Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Methods Mol Biol ; 2451: 569-577, 2022.
Article in English | MEDLINE | ID: mdl-35505033

ABSTRACT

Recently, it has become clear that a prerequisite requirement for most cancer therapies is controlling the negative impact of the activity of immunosuppressory cell populations. It is therefore of a considerable interest to develop treatments for containing the operation of major myeloid and lymphoid immunoregulatory cell populations. We have reported that acid ceramidase inhibitor LCL521 effectively overrides the activity of immunoregulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) engaged in the context of tumor response to photodynamic therapy (PDT). The present communication dissects and describes in detail the procedure for the use of LCL521 as an adjuvant to PDT for improved cure rates of treated tumors based on restricting the activity of immunoregulatory cell populations.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Photochemotherapy , Humans , Myeloid-Derived Suppressor Cells/pathology , Neoplasms/drug therapy , Neoplasms/pathology , T-Lymphocytes/pathology
2.
Bioorg Med Chem ; 32: 116011, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33461145

ABSTRACT

B13 is an acid ceramidase (ACDase) inhibitor. The two chiral centers of this aromatic amido alcohol lead to four stereoisomers, yet we have little knowledge about its erythro- enantiomers, (1R, 2S) and (1S, 2R). In this paper, for the first time, the synthesis of two erythro- enantiomers is described, and the compounds are evaluated along with two threo- enantiomers, (1R, 2R) and (1S, 2S). The key metabolites and sphingolipid (SL) profile of the full set of B13 stereoisomers in MCF7 breast carcinoma cells are presented. The results demonstrated that the erythro- enantiomers were more effective than the threo- enantiomers on growth inhibition in MCF7 cells, although there were no statistically significant differences within the threo- and erythro- series. Measurement of intracellular levels of the compounds indicated that the erythro- seemed a little more cell permeable than the threo- enantiomers; also, the (1R, 2S) isomer with the same stereo structure as natural ceramide (Cer) could be hydrolyzed and phosphorylated in MCF7 cells. Furthermore, we also observed the formation of C16 homologs from the full set of B13 isomers within the cells, indicating the occurrence of de-acylation and re-acylation of the amino group of the aromatic alcohol. Moreover, the decrease in the Cer/Sph ratio suggests that the growth inhibition from (1R, 2S) isomer is not because of the inhibition of ceramidases. Taken together, (1R, 2S) could be developed as a substitute of natural Cer.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Propanolamines/pharmacology , Sphingolipids/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Molecular Structure , Propanolamines/chemical synthesis , Propanolamines/chemistry , Sphingolipids/metabolism , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
3.
Photochem Photobiol Sci ; 19(9): 1145-1151, 2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32821888

ABSTRACT

Our recent investigation uncovered that the acid ceramidase inhibitor LCL521 enhances the direct tumor cell killing effect of photodynamic therapy (PDT) treatment. The present study aimed at elucidating the mechanisms underlying this effect. Exposing mouse squamous cell carcinoma SCCVII cells treated with temoporfin-based PDT to LCL521 (rising ceramide concentration) produced a much greater decrease in cell survival than comparable exposure to the sphingosine kinase-1 inhibitor PF543 (that reduces sphingosine-1-phosphate concentration). This is consistent with recognizing the rising levels of pro-apoptotic sphingolipid ceramide as being more critical in promoting the death of PDT-treated cells than the reduction in the availability of pro-survival acting sphingosine-1 phosphate. This pro-apoptotic impact of LCL521, which was suppressed by the apoptosis inhibitor bongkrekic acid, involves the interaction with the cellular stress signaling network. Hence, inhibiting the key elements of these pathways markedly influenced the adjuvant effect of LCL521 on the PDT response. Particularly effective was the inositol-requiring element-1 (IRE1) kinase inhibitor STF-083010 that dramatically enhanced the killing of cells treated with PDT plus LCL521. An important role in the survival of these cells was exhibited by master transcription factors STAT3 and HIF-1α. The STAT3 inhibitor NSC 74859 was especially effective in further reducing the cell survival rates, suggesting its possible exploitation for therapeutic gain. An additional finding in this study is that LCL521-promoted PDT-mediated cell killing through ceramide-mediated lethal effects is extended to the interaction with other cancer treatment modalities with a rapid cellular stress impact such as photothermal therapy (PTT) and cryoablation therapy (CAT).


