Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Oncogene ; 29(22): 3297-306, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20305696

ABSTRACT

Biologically active membrane gangliosides, expressed and released by many human tumors, are hypothesized to significantly impact tumor progression. Lack of a model of complete and specific tumor ganglioside depletion in vivo, however, has hampered elucidation of their role. Here, we report the creation of a novel, stable, genetically induced tumor cell system resulting in specific and complete blockade of ganglioside synthesis. Wild-type (WT) and GM3 synthase/GM2 synthase double knockout (DKO) murine embryonic fibroblasts were transformed using amphotropic retrovirus-transduced oncogenes (pBABE-c-Myc(T58A)+H-RasG12V). The transformed cells, WT(t) and DKO(t) respectively, evidenced comparable integrated copy numbers and oncogene expression. Ganglioside synthesis was completely blocked in the DKO(t) cells, importantly without triggering an alternate pathway of ganglioside synthesis. Ganglioside depletion (to <0.5 nmol/10(7) cells from 9 to 11 nmol/10(7) WT(t) or untransfected normal fibroblasts) did not adversely affect cell proliferation kinetics but did reduce cell migration on fibronectin-coated wells, consistent with our previous observations in ganglioside-depleted normal human fibroblasts. Strikingly, despite similar oncogene expression and growth kinetics, DKO(t) cells evidenced significantly impaired tumor growth in syngeneic immunocompetent mice, underscoring the pivotal role of tumor cell gangliosides and providing an ideal system for probing their mechanisms of action in vivo.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gangliosides/metabolism , N-Acetylgalactosaminyltransferases/deficiency , Oncogenes , Sialyltransferases/deficiency , Animals , Cell Growth Processes/physiology , Cell Movement/physiology , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Gangliosides/biosynthesis , Gangliosides/genetics , Genes, myc , Genes, ras , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism
2.
Biomed Khim ; 52(3): 245-57, 2006.
Article in Russian | MEDLINE | ID: mdl-16898583

ABSTRACT

Tumor cell gangliosides are bioactive molecules involved in tumor-host interactions. To investigate their role in tumor formation and angiogenesis, we sought to develop an inhibitory model targeting human GM3 synthase, an essential enzyme in the ganglioside synthesis pathway, by antisense transfection. We prepared a number of transfectants from DAOY human medulloblastoma cells and isolated clones that stably expressed a 560-bp fragment of human GM3 synthase cDNA, in either sense or antisense orientation, as well as clones transfected with an empty vector. Both sense and antisense clones permanently incorporated mammalian expression vectors into their genomes. The DAOY cell clones were screened for ganglioside content using total lipid extraction, ganglioside isolation, and HPTLC. One antisense-transfected clone, 7.2A, in which total ganglioside content was reduced by 70%, was selected for further study. All sense- and sham-transfectants had ganglioside levels not different from that of untransfected DAOY cells. After 10 passages however, while antisense mRNA expression was fully maintained, the ganglioside content of 7.2A cells had reverted to normal levels. Antisense RNA transfection can sometimes have a reversible effect on the expression of a target. Possible regulatory mechanisms of this previously unrecognized process of reversion to wild type phenotype are discussed.


Subject(s)
Gangliosides/biosynthesis , Oligonucleotides, Antisense/pharmacology , Sialyltransferases/metabolism , Transfection , Cell Line, Tumor , Chromatography, Thin Layer , Humans , Medulloblastoma , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sialyltransferases/antagonists & inhibitors , Sialyltransferases/genetics
3.
Pediatr Blood Cancer ; 46(2): 228-33, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16047354

