide then induces the transient formation of non-lamellar structural intermediates, which cause the loss of lipid asymmetry in the bilayer, i.e. the transbilayer movement of ceramide together with other lipids. As direct targets for ceramide tend to be intracellular, these observations may be relevant to the mechanism of transmembrane signaling by means of the sphingomyelin pathway.


Subject(s)
Cell Membrane/metabolism , Lipid Bilayers/metabolism , Membrane Lipids/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Biological Transport , Erythrocyte Membrane/metabolism , Fluorescence Resonance Energy Transfer , Gangliosides/metabolism , Humans , Kinetics
20.
Endocrinology ; 144(1): 69-74, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12488331

ABSTRACT

The industrial chemical, 4-vinylcyclohexene diepoxide (VCD), kills oocytes within immature follicles in the ovaries of mice and rats and is considered a potential occupational health hazard. It has been reported that VCD-induced follicle loss occurs via a cell death process involving elevated expression of Bax, a proapoptotic Bcl-2 family member, and increased caspase-3-like activity. We have previously shown that oocytes lacking acid sphingomyelinase (ASMase; an enzyme that generates the proapoptotic stress sensor ceramide), the aromatic hydrocarbon receptor (Ahr), Bax, or caspase-2 are resistant to apoptosis induced by other chemical toxicants. Therefore, this study was designed to investigate the functional importance of ASMase, Ahr, Bax, and caspase-2 as well as the related executioner enzyme caspase-3 to VCD-induced ovotoxicity in mice using gene knockout technology. For each gene mutant mouse line, wild-type and homozygous-null female siblings derived from heterozygous matings were given once-daily ip injections of either vehicle (sesame oil) or VCD (80 mg/kg body weight) for 15 d (three or four mice per treatment group per genotype). Ovaries were collected 24 h after the final injection and analyzed for the total number of nonatretic primordial and primary follicles remaining per ovary. No differences in the extent of primordial or primary follicle destruction resulting from VCD exposure were observed in wild-type vs. ASMase- or Ahr-deficient mice. By contrast, the extent of VCD-induced primordial follicle depletion in Bax-deficient mice (45 +/- 11%) was significantly (P < 0.05) lower than that in wild-type females (85 +/- 2%). The extent of primary follicle loss in bax-null mice exposed to VCD (3 +/- 22%) was also significantly (P < 0.05) lower than that in their wild-type sisters (86 +/- 4%). In caspase-2-deficient mice, significantly (P < 0.05) fewer oocyte-containing primary follicles were destroyed by VCD (17 +/- 19%) vs. wild-type controls (71 +/- 6%); however, no significant difference in the extent of VCD-induced primordial follicle destruction was observed in caspase-2-null vs. wild-type females. Finally, in caspase-3-deficient mice, significantly (P < 0.05) fewer oocyte-containing primary follicles were destroyed by VCD (33 +/- 3%) vs. wild-type controls (71 +/- 2%); however, no significant difference in the extent of VCD-induced primordial follicle destruction was observed in caspase-3-null vs. wild-type females. We conclude that Bax, caspase-2, and caspase-3, but not ASMase or Ahr, are functionally important in VCD-induced follicle loss. However, as a loss of Bax, caspase-2, or caspase-3 function conveyed only partial protection from the ovotoxic effects of VCD, other cell death pathways that either function independently of Bax, caspase-2, and caspase-3 or are not apoptotic in nature are also involved.


Subject(s)
Caspases/physiology , Cyclohexanes/toxicity , Ovarian Follicle/drug effects , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Animals , Apoptosis/drug effects , Caspase 2 , Caspase 3 , Caspases/deficiency , Caspases/genetics , Cell Count , Cyclohexenes , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/drug effects , Oocytes/enzymology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/physiology , Species Specificity , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/physiology , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...