Subject(s)
Acetates/pharmacology , Amines/pharmacology , Antineoplastic Agents/pharmacology , Ceramidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hyperthermia, Induced , Photochemotherapy , Acetates/chemical synthesis , Acetates/chemistry , Amines/chemical synthesis , Amines/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Ceramidases/metabolism , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Mice , Tumor Cells, Cultured
4.
Cytokine ; 135: 155219, 2020 11.
Article in English | MEDLINE | ID: mdl-32738771

ABSTRACT

The bioactive sphingolipid ceramide affects immune responses although its effect on antigen (Ag) processing and delivery by HLA class II to CD4+T-cells remains unclear. Therefore, we examined the actions of a novel cell-permeable acid ceramidase (AC) inhibitor [(1R,2R) N myristoylamino-(4'-nitrophenyl)-propandiol-1,3] on antigen presentation and inflammatory cytokine production by Ag-presenting cells (APCs) such as B-cells, macrophages, and dendritic cells. We found that AC inhibition in APCs perturbed Ag-processing and presentation via HLA-DR4 (MHC class II) proteins as measured by coculture assay and T-cell production of IL-2. Mass spectral analyses showed that B13 treatment significantly raised levels of four types of ceramides in human B-cells. B13 treatment did not alter Ag internalization and class II protein expression, but significantly inhibited lysosomal cysteinyl cathepsins (B, S and L) and thiol-reductase (GILT), HLA class II Ag-processing, and generation of functional class II-peptide complexes. Ex vivo Ag presentation assays showed that inhibition of AC impaired primary and recall CD4+T-cell responses and cytokine production in response against type II collagen. Further, B13 delayed onset and reduced severity of inflamed joints and cytokine production in the collagen-induced arthritis mouse model in vivo. These findings suggest that inhibition of AC in APCs may dysregulate endolysosomal proteases and HLA class II-associated self-antigen presentation to CD4+T-cells, attenuating inflammatory cytokine production and suppressing host autoimmune responses.


Subject(s)
Acid Ceramidase/immunology , Antigen Presentation/immunology , Arthritis, Experimental/immunology , Autoimmune Diseases/immunology , Histocompatibility Antigens Class II/immunology , Animals , Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cathepsins/immunology , Cell Line , HLA-DR4 Antigen/immunology , Humans , Macrophages/immunology , Mice , Mice, Inbred DBA
5.
Prostate ; 79(8): 896-908, 2019 06.
Article in English | MEDLINE | ID: mdl-30900312

ABSTRACT

Numerous genetic alterations have been identified during prostate cancer progression. The influence of environmental factors, particularly the diet, on the acceleration of tumor progression is largely unknown. Expression levels and/or activity of Src kinase are highly elevated in numerous cancers including advanced stages of prostate cancer. In this study, we demonstrate that high-fat diets (HFDs) promoted pathological transformation mediated by the synergy of Src and androgen receptor in vivo. Additionally, a diet high in saturated fat significantly enhanced proliferation of Src-mediated xenograft tumors in comparison with a diet high in unsaturated fat. The saturated fatty acid palmitate, a major constituent in a HFD, significantly upregulated the biosynthesis of palmitoyl-CoA in cancer cells in vitro and in xenograft tumors in vivo. The exogenous palmitate enhanced Src-dependent mitochondrial ß-oxidation. Additionally, it elevated the amount of C16-ceramide and total saturated ceramides, increased the level of Src kinase localized in the cell membrane, and Src-mediated downstream signaling, such as the activation of mitogen-activated protein kinase and focal adhesion kinase. Our results uncover how the metabolism of dietary palmitate cooperates with elevated Src kinase in the acceleration of prostate tumor progression.


Subject(s)
Palmitates/administration & dosage , Prostatic Neoplasms/etiology , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Diet, High-Fat/adverse effects , Disease Progression , HEK293 Cells , Heterografts , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , PC-3 Cells , Palmitates/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
6.
Bioorg Med Chem ; 26(23-24): 6067-6075, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30448190

ABSTRACT

The function of acid ceramidase (ACDase), whose congenital deficiency leads to Farber disease, has been recognized to be vital to tumor cell biology, and inhibition of its activity may be beneficial in cancer therapy. Therefore, manipulation of the activity of this enzyme may have significant effect, especially on cancer cells. LCL521, Di-DMG-B13, is a lysosomotropic inhibitor of ACDase. Here we define complexities in the actions of LCL521 on ACDase. Systematic studies in MCF7 cells showed dose and time divergent action of LCL521 on ACDase protein expression and sphingolipid levels. Low dose of LCL521 (1 µM) effectively inhibited ACDase in cells, but the effects were transient. A higher dose of LCL521 (10 µM) caused a profound decrease of sphingosine and increase of ceramide, but additionally affected the processing and regeneration of the ACDase protein, with biphasic and reversible effects on the expression of ACDase, which paralleled the long term changes of cellular sphingosine and ceramide. Finally, the higher concentrations of LCL521 also inhibited Dihydroceramide desaturase (DES-1). In summary, LCL521 exhibits significant effects on ACDase in a dose and time dependent manner, but dose range and treatment time need to be paid attention to specify its future exploration on ACDase targeted cancer treatment.