ABSTRACT

BACKGROUND: Diabetes insipidus (DI) is the most frequent central nervous system (CNS)-related permanent consequence in Langerhans cell histiocytosis (LCH), which mostly requires life-long hormone replacement therapy. In an attempt to define the population at risk for DI, 1,741 patients with LCH registered on the trials DALHX 83 and DALHX 90, LCH I and LCH II were studied. RESULTS: Overall 212 of 1,741 patients (12%) was reported to have DI. In 102 of 1,741 patients (6%) DI was present at diagnosis of LCH. One thousand one hundred eighty three of 1,539 patients without DI at diagnosis had follow up information. One hundred ten of these (9%) later developed DI. The risk of developing DI was 20% at 15 years after diagnosis. Multisystem disease patients at diagnosis carried a 4.6-fold risk for DI compared to single system patients. Craniofacial lesions, in particular in the "ear," "eye," and oral region were associated with a significantly increased risk for DI (relative hazard rate, RHR 1.7), independent of the extent of disease. No influence of the duration of therapy could be determined, but the duration of initial disease activity (RHR 1.5) and the occurrence of reactivations (RHR 3.5) significantly increased the risk for DI. CONCLUSIONS: Patients with multisystem disease and craniofacial involvement at diagnosis, in particular of the "ear," "eye," and the oral region carry a significantly increased risk to develop DI during their course. This risk is augmented when the disease remains active for a longer period or reactivates.


Subject(s)
Diabetes Insipidus, Neurogenic/pathology , Histiocytosis, Langerhans-Cell/pathology , Clinical Trials as Topic , Diabetes Insipidus, Neurogenic/complications , Diabetes Insipidus, Neurogenic/drug therapy , Female , Follow-Up Studies , Histiocytosis, Langerhans-Cell/drug therapy , Histiocytosis, Langerhans-Cell/etiology , Hormone Replacement Therapy , Humans , Male , Retrospective Studies , Risk Factors
4.
Br J Cancer ; 91(2): 389-97, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15187999

ABSTRACT

Recent findings link increased expression of the structurally complex 'b' pathway gangliosides GD1b, GT1b, GQ1b (CbG) to a favourable clinical and biological behaviour in human neuroblastoma (NB). Seeking a model to probe these observations, we evaluated four human NB cell lines. Very low CbG content (4-10%) in three of the four cell lines (LAN-5, LAN-1, SMS-KCNR) reflected the ganglioside pattern observed in the most aggressive NB tumours. Pharmacological alterations of complex ganglioside synthesis in vitro by a 5-7 day exposure to 5-10 microM retinoic acid, which is employed in maintenance therapy of disseminated NB, included markedly increased (i) relative expression of CbG (6.6+/-2.0-fold increase, P=0.037), (ii) relative expression of the analogous 'a' pathway gangliosides, termed CaG (6.4+/-1.4-fold increase in GM1a and GD1a; P=0.010), and (iii) total cellular ganglioside content (2.0-6.3-fold), which in turn amplified the accumulation of structurally complex gangliosides. Substantial increases (2.7-2.9-fold) in the activity of GD1b/GM1a synthase (beta-1,3-galactosyltransferase), which initiates the synthesis of CbG and CaG, accompanied the all-trans retinoic acid (ATRA)-induced ganglioside changes. Thus, increased CbG synthesis in NB cell lines is attributable to a specific effect of ATRA, namely induction of GD1b/GM1a synthase activity. Since the shift towards higher expression of CbG and CaG during retinoic acid-induced cellular differentiation reflects a ganglioside pattern found in clinically less-aggressive tumours, our studies suggest that complex gangliosides may play a role in the biological and clinical behaviour of NB.


Subject(s)
Antineoplastic Agents/pharmacology , Galactosyltransferases/metabolism , Gangliosides/metabolism , Glucosyltransferases/biosynthesis , Neuroblastoma/drug therapy , Tretinoin/pharmacology , Cell Differentiation , Humans , Neuroblastoma/metabolism , Tumor Cells, Cultured
5.
Bone Marrow Transplant ; 33(12): 1253-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15146169

ABSTRACT

We report here a monosomy 7 transformation of donor cells following matched-unrelated, same sex, allogeneic bone marrow transplantation in a patient with severe congenital aplastic anemia. A PCR technique was employed to amplify microsatellite markers on chromosome 7 to confirm donor/recipient identity. We found that the transformation of monosomy 7 occurred in previously genetically normal donor cells. This study suggests that the microenvironment of the bone marrow of our patient with severe congenital aplastic anemia may have played a critical role in the development of monosomy 7 of normal donor cells and we conclude that chromosomal microsatellite marker analysis can be a valuable tool for precise donor/recipient differentiation in engraftment monitoring.


Subject(s)
Anemia, Aplastic/therapy , Bone Marrow Transplantation , Cell Transformation, Neoplastic/genetics , Monosomy , Tissue Donors , Anemia, Aplastic/complications , Anemia, Aplastic/congenital , Chromosomes, Human, Pair 7 , Cytogenetic Analysis , Female , Humans , Infant , Polymerase Chain Reaction , Transplantation, Homologous
7.
J Biol Chem ; 276(46): 42782-92, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11535585

ABSTRACT

In a recent study, inhibition of cellular ganglioside synthesis blocked growth factor-induced fibroblast proliferation. Conversely, enrichment of cell membrane gangliosides by ganglioside preincubation enhanced growth factor-elicited cell proliferation. In the absence of serum and growth factors, NeuNAcalpha2-3Galbeta1-3GalNAcbeta1-4(NeuNAcalpha2-3)Galbeta1-4Glcbeta1-1Cer (G(D1a)) acted like a growth factor when cells were pretreated with the ganglioside, stimulating proliferation of normal human dermal fibroblasts and Swiss 3T3 fibroblasts. In contrast, growth inhibition was observed when high concentrations of gangliosides were continuously present in the culture medium during incubation of fibroblasts with growth factors (Li, R., Manela, J., Kong, Y., and Ladisch, S. (2000) J. Biol. Chem. 275, 34213-34223). Here, we investigated the mechanisms whereby gangliosides elicit proliferation-coupled signaling in normal human dermal fibroblasts. Incubation of the fibroblasts with G(D1a) enhanced epidermal growth factor (EGF) receptor autophosphorylation and Ras and MAPK activation in a dose-dependent manner. Exposure of the cells to G(D1a) also enhanced the phosphorylation of Elk-1 by the activated MAPK. Brief pretreatment of the cells with PD98059 blocked the enhancing effect of gangliosides on EGF-induced MAPK activation. In the absence of serum and growth factors, G(D1a) incubation induced phosphorylation of Src kinase, Ras activation, and phosphorylation of MAPK and Elk-1 in a dose-dependent manner. The activation of Src kinase was confirmed by enhanced Src kinase activity. Brief treatment of the cells with PP1 blocked the activation of Src kinase and MAPK. Again, PD98059 treatment inhibited ganglioside-elicited MAPK phosphorylation. Among the gangliosides tested, G(D1a), was the most active molecule, whereas lactosylceramide was the least active one, indicating relative structural specificity of the ganglioside action. In conclusion, gangliosides promote fibroblast proliferation through enhancement of growth factor signaling and activation of Src kinase.


Subject(s)
Epidermal Growth Factor/metabolism , Gangliosides/metabolism , Signal Transduction , src-Family Kinases/metabolism , Blotting, Western , CSK Tyrosine-Protein Kinase , Cell Division , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Flavonoids/pharmacology , Guanosine Triphosphate/metabolism , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Time Factors
8.
Int J Cancer ; 93(1): 131-8, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11391632

ABSTRACT

As a strategy to enhance tumor cell sensitivity to vincristine, we tested whether modulation of sphingolipid metabolism would alter vincristine cytotoxicity since this is linked to accumulation of the intermediate metabolite, ceramide. We blocked ceramide metabolism in a series of variably vincristine-resistant cell lines derived from CCRF-CEM leukemia cells using an inhibitor of glucosylceramide synthase, DL-threo-phenyl-2-hexadecanoylamino-3-pyrrolidino-1-propanol (PPPP). PPPP alone (1.0 microM), while nearly completely blocking glucosylceramide synthesis, was not toxic and did not increase cellular ceramide levels. Vincristine alone was toxic, caused apoptosis or programmed cell death (PCD) and caused an elevation in ceramide levels. Strikingly, the combination of PPPP and vincristine resulted in a further increase, over that of vincristine alone, of (i) cellular ceramide concentration, (ii) cytotoxicity associated with PCD and (iii) G2/M cell-cycle arrest. PPPP had no effect on P-glycoprotein expression or function. We conclude that vincristine cytotoxicity occurs in part through a ceramide-dependent mechanism, resulting in both G2/M block as well as PCD, and that the blockade of glucosylceramide synthase, in itself not toxic, causes augmented accumulation of ceramide resulting from vincristine exposure, which in turn maximizes ceramide-dependent, vincristine-induced cytotoxicity. Inhibition of glucosylceramide synthesis may be a means of circumventing drug resistance by enhancing signaling through a cell-death pathway.