Subject(s)
Acetates/pharmacology , Acid Ceramidase/antagonists & inhibitors , Amines/pharmacology , Enzyme Inhibitors/pharmacology , Sphingolipids/antagonists & inhibitors , Acid Ceramidase/metabolism , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Molecular Structure , Sphingolipids/metabolism , Structure-Activity Relationship , Time Factors , Tumor Cells, Cultured
7.
Cancer Res ; 77(24): 6950-6962, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29038344

ABSTRACT

Protein N-myristoylation enables localization to membranes and helps maintain protein conformation and function. N-myristoyltransferases (NMT) catalyze co- or posttranslational myristoylation of Src family kinases and other oncogenic proteins, thereby regulating their function. In this study, we provide genetic and pharmacologic evidence that inhibiting the N-myristoyltransferase NMT1 suppresses cell-cycle progression, proliferation, and malignant growth of prostate cancer cells. Loss of myristoylation abolished the tumorigenic potential of Src and its synergy with androgen receptor in mediating tumor invasion. We identified the myristoyl-CoA analogue B13 as a small-molecule inhibitor of NMT1 enzymatic activity. B13 exposure blocked Src myristoylation and Src localization to the cytoplasmic membrane, attenuating Src-mediated oncogenic signaling. B13 exerted its anti-invasive and antitumor effects against prostate cancer cells, with minimal toxic side-effects in vivo Structural optimization based on structure-activity relationships enabled the chemical synthesis of LCL204, with enhanced inhibitory potency against NMT1. Collectively, our results offer a preclinical proof of concept for the use of protein myristoylation inhibitors as a strategy to block prostate cancer progression. Cancer Res; 77(24); 6950-62. ©2017 AACR.


Subject(s)
Acyltransferases/physiology , Myristic Acid/metabolism , Phosphotransferases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Acyltransferases/antagonists & inhibitors , Acyltransferases/genetics , Acyltransferases/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , Disease Progression , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Mutation, Missense , Phosphorylation/drug effects , Phosphorylation/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Protein Processing, Post-Translational/genetics , Proto-Oncogene Proteins pp60(c-src)/chemistry , Proto-Oncogene Proteins pp60(c-src)/genetics , Structure-Activity Relationship , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
PLoS One ; 12(6): e0177805, 2017.
Article in English | MEDLINE | ID: mdl-28614356

ABSTRACT

Acid ceramidase, which catalyzes ceramide hydrolysis to sphingosine and free fatty acid mainly in the lysosome, is being recognized as a potential therapeutic target for cancer. B13 is an effective and selective acid ceramidase inhibitor in vitro, but not as effective in cells due to poor access to the lysosomal compartment. In order to achieve targeting of B13 to the lysosome, we designed lysosomotropic N, N-dimethyl glycine (DMG)-conjugated B13 prodrug LCL521 (1,3-di-DMG-B13). Our previous results indicated the efficient delivery of B13 to the lysosome resulted in augmented effects of LCL521 on cellular acid ceramidase as evaluated by effects on substrate/product levels. Our current studies indicate that functionally, this translated into enhanced inhibition of cell proliferation. Moreover, there were greater synergistic effects of LCL521 with either ionizing radiation or Tamoxifen. Taken together, these results clearly indicate that compartmental targeting for the inhibition of acid ceramidase is an efficient and valuable therapeutic strategy.


Subject(s)
Acid Ceramidase/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/enzymology , Nitrobenzenes/chemistry , Prodrugs/chemical synthesis , Propanolamines/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Drug Synergism , Female , Humans , Prodrugs/chemistry , Prodrugs/pharmacology , Tamoxifen/pharmacology
9.
J Lipid Res ; 58(7): 1439-1452, 2017 07.
Article in English | MEDLINE | ID: mdl-28490444