Subject(s)
Cell Survival/physiology , Enzyme Inhibitors/pharmacology , Glucosyltransferases/metabolism , Propanolamines/pharmacology , Pyrrolidines/pharmacology , Vincristine/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Survival/drug effects , Drug Resistance, Multiple , Glucosyltransferases/antagonists & inhibitors , Humans , Leukemia, T-Cell , Sphingolipids/metabolism , Tumor Cells, Cultured
9.
Biochem Biophys Res Commun ; 282(4): 1031-7, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11352656

ABSTRACT

Tumor progression requires normally quiescent endothelial cells to form new vascular networks. This angiogenesis is dependent upon several soluble factors, prominent among which is vascular endothelial growth factor (VEGF). Other tumor-associated molecules, such as gangliosides, sialic acid-containing glycosphingolipids expressed by tumor cells and shed into the tumor microenvironment, may also modulate tumor angiogenesis. Here we assessed the influence of a highly purified ganglioside, G(D1a), on responses of normal human umbilical vein endothelial cells (HUVEC) to VEGF. Preincubation of HUVEC with G(D1a) enhanced VEGF-induced cell proliferation; 10 microM G(D1a) caused a twofold increase in DNA synthesis. The migration of HUVEC across a VEGF gradient was also enhanced by 50%, even with only a brief (1 h) preexposure of the cells to the same concentration of G(D1a). These findings suggest that gangliosides shed by tumor cells can promote tumor angiogenesis by enhancing the VEGF response of endothelial cells in the tumor microenvironment.


Subject(s)
Cell Movement/drug effects , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Gangliosides/pharmacology , Lymphokines/pharmacology , Cell Division/drug effects , Cells, Cultured , DNA/biosynthesis , Dose-Response Relationship, Drug , Drug Synergism , Endothelium, Vascular/cytology , Humans , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
J Pediatr ; 138(5): 728-34, 2001 May.
Article in English | MEDLINE | ID: mdl-11343051

ABSTRACT

OBJECTIVE: To compare 2 active agents, vinblastine and etoposide, in the treatment of multisystem Langerhans' cell histiocytosis (LCH) in an international randomized study. STUDY DESIGN: One hundred forty-three untreated patients were randomly assigned to receive 24 weeks of vinblastine (6 mg/m(2), given intravenously every week) or etoposide (150 mg/m(2)/d, given intravenously for 3 days every 3 weeks), and a single initial dose of corticosteroids. RESULTS: Vinblastine and etoposide were equivalent (P > or = .2) in all respects: response at week 6 (57% and 49%); response at the last evaluation (58% and 69%); toxicity (47% and 58%); and probability of survival (76% and 83%) [corrected], of disease reactivation (61% and 55%), and of developing permanent consequences (39% and 51%) including diabetes insipidus (22% and 23%). LCH reactivations were usually mild, as was toxicity. All children > or = 2 years old without risk organ involvement (liver, lungs, hematopoietic system, or spleen) survived. With such involvement, lack of rapid (within 6 weeks) response was identified as a new prognostic indicator, predicting a high (66%) mortality rate. CONCLUSIONS: Vinblastine and etoposide, with one dose of corticosteroids, are equally effective treatments for multisystem LCH, but patients who do not respond within 6 weeks are at increased risk for treatment failure and may require different therapy.


Subject(s)
Etoposide/therapeutic use , Histiocytosis, Langerhans-Cell/drug therapy , Methylprednisolone/therapeutic use , Vinblastine/therapeutic use , Adolescent , Child , Drug Therapy, Combination , Etoposide/adverse effects , Humans , Risk Assessment , Survival Analysis , Treatment Outcome , Vinblastine/adverse effects
11.
Cancer ; 91(4): 785-93, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11241247

ABSTRACT

BACKGROUND: Gangliosides are membrane-bound glycolipid molecules particularly prominent in neural tissue. Changes in ganglioside expression during embryologic development result from a shift in biosynthesis from the fetal b pathway to the adult a pathway. Tumor gangliosides may play a role in the clinical behavior of certain subtypes of neuroblastoma. Because neuroblastoma, which presents in infancy, has a different biologic and clinical phenotype than that which presents in older children, the authors determined whether differences in ganglioside biosynthesis exist between these two neuroblastoma subgroups. METHODS: Sixty-eight tumor specimens (25 diagnosed by screening and 43 diagnosed clinically) were obtained from the Quebec Neuroblastoma Screening Project. Gangliosides were isolated and purified by solvent partitioning, separated by high performance thin-layer chromatography, and quantitated by scanning densitometry. The sum of a and b pathway gangliosides were determined for each tumor. RESULTS: Gangliosides of the b (fetal) pathway predominated in both screened and clinically diagnosed tumors of patients younger than 1 year of age. Twenty-three of 25 screened patients (92%) and 21 of 23 patients with clinically diagnosed tumors at younger than 1 year of age (91%) had tumor b pathway ganglioside content greater than 60%. In contrast, tumors of only 8 of 20 patients 1 year or older (40%) had b pathway ganglioside predominance. Predominance of b pathway tumor gangliosides correlated with improved outcome. Event free survival was significantly higher among patients with b pathway ganglioside tumor content greater than 60% versus those with b pathway ganglioside tumor content less than 60% (118.1 +/- 3.9 months vs. 69.2 +/- 8.6 months, P < 0.01). CONCLUSIONS: Fetal patterns of ganglioside biosynthesis predominate in neuroblastoma tumors from patients younger than 1 year of age and adult patterns of ganglioside biosynthesis predominate in tumors from older children, supporting the view that neuroblastoma consists of distinct but overlapping disorders, and that gangliosides may play a biologic role in the clinical differences among these patients.


Subject(s)
Gangliosides/metabolism , Neuroblastoma/metabolism , Chromatography, Thin Layer , Humans , Infant , Infant, Newborn , Neuroblastoma/diagnosis , Neuroblastoma/pathology , Statistics, Nonparametric , Survival Analysis
12.
Anticancer Res ; 21(6A): 3785-91, 2001.
Article in English | MEDLINE | ID: mdl-11911248

ABSTRACT

BACKGROUND: Integrin alpha v promotes brain microvessel endothelium survival, yet its role in brain tumor cells is unknown. MATERIALS AND METHODS: Alpha v synthesis in medulloblastoma cells Daoy, D341Med, and D283Med, was inhibited with antisense oligonucleotides (ASODN) to test the effect on growth and survivaL RESULTS: ASODN reduced alpha v surface expression 75% in a dose- and time-dependent manner (2 microM, 72 hours). Alpha v-deficient cells grown with vitronectin demonstrated reduced cell spreading, G0-G1 growth arrest, decreased proliferation and increased apoptosis compared to controls or alpha v-deficient cells grown with collagen. Furthermore, insulin-like growth factor-I (IGF-I) suboptimally stimulated proliferation and survival of alpha v-deficient cells, suggesting that alpha v-IGF-I interactions potentiate medulloblastoma growth. Finally, treatment with alpha v-blocking antibody induced caspase-8 and caspase-3 expression, while apoptosis of alpha v-deficient cells was associated only with increased caspase-3. CONCLUSION: Alpha v integrin supports medulloblastoma growth by activating adhesion-dependent and -independent survival pathways and thus may serve as a novel therapeutic target in this tumor.


Subject(s)
Antigens, CD/biosynthesis , Antigens, CD/genetics , Apoptosis/drug effects , Medulloblastoma/metabolism , Medulloblastoma/pathology , Oligonucleotides, Antisense/pharmacology , Apoptosis/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Growth Inhibitors/pharmacology , Humans , Integrin alphaV , Tumor Cells, Cultured
14.
Biochim Biophys Acta ; 1486(2-3): 265-74, 2000 Jul 19.
Article in English | MEDLINE | ID: mdl-10903477