ABSTRACT

Acute kidney injury (AKI), resulting from chemotherapeutic agents such as cisplatin, remains an obstacle in the treatment of cancer. Cisplatin-induced AKI involves apoptotic and necrotic cell death, pathways regulated by sphingolipids such as ceramide and glucosylceramide. Results from this study indicate that C57BL/6J mice treated with cisplatin had increased ceramide and hexosylceramide levels in the renal cortex 72 h following cisplatin treatment. Pretreatment of mice with inhibitors of acid sphingomyelinase and de novo ceramide synthesis (amitriptyline and myriocin, respectively) prevented accumulation of ceramides and hexosylceramide in the renal cortex and protected from cisplatin-induced AKI. To determine the role of ceramide metabolism to hexosylceramides in kidney injury, we treated mice with a potent and highly specific inhibitor of glucosylceramide synthase, the enzyme responsible for catalyzing the glycosylation of ceramides to form glucosylceramides. Inhibition of glucosylceramide synthase attenuated the accumulation of the hexosylceramides and exacerbated ceramide accumulation in the renal cortex following treatment of mice with cisplatin. Increasing ceramides and decreasing glucosylceramides in the renal cortex sensitized mice to cisplatin-induced AKI according to markers of kidney function, kidney injury, inflammation, cell stress, and apoptosis. Under conditions of high ceramide generation, data suggest that metabolism of ceramides to glucosylceramides buffers kidney ceramides and helps attenuate kidney injury.-Dupre, T. V., M. A. Doll, P. P. Shah, C. N. Sharp, D. Siow, J. Megyesi, J. Shayman, A. Bielawska, J. Bielawski, L. J. Beverly, M. Hernandez-Corbacho, C. J. Clarke, A. J. Snider, R. G. Schnellmann, L. M. Obeid, Y. A. Hannun, and L. J. Siskind. Inhibiting glucosylceramide synthase exacerbates cisplatin-induced acute kidney injury. J. Lipid Res 2017. 58: 1439-1452.


Subject(s)
Acute Kidney Injury/chemically induced , Cisplatin/adverse effects , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Animals , Ceramides/metabolism , Kidney Cortex/blood supply , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Male , Mice , Rats , Reperfusion Injury/metabolism
10.
Article in English | MEDLINE | ID: mdl-28377281

ABSTRACT

Sphingosine-1-phosphate (S1P) is a biologically active sphingolipid metabolite which has been implicated in many diseases including cancer and inflammatory diseases. Recently, sphingosine kinase 1 (SK1), one of the isozymes which generates S1P, has been implicated in the development and progression of inflammatory bowel disease (IBD). Based on our previous work, we set out to determine the efficacy of a novel SK1 selective inhibitor, LCL351, in a murine model of IBD. LCL351 selectively inhibits SK1 both in vitro and in cells. LCL351, which accumulates in relevant tissues such as colon, did not have any adverse side effects in vivo. In mice challenged with dextran sodium sulfate (DSS), a murine model for IBD, LCL351 treatment protected from blood loss and splenomegaly. Additionally, LCL351 treatment reduced the expression of pro-inflammatory markers, and reduced neutrophil infiltration in colon tissue. Our results suggest inflammation associated with IBD can be targeted pharmacologically through the inhibition and degradation of SK1. Furthermore, our data also identifies desirable properties of SK1 inhibitors.


Subject(s)
Colitis/drug therapy , Colitis/immunology , Dextran Sulfate/adverse effects , Guanidines/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Sphingosine/pharmacology , A549 Cells , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Colitis/chemically induced , Colitis/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Guanidines/therapeutic use , Humans , Sphingosine/therapeutic use , Tumor Necrosis Factor-alpha/genetics
11.
Anticancer Res ; 37(3): 1213-1218, 2017 03.
Article in English | MEDLINE | ID: mdl-28314284

ABSTRACT

BACKGROUND/AIM: Because patients with cancer of apparently equivalent stage often have different outcomes, it is necessary to gather additional information to complement cancer staging. Dysregulated sphingolipid metabolism contributes to carcinogenesis. In this retrospective pilot study, we tested the hypothesis that changes in serum levels of sphingolipids are associated with stage IV colorectal cancer (CRC). PATIENTS AND METHODS: We used commercially available serum samples from healthy males and patients with CRC (adenocarcinoma of the large intestine, stage IV with metastases). Blood samples were obtained immediately prior to anesthesia/surgery. We measured sphingolipid levels in sera using mass spectrometry. RESULTS: In serum of patients with CRC, the levels of C16-, C18-, C18:1-, and C24:1-ceramide, as well as those of sphingosine, were significantly higher than those of controls. In contrast, the levels of C24-sphingomyelin were significantly lower than those of controls. A global test of association showed that ceramides and sphingomyelins but not hexosylceramides were significantly associated with stage IV CRC. CONCLUSION: Sphingolipids have a potential of serving as novel, non-invasive, inexpensive, and effective blood-based biomarkers to complement CRC staging for better prognosis and more personalized medicine.


Subject(s)
Ceramides/blood , Colorectal Neoplasms/blood , Sphingomyelins/blood , Sphingosine/blood , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism , Male , Middle Aged , Pilot Projects , Prognosis , Retrospective Studies
12.
Anticancer Res ; 37(2): 455-463, 2017 02.
Article in English | MEDLINE | ID: mdl-28179290

ABSTRACT

BACKGROUND/AIM: Combining an anticancer agent fenretinide (HPR) or C6-pyridinium ceramide (LCL29) with Foscan-mediated photodynamic therapy (FoscanPDT) is expected to augment anticancer benefits of each substance. We showed that treatment with FoscanPDT+HPR enhanced accumulation of C16-dihydroceramide, and that fumonisin B1 (FB), an inhibitor of ceramide synthase, counteracted caspase-3 activation and colony-forming ability of head and neck squamous cell carcinoma (HNSCC) cells. Because cancer cells appear to be more susceptible to increased levels of the endoplasmic reticulum (ER) stress than normal cells, herein we tested the hypothesis that FoscanPDT combined with HPR or LCL29 induces FB-sensitive ER stress-associated apoptosis that affects cell survival. MATERIALS AND METHODS: Using an HNSCC cell line, we determined: cell survival by clonogenic assay, caspase-3 activity by spectrofluorometry, the expression of the ER markers BiP and CHOP by quantitative real-time polymerase chain reaction and western immunoblotting, and sphingolipid levels by mass spectrometry. RESULTS: Similar to HPR+FoscanPDT, LCL29+FoscanPDT induced enhanced loss of clonogenicity and caspase-3 activation, that were both inhibited by FB. Our additional pharmacological evidence showed that the enhanced loss of clonogenicity after the combined treatments was singlet oxygen-, ER stress- and apoptosis-dependent. The combined treatments induced enhanced, FB-sensitive, up-regulation of BiP and CHOP, as well as enhanced accumulation of sphingolipids. CONCLUSION: Our data suggest that enhanced clonogenic cell killing after the combined treatments is dependent on oxidative- and ER-stress, apoptosis, and FB-sensitive sphingolipid production, and should help develop more effective mechanism-based therapeutic strategies.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Ceramides/pharmacology , Endoplasmic Reticulum Stress/drug effects , Fenretinide/pharmacology , Fumonisins/pharmacology , Head and Neck Neoplasms/drug therapy , Mesoporphyrins/pharmacology , Photochemotherapy/methods , Pyridinium Compounds/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Combined Modality Therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Radiation-Sensitizing Agents/pharmacology , Squamous Cell Carcinoma of Head and Neck
13.
Cell Signal ; 29: 52-61, 2017 01.
Article in English | MEDLINE | ID: mdl-27702691

ABSTRACT

Despite great promise, combining anti-angiogenic and conventional anti-cancer drugs has produced limited therapeutic benefit in clinical trials, presumably because mechanisms of anti-angiogenic tissue response remain only partially understood. Here we define a new paradigm, in which anti-angiogenic drugs can be used to chemosensitize tumors by targeting the endothelial acid sphingomyelinase (ASMase) signal transduction pathway. We demonstrate that paclitaxel and etoposide, but not cisplatin, confer ASMase-mediated endothelial injury within minutes. This rapid reaction is required for human HCT-116 colon cancer xenograft complete response and growth delay. Whereas VEGF inhibits ASMase, anti-VEGFR2 antibodies de-repress ASMase, enhancing endothelial apoptosis and drug-induced tumor response in asmase+/+, but not in asmase-/-, hosts. Such chemosensitization occurs only if the anti-angiogenic drug is delivered 1-2h before chemotherapy, but at no other time prior to or post chemotherapy. Our studies suggest that precisely-timed administration of anti-angiogenic drugs in combination with ASMase-targeting anti-cancer drugs is likely to optimize anti-tumor effects of systemic chemotherapy. This strategy warrants evaluation in future clinical trials.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Molecular Targeted Therapy , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cattle , Ceramides/metabolism , Drug Delivery Systems , Endothelium/metabolism , Enzyme Activation/drug effects , HCT116 Cells , Humans , Male , Mice, Inbred C57BL , Paclitaxel/pharmacology , Sphingomyelin Phosphodiesterase/metabolism
14.
Oncotarget ; 7(51): 83907-83925, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27880732

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are immune suppressive cells that are hallmarks of human cancer. MDSCs inhibit cytotoxic T lymphocytes (CTLs) and NK cell functions to promote tumor immune escape and progression, and therefore are considered key targets in cancer immunotherapy. Recent studies determined a key role of the apoptosis pathways in tumor-induced MDSC homeostasis and it is known that ceramide plays a key role in regulation of mammalian cell apoptosis. In this study, we aimed to determine the efficacy and underlying molecular mechanism of ceramide in suppression of MDSCs. Treatment of tumor-bearing mice with LCL521, a lysosomotropic inhibitor of acid ceramidase, significantly decreased MDSC accumulation in vivo. Using a MDSC-like myeloid cell model, we determined that LCL521 targets lysosomes and increases total cellular C16 ceramide level. Although MDSC-like cells have functional apoptosis pathways, LCL521-induced MDSC death occurs in an apoptosis- and necroptosis-independent mechanism. LCL521 treatment resulted in an increase in the number of autophagic vesicles, heterolysosomes and swollen ERs. Finally, concomitant inhibition of cathepsin B and cathepsin D was required to significantly decrease LCL521-induced cell death. Our observations indicate that LCL521 targets lysosomes to activate cathepsin B and cathepsin D, resulting in interrupted autophagy and ER stress that culminates in MDSC death. Therefore, a ceramidase inhibitor is potentially an effective adjunct therapeutic agent for suppression of MDSCs to enhance the efficacy of CTL-based cancer immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cathepsin B/metabolism , Cathepsin D/metabolism , Ceramides/metabolism , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , Lysosomes/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Sarcoma/drug therapy , Signal Transduction/drug effects , Acid Ceramidase/antagonists & inhibitors , Acid Ceramidase/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Lysosomes/enzymology , Lysosomes/pathology , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/pathology , Sarcoma/enzymology , Sarcoma/immunology , Sarcoma/pathology , Time Factors
15.
J Lipid Res ; 57(11): 2028-2039, 2016 11.
Article in English | MEDLINE | ID: mdl-27623848

ABSTRACT

Th1 pro-inflammatory cytokines, i.e., TNF-α and IFN-γ, in combination are known to induce cell death in several cell types, including oligodendrocytes, but the mechanism of their synergistic cytotoxicity is unclear. Although ceramide (Cer) has been implicated in cytokine- and stress-induced cell death, its intracellular levels alone cannot explain cytokine synergy. We considered the possibility that Cer released as part of extracellular vesicles may contribute to cytokine-induced synergistic cell death. Using a human oligodendroglioma (HOG) cell line as a model, here we show that exosomes derived from TNF-α-treated "donor" cells, while being mildly toxic to fresh cultures (similar to individual cytokines), induce enhanced cell death when added to IFN-γ-primed target cultures in a fashion resembling the effect of cytokine combination. Further, the sphingolipid profiles of secreted exosomes, as determined by HPLC-MS/MS, revealed that the treatment with the cytokines time-dependently induced the formation and exosomal release, in particular of C16-, C24-, and C24:1-Cer species; C16-, C24-, and C24:1-dihydroCer species; and C16-, C24-, and C24:1-SM species. Finally, exogenous C6-Cer or C16-Cer mimicked and enhanced the cytotoxic effects of the cytokines upon HOG cells, thereby supporting the cell death-signaling role of extracellular Cer.


Subject(s)
Ceramides/metabolism , Interferon-gamma/metabolism , Oligodendroglioma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Death/genetics , Cell Line, Tumor , Ceramides/chemistry , Ceramides/genetics , Chromatography, High Pressure Liquid , Exosomes , Extracellular Vesicles/metabolism , Humans , Interferon-gamma/administration & dosage , Interferon-gamma/genetics , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglioma/pathology , Sphingolipids/chemistry , Sphingolipids/metabolism , Tandem Mass Spectrometry , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/genetics
16.
Int J Cancer ; 139(6): 1372-8, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27136745

ABSTRACT

Acid ceramidase has been identified as a promising target for cancer therapy. One of its most effective inhibitors, LCL521, was examined as adjuvant to photodynamic therapy (PDT) using mouse squamous cell carcinoma SCCVII model of head and neck cancer. Lethal effects of PDT, assessed by colony forming ability of in vitro treated SCCVII cells, were greatly enhanced when combined with 10 µM LCL521 treatment particularly when preceding PDT. When PDT-treated SCCVII cells are used to vaccinate SCCVII tumor-bearing mice (PDT vaccine protocol), adjuvant LCL521 treatment (75 mg/kg) resulted in a marked retardation of tumor growth. This effect can be attributed to the capacity of LCL521 to effectively restrict the activity of two main immunoregulatory cell populations (Tregs and myeloid-derived suppressor cells, MDSCs) that are known to hinder the efficacy of PDT vaccines. The therapeutic benefit with adjuvant LCL521 was also achieved with SCCVII tumors treated with standard PDT when using immunocompetent mice but not with immunodeficient hosts. The interaction of LCL521 with PDT-based antitumor mechanisms is dominated by immune system contribution that includes overriding the effects of immunoregulatory cells, but could also include a tacit contribution from boosting direct tumor cell kill.


Subject(s)
Acid Ceramidase/antagonists & inhibitors , Cancer Vaccines , Enzyme Inhibitors/pharmacology , Photochemotherapy , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Humans , Immunomodulation , Mice , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
17.
J Photochem Photobiol B ; 159: 191-5, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27085050

ABSTRACT

We and others have shown that stresses, including photodynamic therapy (PDT), can disrupt the de novo sphingolipid biosynthesis pathway, leading to changes in the levels of sphingolipids, and subsequently, modulation of cell death. The de novo sphingolipid biosynthesis pathway includes a ceramide synthase-dependent reaction, giving rise to dihydroceramide, which is then converted in a desaturase-dependent reaction to ceramide. In this study we tested the hypothesis that combining Foscan-mediated PDT with desaturase inhibitor fenretinide (HPR) enhances cancer cell killing. We discovered that by subjecting SCC19 cells, a human head and neck squamous cell carcinoma cell line, to PDT+HPR resulted in enhanced accumulation of C16-dihydroceramide, not ceramide. Concomitantly, mitochondrial depolarization was enhanced by the combined treatment. Enhanced activation of caspase-3 after PDT+HPR was inhibited by FB. Enhanced clonogenic cell death after the combination was sensitive to FB, as well as Bcl2- and caspase inhibitors. Treatment of mouse SCCVII squamous cell carcinoma tumors with PDT+HPR resulted in improved long-term tumor cures. Overall, our data showed that combining PDT with HPR enhanced apoptotic cancer cell killing and antitumor efficacy of PDT. The data suggest the involvement of the de novo sphingolipid biosynthesis pathway in enhanced apoptotic cell killing after PDT+HPR, and identify the combination as a novel more effective anticancer treatment than either treatment alone.


Subject(s)
Apoptosis , Fenretinide/therapeutic use , Mesoporphyrins/therapeutic use , Photochemotherapy , Photosensitizing Agents/therapeutic use , Sphingolipids/biosynthesis , Cell Line, Tumor , Drug Therapy, Combination , Fenretinide/administration & dosage , Humans , Mesoporphyrins/administration & dosage , Photosensitizing Agents/administration & dosage
18.
Chem Phys Lipids ; 194: 110-116, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26408265

ABSTRACT

A ceramide commonly found in mammalian cells, C16-ceramide (N-palmitoyl-d-erythro-sphingosine), is capable of forming large, protein-permeable channels in the mitochondrial outer membrane (MOM). However, C16-ceramide is unable to permeabilize the plasma membrane of erythrocytes. This specificity is unexpected considering that ceramide forms channels in simple phosphoglycerolipid membranes. Synthetic analogs of C16-ceramide with targeted changes at each of the functional regions of the molecule including methylation, altered hydrocarbon chain length, and changes in the stereochemistry, were tested to probe the role of ceramide's molecular features on its ability to form channels in these two different membrane types. The ability to permeabilize the MOM was relatively insensitive to modifications of the various functional groups of ceramide whereas the same modifications resulted in plasma membrane permeabilization (a gain of function rather than a loss of function). Some analogs (ceramine, NBD-labeled ceramide, C18,1 ceramide) gained another function, the ability to inhibit cytochrome oxidase. The gain of deleterious functions indicates that constraints on the structure of ceramide that is formed by the cell's synthetic machinery includes the avoidance of deleterious interactions. We propose that the specific structure of ceramide limits the size of its interactome (both proteins and lipids) thus reducing the likelihood of unwanted side effects.


Subject(s)
Cell Membrane/metabolism , Ceramides/chemistry , Ceramides/metabolism , Mitochondrial Membranes/metabolism , Animals , Cell Membrane/chemistry , Erythrocytes/cytology , Mitochondria, Liver/metabolism , Mitochondrial Membranes/chemistry , Molecular Structure , Rats , Rats, Sprague-Dawley
19.
J Biol Chem ; 290(21): 13157-67, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25839235

ABSTRACT

A family of six ceramide synthases with distinct but overlapping substrate specificities is responsible for generation of ceramides with acyl chains ranging from ∼14-26 carbons. Ceramide synthase 6 (CerS6) preferentially generates C14- and C16-ceramides, and we have previously shown that down-regulation of this enzyme decreases apoptotic susceptibility. In this study, we further evaluated how increased CerS6 expression impacts sphingolipid composition and metabolism. Overexpression of CerS6 in HT29 colon cancer cells resulted in increased apoptotic susceptibility and preferential generation of C16-ceramide, which occurred at the expense of very long chain, saturated ceramides. These changes were also reflected in sphingomyelin composition. HT-CerS6 cells had increased intracellular levels of sphingosine, which is generated by ceramidases upon hydrolysis of ceramide. qRT-PCR analysis revealed that only expression of acid ceramidase (ASAH1) was increased. The increase in acid ceramidase was confirmed by expression and activity analyses. Pharmacological inhibition of JNK (SP600125) or curcumin reduced transcriptional up-regulation of acid ceramidase. Using an acid ceramidase promoter driven luciferase reporter plasmid, we demonstrated that CerS1 has no effect on transcriptional activation of acid ceramidase and that CerS2 slightly but significantly decreased the luciferase signal. Similar to CerS6, overexpression of CerS3-5 resulted in an ∼2-fold increase in luciferase reporter gene activity. Exogenous ceramide failed to induce reporter activity, while a CerS inhibitor and a catalytically inactive mutant of CerS6 failed to reduce it. Taken together, these results suggest that increased expression of CerS6 can mediate transcriptional activation of acid ceramidase in a JNK-dependent manner that is independent of CerS6 activity.


Subject(s)
Acid Ceramidase/metabolism , Apoptosis/drug effects , Ceramides/pharmacology , Colonic Neoplasms/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins/metabolism , Sphingosine N-Acyltransferase/metabolism , Acid Ceramidase/genetics , Antimetabolites, Antineoplastic/pharmacology , Blotting, Western , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Flow Cytometry , Fluorouracil/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Membrane Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sphingolipids/metabolism , Sphingosine N-Acyltransferase/genetics , Tumor Cells, Cultured
20.
Int J Oncol ; 46(5): 2003-10, 2015 May.
Article in English | MEDLINE | ID: mdl-25739041

ABSTRACT

Because photodynamic therapy (PDT) alone is not always effective as an anticancer treatment, PDT is combined with other anticancer agents for improved efficacy. The clinically-relevant fenretinide [N-(4-hydroxyphenyl) retinamide; 4HPR], was combined with the silicon phthalocyanine photosensitizer Pc4-mediated PDT to test for their potential to enhance killing of SCC17B cells, a clinically-relevant model of human head and neck squamous cell carcinoma. Because each of these treatments induces apoptosis and regulates the de novo sphingolipid (SL) biosynthesis pathway, the role of ceramide synthase, the pathway-associated enzyme, in PDT+4HPR-induced apoptotic cell death was determined using the ceramide synthase inhibitor fumonisin B1 (FB). PDT+4HPR enhanced loss of clonogenicity. zVAD-fmk, a pan-caspase inhibitor, and FB, protected cells from death post-PDT+4HPR. In contrast, the anti-apoptotic protein Bcl2 inhibitor ABT199 enhanced cell killing after PDT+4HPR. Combining PDT with 4HPR led to FB-sensitive, enhanced Bax associated with mitochondria and cytochrome c redistribution. Mass spectrometry data showed that the accumulation of C16-dihydroceramide, a precursor of ceramide in the de novo SL biosynthesis pathway, was enhanced after PDT+4HPR. Using quantitative confocal microscopy, we found that PDT+4HPR enhanced dihydroceramide/ceramide accumulation in the ER, which was inhibited by FB. The results suggest that SCC17B cells are sensitized to PDT by 4HPR via the de novo SL biosynthesis pathway and apoptosis, and imply potential clinical relevance of the combination for cancer treatment.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Fenretinide/therapeutic use , Head and Neck Neoplasms/pathology , Photochemotherapy/methods , Sphingolipids/biosynthesis , Tumor Stem Cell Assay/methods , Carcinoma, Squamous Cell/drug therapy , Combined Modality Therapy , Head and Neck Neoplasms/drug therapy , Humans , Microscopy, Confocal , Spectrometry, Mass, Electrospray Ionization , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...