ABSTRACT

Tumor cell ganglioside shedding has been implicated in the process of tumor formation. Previously, we identified three forms of tumor ganglioside shedding: micelles, monomers and membrane vesicles. Here, we have explored the membrane vesicle form of ganglioside shedding, using a newly identified human ovarian carcinoma cell line, CABA I. These cells synthesize and express a spectrum of gangliosides, including the disialoganglioside, G(D3). Immunostaining using the monoclonal antibody R24 confirmed G(D3) expression and its presence in the plasma membrane of these cells. Cellular gangliosides were detected in the culture supernatant by HPTLC autoradiography, confirming an active shedding rate of 3% of cellular gangliosides/24 h. CABA I cell membranes also express caveolin-1, a characteristic protein marker for caveolae, which was detected by flow cytometric analysis and by Western blotting in both the cell membranes and the isolated membrane vesicles. To further define the expression of G(D3) and caveolin-1, we used immunogold electron microscopy. This revealed localization of G(D3) in small clusters in the plasma membrane as well as enrichment and localization of ganglioside G(D3) and caveolin-1 in shed membrane vesicles, with 58-78% of vesicles carrying both G(D3) and caveolin-1. Together, these results suggest that membrane vesicle shedding originates in plasma membrane domains enriched in gangliosides and caveolin-1.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/analysis , Biomarkers, Tumor/analysis , Caveolins , Cell Membrane/metabolism , Gangliosides/analysis , Membrane Proteins/analysis , Ovarian Neoplasms/metabolism , Caveolin 1 , Female , Flow Cytometry , Humans , Immunohistochemistry , Microscopy, Immunoelectron , Tumor Cells, Cultured
15.
J Biol Chem ; 275(44): 34213-23, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10859325

ABSTRACT

Cell surface gangliosides have been proposed as modulators of transmembrane signaling. In this study, we used two complementary approaches to investigate the function of cellular gangliosides in the response of mammalian fibroblasts to growth factors. First, inhibition of glucosyl ceramide synthase by a new specific inhibitor of d-l-threo-1-phenyl-2-hexadecanoylamino-3 -pyrrolidino-1-propanol-HC l (glucosylceramide synthase), which depletes cellular gangliosides at a concentration of 1 microm without causing an increase in ceramide levels, blocked epidermal growth factor-stimulated proliferation of fibroblasts. Similarly, responses to several other growth factors that activate receptor tyrosine kinases, including fibroblast growth factor, insulin-like growth factor-I, and platelet-derived growth factor, were inhibited by 50-100%. Conversely, enrichment of cellular gangliosides by preincubation of the mouse and human fibroblasts with exogenously added gangliosides enhanced growth factor-elicited cell proliferation. Novel findings of this study, distinguishing it from previous similar studies, include differential effects of preincubation versus continuous incubation of cells with gangliosides on growth factor-dependent cell proliferation and the growth factor-like action of NeuNAc alpha 2-3Gal beta 1-3GalNAc beta 1-4(NeuNAc alpha 2-3)Gal beta 1-4Glc beta 1-1Cer when cells are pretreated with the ganglioside.


Subject(s)
Cell Division/physiology , Gangliosides/physiology , Growth Substances/physiology , 3T3 Cells , Animals , Ceramides/metabolism , Down-Regulation , Fibroblasts/cytology , Humans , Mice , Peptidoglycan/metabolism , Sphingomyelins/metabolism
16.
J Natl Cancer Inst ; 92(11): 912-7, 2000 Jun 07.
Article in English | MEDLINE | ID: mdl-10841826

ABSTRACT

BACKGROUND: Gangliosides are immunosuppressive cell surface molecules that are often present in high concentrations in and shed actively by tumor cells. These molecules inhibit the antitumor immune response that is implicated in tumor rejection. We therefore determined the ability of tumor cells pharmacologically depleted of gangliosides to form tumors in mice. METHODS: We tested a ganglioside-rich subline of B16 murine melanoma, MEB4, and MEB4 cells that had been depleted of endogenous gangliosides by incubation with 0.5 microM 1-phenyl-2-hexadecanoylamino-3-pyrrolidino-1-propanol, a specific inhibitor of the enzyme glucosylceramide synthase. Tumor formation was assessed twice a week for 10 weeks after the intradermal injection of tumor cells, and metastatic potential was assessed 4 weeks after tail vein injection of tumor cells. All P values are from two-sided tests. RESULTS: Reduction of the ganglioside content of MEB4 cells, which was not cytotoxic to cells and did not inhibit cell proliferation in vitro, markedly reduced their ability to form tumors. Only 40% of the mice given an intradermal injection of 10(5) ganglioside-depleted MEB4 cells developed tumors compared with 100% of the mice given an injection of 10(5) control MEB4 cells (P<.001). Ganglioside depletion also reduced metastasis: A mean of five pulmonary metastases was detected per mouse given an injection of 2 x 10(5) ganglioside-depleted MEB4 cells compared with a mean of 25 per mouse given an injection of 2 x 10(5) control MEB4 cells. CONCLUSION: Tumor cells with a pharmacologically decreased concentration of gangliosides produce fewer tumors in mice than do untreated cells, suggesting that pharmacologic depletion of gangliosides should be explored further as a therapeutic approach to cancer.


Subject(s)
Enzyme Inhibitors/pharmacology , Gangliosides/metabolism , Glucosyltransferases/antagonists & inhibitors , Melanoma, Experimental/metabolism , Propanolamines/pharmacology , Pyrrolidines/pharmacology , Animals , Gangliosides/biosynthesis , Mice , Tumor Cells, Cultured
17.
Med Pediatr Oncol ; 33(5): 482-5, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10531573

ABSTRACT

BACKGROUND: Treatment of multisystem Langerhans cell histiocytosis (LCH) remains difficult. Various regimens of single and multiagent chemotherapy have been used, but a significant proportion of patients fail to respond to treatment. PROCEDURE: We have evaluated the use of cyclosporine A (CSA) in a controlled group of patients, who had received a systematic primary therapy (LCH-I). Patients received CSA either as a single agent (10 patients) or in combination with vinblastine, etoposide, prednisolone, and/or antithymocyte globulin (16 patients). RESULTS: Among the total of 26 patients treated, a single patient developed a complete response and three a partial response, whereas 85% (22 patients) had no response to CSA. CONCLUSIONS: CSA is at best of limited value in the treatment of patients with multisystem LCH, particularly those who had progressive disease while receiving chemotherapy.


Subject(s)
Antifungal Agents/therapeutic use , Cyclosporine/therapeutic use , Histiocytosis, Langerhans-Cell/drug therapy , Adolescent , Antifungal Agents/pharmacology , Child , Child, Preschool , Cyclosporine/pharmacology , Disease Progression , Female , Histiocytosis, Langerhans-Cell/pathology , Humans , Infant , Male , Retrospective Studies , Treatment Outcome
18.
J Immunol ; 163(7): 3718-26, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10490967

ABSTRACT

Tumor gangliosides are highly immunosuppressive membrane glycosphingolipids that are shed into the tumor cell microenvironment. We directly tested the impact of shed gangliosides on the in vivo antitumor immune response in a syngeneic fully autochthonous system (FBL-3 erythroleukemia cells, C57BL/6 mice, and highly purified FBL-3 cell gangliosides). The major FBL-3 ganglioside was identified as GM1b by mass spectrometry. Substantial ganglioside shedding (90 pmol/108 cells/h), a requisite for their inhibition of the immune function of tumor-infiltrating leukocytes, was detected. Immunosuppression by FBL-3 gangliosides was potent; 5-20 microM inhibited the tumor-specific secondary proliferative response (80-100%) and suppressed the generation of tumor-specific CTLs (97% reduction of FBL-3 cell lysis at an E:T ratio of 100:1). In vivo, coinjection of 10 nmol of FBL-3 gangliosides with a primary FBL-3 cell immunization led to a reduced response to a secondary challenge (the increase in the draining popliteal lymph node mass, cell number, and lymphocyte thymidine incorporation were lowered by 70, 69, and 72%, respectively). Coinjection of gangliosides with a secondary tumor challenge led to a 61, 74, and 42% reduction of the increase in lymph node mass, cell number, and thymidine uptake and a 63-74% inhibition of the increase of draining lymph node T cells (CD3+), B cells (CD19+), and dendritic cells/macrophages (Mac-3+). Overall, the clear conclusion that tumor-derived gangliosides inhibit syngeneic antitumor immune responses implicates these molecules as a potent factor in promoting tumor formation and progression.


Subject(s)
Gangliosides/immunology , Immunosuppressive Agents/pharmacology , Leukemia, Erythroblastic, Acute/immunology , Leukemia, Erythroblastic, Acute/prevention & control , Animals , Cell Division/drug effects , Cell Division/immunology , Cytotoxicity, Immunologic/drug effects , Female , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/immunology , G(M1) Ganglioside/isolation & purification , G(M1) Ganglioside/physiology , Gangliosides/physiology , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Lymphocyte Subsets/cytology , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/metabolism
19.
J Biol Chem ; 274(30): 21121-7, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10409665

ABSTRACT

Ceramide is a newly discovered second messenger that has been shown to cause cell growth arrest and apoptosis. Here, we present evidence that exogenously added C(6)-ceramide induces enlargement of late endosomes and lysosomes. 10 microM C(6)-ceramide caused the formation of numerous vesicles of varying sizes (2-10 micrometers) in fibroblasts (3T3-L1 and 3T3-F442A), without toxic effects. Vesicle formation induced by C(6)-ceramide was time- and dose-dependent, rapid, and reversible. Numerous small vesicles appeared within 8 h of treatment with 10 microM C(6)-ceramide. They enlarged with time, with large vesicles found in the perinuclear region and small ones observed at the cell periphery. Within 24 h of treatment, approximately 30% of the cells exhibited these vesicles. Removal of ceramide from the culture medium caused disappearance of the vesicles, which reappeared upon readdition of ceramide. Confocal immunofluorescence microscopic analysis using an anti-lysosome-associated membrane protein antibody identified the enlarged vesicles as late endosomes/lysosomes. The fluorescent C(6)-NBD-ceramide, a vital stain for the Golgi apparatus, did not stain these vesicles. The effect on vesicle formation was influenced by ceramide structure; D-erythro-C(6)-ceramide was the most active ceramide analogue tested. Short chain ceramide metabolites, such as sphingosine, sphingosine 1-phosphate, N-hexanoyl-sphingosylphosphorylcholine, N-acetylpsychosine, and C(2)-ceramide G(M3), (G(M3), N-acetylneuraminosyl-alpha(2, 3)-galactosyl-beta(1,4)-glucosylceramide), were inactive in causing vesicle formation when added exogenously. Together, these studies demonstrate that exogenous C(6)-ceramide induces endocytic vesicle formation and causes enlarged late endosomes and lysosomes in mouse fibroblasts.


Subject(s)
Ceramides/pharmacology , Endocytosis/drug effects , 3T3 Cells , Animals , Endosomes/drug effects , Endosomes/ultrastructure , Lysosomes/drug effects , Lysosomes/ultrastructure , Mice , Microscopy, Confocal , Time Factors
20.
Biochim Biophys Acta ; 1394(1): 43-56, 1998 Oct 02.
Article in English | MEDLINE | ID: mdl-9767101

ABSTRACT

Gangliosides shed by tumor cells are immunosuppressive molecules, but the mechanisms of shedding are poorly understood. We therefore conducted a comprehensive study of shedding to identify the natural forms of shed gangliosides. By chemical detection and mass spectrometric analysis of the gangliosides of YAC-1 murine lymphoma cells, we first confirmed that all major ganglioside species are released. Then, by the combination of metabolic and cell surface radiolabeling, we further demonstrated that gangliosides are released directly from the cell plasma membrane, i.e. by shedding. Ultracentrifugation separated the conditioned medium of metabolically radiolabeled cells cultured in either serum-free or serum-containing medium into: (1) a pellet of 100-200 nm membrane vesicles (visualized by electron microscopy) containing nearly one-third of total shed gangliosides; and (2) the supernatant, which contained soluble gangliosides (two-thirds of the total shed gangliosides). Although the ganglioside concentration in the conditioned medium (6-14x10-8 M) was above the critical micelle concentration of purified YAC-1 gangliosides (<1x10-8 M), by gel filtration >90% of the soluble gangliosides were found in monomeric form (MW <2 kDa) and only <10% in micelles (130 kDa). Ultrafiltration of fresh conditioned medium likewise showed the existence of monomers, and the findings were confirmed in human Daoy medulloblastoma and mouse MEB4 melanoma cells. Thus, in their natural states, shed tumor cell gangliosides exist in three forms: membrane vesicles, micelles, and monomers.


Subject(s)
Gangliosides/analysis , Neoplasms/chemistry , Animals , Chromatography, Gel , Humans , Lymphoma/metabolism , Mice , Microscopy, Electron , Neuroblastoma/chemistry , Tumor Cells, Cultured , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL