1
|
Suchanski J, Reza S, Urbaniak A, Woldanska W, Kocbach B, Ugorski M. Galactosylceramide Upregulates the Expression of the BCL2 Gene and Downregulates the Expression of TNFRSF1B and TNFRSF9 Genes, Acting as an Anti-Apoptotic Molecule in Breast Cancer Cells. Cancers (Basel) 2024; 16:389. [PMID: 38254878 PMCID: PMC10813928 DOI: 10.3390/cancers16020389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/27/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Galactosylceramide (GalCer) increases the resistance of breast cancer cells to doxorubicin, paclitaxel, and cisplatin by acting as an anti-apoptotic molecule. GalCer was found to specifically downregulate the levels of the pro-apoptotic TNFRSF1B and TNFRSF9 genes and upregulate the levels of the anti-apoptotic BCL2 gene, suggesting that this glycosphingolipid regulates their expression at the transcriptional level. Consistent with this hypothesis, MDA-MB-231 and MCF7 breast cancer cells with high levels of GalCer showed lower activity of the TNFRSF1B and TNFRSF9 promoters than cells lacking GalCer. In contrast, the activity of the BCL2 promoter was higher in MCF7 cells overproducing GalCer than in MCF7 cells without GalCer. However, no difference in BCL2 promoter activity was observed between MDA-MB-231 cells with high and no GalCer content. Instead, we found that high levels of GalCer increased the stability of Bcl-2 mRNA. Subsequent studies showed that breast cancer cells with high levels of GalCer are characterized by significantly lower expression of P53. Importantly, inhibition of P53 expression by siRNA in MCF7 and MDA-MB-231 cells lacking GalCer resulted in decreased expression and promoter activity of the TNFRS1B and TNFRSF9 genes. On the other hand, increased expression and promoter activity of the BCL2 gene was found in such MCF7 cells, and increased stability of Bcl-2 transcripts was observed in such MDA-MB-231 cells. Taken together, these data strongly suggest that the regulatory protein that simultaneously increases the expression of the TNFRSF1B and TNFRSF9 genes and decreases the expression of the BCL2 gene and the stability of Bcl-2 transcripts is most likely P53, the expression of which is GalCer dependent.
Collapse
Affiliation(s)
| | | | | | | | | | - Maciej Ugorski
- Department of Biochemistry and Molecular Biology, Wroclaw University of Environmental and Life Sciences, C. K. Norwida 31, 50-375 Wroclaw, Poland; (J.S.)
| |
Collapse
|
2
|
Sarkar A, Banerjee S, Biswas K. Multi-dimensional role of gangliosides in modulating cancer hallmarks and their prospects in targeted cancer therapy. Front Pharmacol 2023; 14:1282572. [PMID: 38089042 PMCID: PMC10711107 DOI: 10.3389/fphar.2023.1282572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/24/2023] [Accepted: 11/14/2023] [Indexed: 12/10/2024] Open
Abstract
Gangliosides are glycosphingolipids with prevalence in nervous tissue and their involvement in certain neuronal diseases have been widely known. Interestingly, many recent studies highlighted their importance in the development and progression of various cancers through orchestration of multiple attributes of tumorigenesis, i.e., promoting migration, invasion, escaping the host immune system, and influencing other cancer hallmarks. Therefore, the multidimensional role of gangliosides in different cancers has established them as potential cancer targets. However, the tremendous structural complexity and functional heterogeneity are the major challenges in ganglioside research. Moreover, despite numerous immunotherapeutic attempts to target different gangliosides, it has failed to yield consistent results in clinical trials owing to their poor immunogenicity, a broad range of cross-reactivity, severe side effects, lack of uniform expression as well as heterogeneity. The recent identification of selective O-acetylated ganglioside expression in cancer tissues, but not in normal tissues, has strengthened their potential as a better and specific target for treating cancer patients. It was further supported by reduced cross-reactivity and side effects in clinical trials, although poor immunogenicity remains a major concern. Therefore, in addition to characterization and identification of the biological importance of O-acetylated gangliosides, their specific and efficient targeting in cancer through engineered antibodies is an emerging area of glycobiology research. This review highlights the modulatory effect of select gangliosides on different hallmarks of cancer and presents the overall development of ganglioside targeted immunotherapies along with recent progress. Here, we have also discussed its potential for future modifications aimed towards improvement in ganglioside-based cancer therapies.
Collapse
Affiliation(s)
| | | | - Kaushik Biswas
- Department of Biological Sciences, Bose Institute, Kolkata, India
| |
Collapse
|
3
|
van der Haar Àvila I, Windhouwer B, van Vliet SJ. Current state-of-the-art on ganglioside-mediated immune modulation in the tumor microenvironment. Cancer Metastasis Rev 2023; 42:941-958. [PMID: 37266839 PMCID: PMC10584724 DOI: 10.1007/s10555-023-10108-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/23/2023] [Accepted: 05/13/2023] [Indexed: 06/03/2023]
Abstract
Gangliosides are sialylated glycolipids, mainly present at the cell surface membrane, involved in a variety of cellular signaling events. During malignant transformation, the composition of these glycosphingolipids is altered, leading to structural and functional changes, which are often negatively correlated to patient survival. Cancer cells have the ability to shed gangliosides into the tumor microenvironment, where they have a strong impact on anti-tumor immunity and promote tumor progression. Since most ganglioside species show prominent immunosuppressive activities, they might be considered checkpoint molecules released to counteract ongoing immunosurveillance. In this review, we highlight the current state-of-the-art on the ganglioside-mediated immunomodulation, specified for the different immune cells and individual gangliosides. In addition, we address the dual role that certain gangliosides play in the tumor microenvironment. Even though some ganglioside species have been more extensively studied than others, they are proven to contribute to the defense mechanisms of the tumor and should be regarded as promising therapeutic targets for inclusion in future immunotherapy regimens.
Collapse
Affiliation(s)
- Irene van der Haar Àvila
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, the Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Britt Windhouwer
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, the Netherlands
| | - Sandra J van Vliet
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, the Netherlands.
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands.
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Cao S, Hu X, Ren S, Wang Y, Shao Y, Wu K, Yang Z, Yang W, He G, Li X. The biological role and immunotherapy of gangliosides and GD3 synthase in cancers. Front Cell Dev Biol 2023; 11:1076862. [PMID: 36824365 PMCID: PMC9941352 DOI: 10.3389/fcell.2023.1076862] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/22/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Gangliosides are a large subfamily of glycosphingolipids that broadly exist in the nervous system and interact with signaling molecules in the lipid rafts. GD3 and GD2 are two types of disialogangliosides (GDs) that include two sialic acid residues. The expression of GD3 and GD2 in various cancers is mostly upregulated and is involved in tumor proliferation, invasion, metastasis, and immune responses. GD3 synthase (GD3S, ST8SiaI), a subclass of sialyltransferases, regulates the biosynthesis of GD3 and GD2. GD3S is also upregulated in most tumors and plays an important role in the development and progression of tumors. Many clinical trials targeting GD2 are ongoing and various immunotherapy studies targeting gangliosides and GD3S are gradually attracting much interest and attention. This review summarizes the function, molecular mechanisms, and ongoing clinical applications of GD3, GD2, and GD3S in abundant types of tumors, which aims to provide novel targets for future cancer therapy.
Collapse
Affiliation(s)
- Shangqi Cao
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xu Hu
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Shangqing Ren
- 2Robotic Minimally Invasive Surgery Center, Sichuan Academy of Medical Sciences and Sichuan Provincial Peoples Hospital, Chengdu, China
| | - Yaohui Wang
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yanxiang Shao
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Kan Wu
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Zhen Yang
- 3Department of Urology, Chengdu Second People’s Hospital, Chengdu, China
| | - Weixiao Yang
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Gu He
- 4State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China,*Correspondence: Gu He, ; Xiang Li,
| | - Xiang Li
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China,*Correspondence: Gu He, ; Xiang Li,
| |
Collapse
|
5
|
Gangliosides as Signaling Regulators in Cancer. Int J Mol Sci 2021; 22:ijms22105076. [PMID: 34064863 PMCID: PMC8150402 DOI: 10.3390/ijms22105076] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/18/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
At the plasma membrane, gangliosides, a group of glycosphingolipids, are expressed along with glycosphingolipids, phospholipids, and cholesterol in so-called lipid rafts that interact with signaling receptors and related molecules. Most cancers present abnormalities in the intracellular signal transduction system involved in tumor growth, invasion, and metastasis. To date, the roles of gangliosides as regulators of signal transduction have been reported in several cancer types. Gangliosides can be expressed by the exogenous ganglioside addition, with their endogenous expression regulated at the enzymatic level by targeting specific glycosyltransferases. Accordingly, the relationship between changes in the composition of cell surface gangliosides and signal transduction has been investigated by controlling ganglioside expression. In cancer cells, several types of signaling molecules are positively or negatively regulated by ganglioside expression levels, promoting malignant properties. Moreover, antibodies against gangliosides have been shown to possess cytotoxic effects on ganglioside-expressing cancer cells. In the present review, we highlight the involvement of gangliosides in the regulation of cancer cell signaling, and we explore possible therapies targeting ganglioside-expressing cancer.
Collapse
|
6
|
Affiliation(s)
- Ross Mikkelsen
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA 23298-0058, USA.
| |
Collapse
|
7
|
Exogenous and Endogeneous Disialosyl Ganglioside GD1b Induces Apoptosis of MCF-7 Human Breast Cancer Cells. Int J Mol Sci 2016; 17:ijms17050652. [PMID: 27144558 PMCID: PMC4881478 DOI: 10.3390/ijms17050652] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/22/2016] [Revised: 04/13/2016] [Accepted: 04/21/2016] [Indexed: 11/16/2022] Open
Abstract
Gangliosides have been known to play a role in the regulation of apoptosis in cancer cells. This study has employed disialyl-ganglioside GD1b to apoptosis in human breast cancer MCF-7 cells using exogenous treatment of the cells with GD1b and endogenous expression of GD1b in MCF-7 cells. First, apoptosis in MCF-7 cells was observed after treatment of GD1b. Treatment of MCF-7 cells with GD1b reduced cell growth rates in a dose and time dependent manner during GD1b treatment, as determined by XTT assay. Among the various gangliosides, GD1b specifically induced apoptosis of the MCF-7 cells. Flow cytometry and immunofluorescence assays showed that GD1b specifically induces apoptosis in the MCF-7 cells with Annexin V binding for apoptotic actions in early stage and propidium iodide (PI) staining the nucleus of the MCF-7 cells. Treatment of MCF-7 cells with GD1b activated apoptotic molecules such as processed forms of caspase-8, -7 and PARP (Poly(ADP-ribose) polymerase), without any change in the expression of mitochondria-mediated apoptosis molecules such as Bax and Bcl-2. Second, to investigate the effect of endogenously produced GD1b on the regulation of cell function, UDP-gal: β1,3-galactosyltransferase-2 (GD1b synthase, Gal-T2) gene has been transfected into the MCF-7 cells. Using the GD1b synthase-transfectants, apoptosis-related signal proteins linked to phenotype changes were examined. Similar to the exogenous GD1b treatment, the cell growth of the GD1b synthase gene-transfectants was significantly suppressed compared with the vector-transfectant cell lines and transfection activated the apoptotic molecules such as processed forms of caspase-8, -7 and PARP, but not the levels of expression of Bax and Bcl-2. GD1b-induced apoptosis was blocked by caspase inhibitor, Z-VAD. Therefore, taken together, it was concluded that GD1b could play an important role in the regulation of breast cancer apoptosis.
Collapse
|
8
|
Abstract
Glycosphingolipids (GSLs) are a family of bioactive lipids that in addition to their role in the regulation of structural properties of membrane bilayers have emerged as crucial players in many biological processes and signal transduction pathways. Rather than being uniformly distributed within membrane bilayers, GSLs are localized in selective domains called lipid rafts where many signaling platforms operate. One of the most important functions of GSLs, particularly ceramide, is their ability to regulate cell death pathways and hence cell fate. This complex role is accomplished by the ability of GSLs to act in distinct subcellular strategic centers, such as mitochondria, endoplasmic reticulum (ER) or lysosomes to mediate apoptosis, ER stress, autophagy, lysosomal membrane permeabilization and necroptosis. Hence better understanding the role of GSLs in cell death may be of relevance for a number of pathological processes and diseases, including neurodegeneration, metabolic liver diseases and cancer.
Collapse
|
9
|
Miklavcic JJ, Hart TDL, Lees GM, Shoemaker GK, Schnabl KL, Larsen BMK, Bathe OF, Thomson ABR, Mazurak VC, Clandinin MT. Increased catabolism and decreased unsaturation of ganglioside in patients with inflammatory bowel disease. World J Gastroenterol 2015; 21:10080-10090. [PMID: 26401073 PMCID: PMC4572789 DOI: 10.3748/wjg.v21.i35.10080] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/01/2015] [Revised: 03/27/2015] [Accepted: 05/04/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether accelerated catabolism of ganglioside and decreased ganglioside content contribute to the etiology of pro-inflammatory intestinal disease.
METHODS: Intestinal mucosa from terminal ileum or colon was obtained from patients with ulcerative colitis or inflammatory Crohn’s disease (n = 11) undergoing bowel resection and compared to control samples of normal intestine from patients with benign colon polyps (n = 6) and colorectal cancer (n = 12) in this observational case-control study. Gangliosides and phospholipids of intestinal mucosa were characterized by class and ceramide or fatty acid composition using liquid chromatography triple-quad mass spectrometry. Content and composition of ganglioside classes GM1, GM3, GD3, GD1a, GT1 and GT3 were compared among subject groups. Content and composition of phospholipid classes phosphatidylcholine (PC) and phosphatidylethanolamine were compared among subject groups. Unsaturation index of individual ganglioside and phospholipid classes was computed and compared among subject groups. Ganglioside catabolism enzymes beta-hexosaminidase A (HEXA) and sialidase-3 (NEU3) were measured in intestinal mucosa using western blot and compared among subject groups.
RESULTS: Relative GM3 ganglioside content was 2-fold higher (P < 0.05) in intestine from patients with inflammatory bowel disease (IBD) compared to control intestine. The quantity of GM3 and ratio of GM3/GD3 was also higher in IBD intestine than control tissue (P < 0.05). Control intestine exhibited 3-fold higher (P < 0.01) relative GD1a ganglioside content than IBD intestine. GD3 and GD1a species of ganglioside containing three unsaturated bonds were present in control intestine, but were not detected in IBD intestine. The relative content of PC containing more than two unsaturated bonds was 30% lower in IBD intestine than control intestine (P < 0.05). The relative content of HEXA in IBD intestine was increased 1.7-fold (P < 0.05) and NEU3 was increased 8.3-fold (P < 0.01) compared to normal intestine. Intestinal mucosa in IBD is characterized by increased GM3 content, decreased GD1a, and a reduction in polyunsaturated fatty acid constituents in GD3, GD1a and PC.
CONCLUSION: This study suggests a new paradigm by proposing that IBD occurs as a consequence of increased metabolism of specific gangliosides.
Collapse
|
10
|
Metabolism, physiological role, and clinical implications of sphingolipids in gastrointestinal tract. BIOMED RESEARCH INTERNATIONAL 2013; 2013:908907. [PMID: 24083248 PMCID: PMC3780527 DOI: 10.1155/2013/908907] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 04/09/2013] [Revised: 06/30/2013] [Accepted: 08/02/2013] [Indexed: 01/21/2023]
Abstract
Sphingolipids in digestive system are responsible for numerous important physiological and pathological processes. In the membrane of gut epithelial cells, sphingolipids provide structural integrity, regulate absorption of some nutrients, and act as receptors for many microbial antigens and their toxins. Moreover, bioactive sphingolipids such as ceramide or sphingosine-1-phosphate regulate cellular growth, differentiation, and programmed cell death-apoptosis. Although it is well established that sphingolipids have clinical implications in gastrointestinal tumorigenesis or inflammation, further studies are needed to fully explore the role of sphingolipids in neoplastic and inflammatory diseases in gastrointestinal tract. Pharmacological agents which regulate metabolism of sphingolipids can be potentially used in the management of colorectal cancer or inflammatory bowel diseases. The aim of this work is to critically the review physiological and pathological roles of sphingolipids in the gastrointestinal tract.
Collapse
|
11
|
Basu S, Ma R, Moskal JR, Basu M, Banerjee S. Apoptosis of Breast Cancer Cells: Modulation of Genes for Glycoconjugate Biosynthesis and Targeted Drug Delivery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 749:233-55. [DOI: 10.1007/978-1-4614-3381-1_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2022]
|
12
|
Birks SM, Danquah JO, King L, Vlasak R, Gorecki DC, Pilkington GJ. Targeting the GD3 acetylation pathway selectively induces apoptosis in glioblastoma. Neuro Oncol 2011; 13:950-60. [PMID: 21807667 DOI: 10.1093/neuonc/nor108] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/12/2023] Open
Abstract
The expression of ganglioside GD3, which plays crucial roles in normal brain development, decreases in adults but is upregulated in neoplastic cells, where it regulates tumor invasion and survival. Normally a buildup of GD3 induces apoptosis, but this does not occur in gliomas due to formation of 9-O-acetyl GD3 by the addition of an acetyl group to the terminal sialic acid of GD3; this renders GD3 unable to induce apoptosis. Using human biopsy-derived glioblastoma cell cultures, we have carried out a series of molecular manipulations targeting GD3 acetylation pathways. Using immunocytochemistry, flow cytometry, western blotting, and transwell assays, we have shown the existence of a critical ratio between GD3 and 9-O-acetyl GD3, which promotes tumor survival. Thus, we have demonstrated for the first time in primary glioblastoma that cleaving the acetyl group restores GD3, resulting in a reduction in tumor cell viability while normal astrocytes remain unaffected. Additionally, we have shown that glioblastoma viability is reduced due to the induction of mitochondrially mediated apoptosis and that this occurs after mitochondrial membrane depolarization. Three methods of cleaving the acetyl group using hemagglutinin esterase were investigated, and we have shown that the baculovirus vector transduces glioma cells as well as normal astroctyes with a relatively high efficacy. A recombinant baculovirus containing hemagglutinin esterase could be developed for the clinic as an adjuvant therapy for glioma.
Collapse
Affiliation(s)
- Suzanne M Birks
- Cellular and Molecular Neuro-oncology Research Group, Institute Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK.
| | | | | | | | | | | |
Collapse
|
13
|
Abstract
Studies of sphingolipids have become one of the most rapidly advancing fields in the last two decades. These highly diverse lipids have been known to have multiple physiological functions and clinical implications in several diseases, including tumorigenesis, inflammation, atherosclerosis and neural degenerative diseases. Unlike other organs, sphingolipids in the intestinal tract are present not only as lipid constituents in the cells but also as dietary compositions for digestion in the lumen. The present review focuses on the presence of sphingolipids and their catalytic enzymes in the gut; the metabolism and the signaling effects of the metabolites and their impacts on barrier functions, cholesterol absorption, inflammatory diseases and tumor development in the gut.
Collapse
Affiliation(s)
- Rui-Dong Duan
- Gastroenterology and Nutrition Laboratory, Biomedical Center, B11, Institution of Clinical Sciences, University of Lund, Lund, Sweden.
| |
Collapse
|
14
|
Kausar T, Ahsan A, Hasan MR, Lin L, Beer DG, Ralhan R. Sperm protein 17 is a novel marker for predicting cisplatin response in esophageal squamous cancer cell lines. Int J Cancer 2010; 126:1494-503. [PMID: 19685492 DOI: 10.1002/ijc.24828] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/08/2022]
Abstract
Expression of sperm protein 17 (Sp17) mRNA has been reported in various malignancies. In an earlier study, we reported the upregulation of Sp17 transcripts in primary esophageal squamous cell carcinomas (ESCCs) using differential display and detected Sp17 transcripts in 86% of ESCCs by RT-PCR, whereas no transcripts were detected in the paired normal esophageal tissues. Herein we hypothesized that Sp17 might be used as a marker for detecting the response of anticancer therapies in ESCCs. Our results indicated that Sp17 protein levels in esophageal squamous cancer cell lines decreased in response to treatment with (i) the HSP90 activity inhibitor geldanamycin, (ii) the tyrosine kinase inhibitor erlotinib and (iii) cisplatin (chemotherapeutic agent commonly used in management of ESCC). In contrast, the Sp17 levels did not decrease in response to radiation therapy and treatment with the chemotherapeutic agent, gemcitabine. Further investigations showed that cisplatin induced decrease in Sp17 levels was due to transcriptional inhibition and cisplatin-resistant cell lines did not show this decrease in Sp17 levels in response to cisplatin treatment. In addition, we also carried our mass spectophotometric analysis to identify the binding partners of Sp17 to characterize its possible involvement in esophageal tumorigenesis and chemoresistance.
Collapse
Affiliation(s)
- Tasneem Kausar
- Department of Biochemistry, All India Institute of Medical Science, New Delhi, India
| | | | | | | | | | | |
Collapse
|
15
|
GD3 synthase overexpression sensitizes hepatocarcinoma cells to hypoxia and reduces tumor growth by suppressing the cSrc/NF-kappaB survival pathway. PLoS One 2009; 4:e8059. [PMID: 19956670 PMCID: PMC2777380 DOI: 10.1371/journal.pone.0008059] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/26/2009] [Accepted: 11/01/2009] [Indexed: 01/29/2023] Open
Abstract
Background Hypoxia-mediated HIF-1α stabilization and NF-κB activation play a key role in carcinogenesis by fostering cancer cell survival, angiogenesis and tumor invasion. Gangliosides are integral components of biological membranes with an increasingly recognized role as signaling intermediates. In particular, ganglioside GD3 has been characterized as a proapoptotic lipid effector by promoting cell death signaling and suppression of survival pathways. Thus, our aim was to analyze the role of GD3 in hypoxia susceptibility of hepatocarcinoma cells and in vivo tumor growth. Methodology/Principal Findings We generated and characterized a human hepatocarcinoma cell line stably expressing GD3 synthase (Hep3B-GD3), which catalyzes the synthesis of GD3 from GM3. Despite increased GD3 levels (2–3 fold), no significant changes in cell morphology or growth were observed in Hep3B-GD3 cells compared to wild type Hep3B cells under normoxia. However, exposure of Hep3B-GD3 cells to hypoxia (2% O2) enhanced reactive oxygen species (ROS) generation, resulting in decreased cell survival, with similar findings observed in Hep3B cells exposed to increasing doses of exogenous GD3. In addition, hypoxia-induced c-Src phosphorylation at tyrosine residues, NF-κB activation and subsequent expression of Mn-SOD were observed in Hep3B cells but not in Hep3B-GD3 cells. Moreover, MnTBAP, an antioxidant with predominant SOD mimetic activity, reduced ROS generation, protecting Hep3B-GD3 cells from hypoxia-induced death. Finally, lower tumor growth, higher cell death and reduced Mn-SOD expression were observed in Hep3B-GD3 compared to Hep3B tumor xenografts. Conclusion These findings underscore a role for GD3 in hypoxia susceptibility by disabling the c-Src/NF-κB survival pathway resulting in lower Mn-SOD expression, which may be of relevance in hepatocellular carcinoma therapy.
Collapse
|
16
|
Li M, Chen F, Liu CP, Li DM, Li X, Wang C, Li JC. Dexamethasone enhances trichosanthin-induced apoptosis in the HepG2 hepatoma cell line. Life Sci 2009; 86:10-6. [PMID: 19891978 DOI: 10.1016/j.lfs.2009.10.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/05/2009] [Revised: 10/11/2009] [Accepted: 10/16/2009] [Indexed: 11/18/2022]
Abstract
AIMS Trichosanthin (TCS) is a type I ribosome-inactivating protein (RIP) with antitumor activities for various cancers. In this paper, we aimed to investigate whether dexamethasone, an important synthetic member of the glucocorticoid steroids, in combination with TCS can be a potential therapy in treating hepatoma. MAIN METHODS Cell viability was investigated using MTT assay, and apoptosis was evaluated with Hoechst 33258 staining. Western blot analysis was used to examine the changes in the expression levels of IkappaB-alpha, NF-kappaB p65 subunit and Cox-2. Additionally, we took advantage of dominant-negative IkappaB (IkappaB-DM) over-expression and chemical inhibitor PDTC to inhibit NF-kappaB activation. KEY FINDINGS Our results demonstrated that dexamethasone could enhance TCS-induced apoptosis in the hepatoma cell line HepG2, decreasing IC50 values from in excess of 200microg/ml to 50microg/ml. In addition, our results demonstrated that TCS could induce rapid degradation of IkappaB-alpha, nuclear translocation of NF-kappaB and decrease of COX-2 expression in HepG2 cells. Inhibition of NF-kappaB by biological (IkappaB-DM) or chemical inhibitor (PDTC) increased HepG2 cells' sensitivity to TCS, resulting in cell viability rate decreasing and apoptotic rate increasing. Simultaneously, dexamethasone increased the level of IkappaB-alpha protein and effectively inhibited TCS-induced degradation of IkappaB-alpha. SIGNIFICANCE These results suggest that dexamethasone could enhance trichosanthin-induced apoptosis in the HepG2, at least in part, by inhibiting the NF-kappaB signaling pathway and thus strengthening the antitumor effects of TCS, which highlights the possibility of combined drug application of TCS and dexamethasone in the clinical treatment of hepatoma.
Collapse
Affiliation(s)
- Meng Li
- Institute of Cell Biology, Zhejiang University Medical School, Hangzhou 310058, China
| | | | | | | | | | | | | |
Collapse
|
17
|
GD3 nuclear localization after apoptosis induction in HUT-78 cells. Biochem Biophys Res Commun 2008; 368:495-500. [DOI: 10.1016/j.bbrc.2007.12.196] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/14/2007] [Accepted: 12/14/2007] [Indexed: 11/22/2022]
|
18
|
Novel sugar-cholestanols as anticancer agents against peritoneal dissemination of tumor cells. Glycoconj J 2008; 25:531-44. [PMID: 18327639 DOI: 10.1007/s10719-008-9108-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2007] [Revised: 12/08/2007] [Accepted: 01/09/2008] [Indexed: 12/21/2022]
Abstract
Chemically synthesized sugar-cholestanols with mono-, di-, and tri-saccharides attached to cholestanol showed strong inhibiting activity against the proliferation of colorectal and gastric cancer cells. In contrast, cholestanol without sugar moieties was totally ineffective. Furthermore, when cancer cells were exposed to GlcNAcRbetacholestanol (R=(-) or beta1-3Gal), the compound was rapidly taken up via the lipid rafts/microdomains on the cell surface. The uptake of sugar-cholestanol in mitochondria increased gradually and was followed by the release of cytochrome c from mitochondria and the activation of apoptotic signals through the mitochondrial pathway and the caspase cascade, leading to apoptotic cell death, characterized by DNA ladder formation and nuclear fragmentation. Additionally, the examination of GlcNAcRbetacholestanol in a mouse model of peritoneal dissemination showed a dramatic reduction of tumor growth (P < 0.003) and prolonged mouse survival time (P<0.0001). Based on these observations, we believe that the sugar-cholestanols described here have clinical potential as novel anticancer agents.
Collapse
|
19
|
Abstract
Sphingolipids (SLs) have been considered for many years as predominant building blocks of biological membranes with key structural functions and little relevance in cellular signaling. However, this view has changed dramatically in recent years with the recognition that certain SLs such as ceramide, sphingosine 1-phosphate and gangliosides, participate actively in signal transduction pathways, regulating many different cell functions such as proliferation, differentiation, adhesion and cell death. In particular, ceramide has attracted considerable attention in cell biology and biophysics due to its key role in the modulation of membrane physical properties, signaling and cell death regulation. This latter function is largely exerted by the ability of ceramide to activate the major pathways governing cell death such as the endoplasmic reticulum and mitochondria. Overall, the evidence so far indicates a key function of SLs in disease pathogenesis and hence their regulation may be of potential therapeutic relevance in different pathologies including liver diseases, neurodegeneration and cancer biology and therapy.
Collapse
Affiliation(s)
- Albert Morales
- Liver Unit and Centro de Investigaciones Biomédicas Esther Koplowitz, IMDiM, Hospital, Clínic i Provincial, Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | | | | | | | | |
Collapse
|
20
|
Faried A, Faried LS, Nakagawa T, Yamauchi T, Kitani M, Sasabe H, Nishimura T, Usman N, Kato H, Asao T, Kuwano H, Yazawa S. Chemically synthesized sugar-cholestanols possess a preferential anticancer activity involving promising therapeutic potential against human esophageal cancer. Cancer Sci 2007; 98:1358-67. [PMID: 17640296 PMCID: PMC11158219 DOI: 10.1111/j.1349-7006.2007.00563.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022] Open
Abstract
The understanding of the cell signaling pathways and the molecular events leading to cell death of cancer cells will provide in-depth perspective into the identification and development of potent anticancer agents. A balance between cell proliferation and cell death has been raised as a rational target for the management of malignant tumors. In the present study, the authors demonstrated that chemically synthesized sugar-cholestanols consisting of GlcNAcbeta-, Galbeta- and GlcNAcbeta1,3Galbeta-cholestanols exerted a strong inhibiting activity against cell proliferation of esophageal cancer cells, but cholestanol itself did not show such an activity against the same cancer cells at all. In addition to their predominant role as an antiproliferation agent, evidence based on the molecular analyses suggested that sugar-cholestanols played a regulatory role in multiple signal transduction pathways inducing apoptosis through both the death signal-extrinsic and the mitochondria-intrinsic pathways. Sugar-cholestanols seemed to be more susceptible to esophageal cancer cells than to non-cancerous esophageal cells at the very early event of their exposure and, further, to suppress specifically the expression of vascular endothelial growth factor. Taken together, these novel functions of sugar-cholestanols indicate that they could have promising therapeutic potential against human esophageal cancer.
Collapse
Affiliation(s)
- Ahmad Faried
- Department of General Surgical Science (Surgery I), Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Sphingolipids (SLs) comprise a class of lipids with important structural functions and increasing relevance in cellular signalling. In particular, ceramide has attracted considerable attention owing to its role as a second messenger modulating several cell functions such as proliferation, gene expression, differentiation, cell cycle arrest and cell death. Increasing evidence documents the role of SLs in stress and death ligand-induced hepatocellular death, which contributes to the progression of several liver diseases including steatohepatitis, ischaemia-reperfusion liver injury or hepatocarcinogenesis. Furthermore, recent data indicate that the accumulation of SLs in specific cell subcompartments, characteristic of many sphingolipidoses, contributes to the hepatic dysfunctions that accompany these inherited diseases. Hence, the regulation of the cell biology and metabolism of SLs may open up a novel therapeutic avenue in the treatment of liver diseases.
Collapse
Affiliation(s)
- Montserrat Marí
- Liver Unit and Centro de Investigaciones Biomédicas Esther Koplowitz, IMDiM, Hospital Clinic i Provincial, CIBER-HEPAD, Instituto Salud Carlos III, IDIBAPS, Barcelona, Spain
| | | |
Collapse
|
22
|
Boyle PJ, Ma R, Tuteja N, Banerjee S, Basu S. Apoptosis of human breast carcinoma cells in the presence of cis-platin and L-/D-PPMP: IV. Modulation of replication complexes and glycolipid: Glycosyltransferases. Glycoconj J 2007; 23:175-87. [PMID: 16691501 DOI: 10.1007/s10719-006-7923-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/24/2022]
Abstract
Apoptosis of human breast carcinoma cells (SKBR-3, MCF-7, and MDA-468) has been observed after treatment of these cells with anti-cancer drug cis-platin and glycosphingolipid biosynthesis inhibitor L- and D-PPMP, respectively. These drugs initiated apoptosis in a dose-dependent manner as measured by phenotypic morphological changes, by binding of a fluorescent phophatidyl serine-specific dye (PSS-380) onto the outer leaflet of the cell membranes, and by activation of caspases, -3, -8, and -9. It was observed that in two hours very little apoptotic process had started but predominant biochemical changes occurred after 6 h. DNA degradation started after 24 hours of drug treatment. However, very little is known about the stability of the ';Replication Complexes'' during the apoptotic process. DNA helicases are motor proteins that catalyze the melting of genomic DNA during its replication, repair, and recombination processes. Previously, DNA helicase-III was characterized as a component of the replication complexes isolated from embryonic chicken brains as well as breast and colon carcinoma cells. Helicase activities were measured by a novel method (ROME assay), and DNA polymerase-alpha activities were determined by regular chain extension of the nicked ACT-DNA, by determining values obtained from +/- aphidicolin-treated incubation mixtures. In all three breast carcinoma cell lines, a common trend was observed: a decrease of activities of DNA polymerase-alpha and Helicase III. A sharp decrease of activities of the glycolipid sialyltransferases: SAT-2 (CMP-NeuAc; GD3 alpha2-8 sialyltransferase) and SAT-4 (CMP-NeuAc: GM1a alpha2-3 sialyltransferase) was observed in the apoptotic carcinoma cells treated with L-PPMP compared with cis-platin.
Collapse
Affiliation(s)
- Patrick J Boyle
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | | | |
Collapse
|
23
|
Omran OM, Saqr HE, Yates AJ. Molecular Mechanisms of GD3-Induced Apoptosis in U-1242 MG Glioma Cells. Neurochem Res 2006; 31:1171-80. [PMID: 17043769 DOI: 10.1007/s11064-006-9147-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/01/2006] [Accepted: 08/24/2006] [Indexed: 11/28/2022]
Abstract
An increasing amount of evidence indicates that the disialoganglioside GD3 is involved in apoptosis in many cell lines. Our previous studies demonstrated that endogenous GD3 expression induced apoptosis in U-1242 MG glioma cells transfected with the GD3 synthase gene (U1242MG-GD3 cells). In this paper, we present further investigations on the molecular mechanisms of GD3-induced apoptosis in this cell line. We found that endogenously synthesized GD3 localizes to the caveolae of this cell line, where it promotes the localization of death receptor 5 (DR5), tumor necrosis factor receptor-1 (TNF-R1), and Fas (Apo-1) to the caveolae. In addition, caspase-8 was translocated to the caveolar fraction and cleaved; the cleaved proteins were then re-located into the high density fractions. However, GD3 had no effect on the distribution of the adapter protein Fas-associated death domain (FADD). We conclude that GD3 functions as a regulatory molecule early in the extrinsic apoptosis pathway.
Collapse
Affiliation(s)
- O M Omran
- Department of Pathology, The Ohio State University, 4170 Graves Hall, 333 W 10Th Ave, Columbus, OH 43210, USA
| | | | | |
Collapse
|
24
|
Llacuna L, Marí M, Garcia-Ruiz C, Fernandez-Checa JC, Morales A. Critical role of acidic sphingomyelinase in murine hepatic ischemia-reperfusion injury. Hepatology 2006; 44:561-72. [PMID: 16941686 DOI: 10.1002/hep.21285] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022]
Abstract
The molecular mechanisms of hepatic ischemia/reperfusion (I/R) damage are incompletely understood. We investigated the role of ceramide in a murine model of warm hepatic I/R injury. This sphingolipid induces cell death and participates in tumor necrosis factor (TNF) signaling. Hepatic ceramide levels transiently increased after the reperfusion phase of the ischemic liver in mice, because of an early activation of acidic sphingomyelinase (ASMase) followed by acid ceramidase stimulation. In vivo administration of an ASMase inhibitor, imipramine, or ASMase knockdown by siRNA decreased ceramide generation during I/R, and attenuated serum ALT levels, hepatocellular necrosis, cytochrome c release, and caspase-3 activation. ASMase-induced ceramide generation activated JNK resulting in BimL phosphorylation and translocation to mitochondria, as the inhibition of ASMase by imipramine prevented these events. In contrast, blockade of ceramide catabolism by N-oleyolethanolamine (NOE), a ceramidase inhibitor, enhanced ceramide levels and potentiated I/R injury compared with vehicle-treated mice. Pentoxifylline treatment prevented TNF upregulation and ASMase activation. Furthermore, 9 of 11 mice treated with imipramine survived 7 days after total liver ischemia, compared with 4 of 12 vehicle-treated mice, whereas 8 of 8 NOE-treated mice died within 2 days of total liver ischemia. In conclusion, ceramide generated from ASMase plays a key role in I/R-induced liver damage, and its modulation may be of therapeutic relevance.
Collapse
Affiliation(s)
- Laura Llacuna
- Liver Unit, Hospital Clínic, Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), and the Department of Experimental Pathology, Instituto Investigaciones Biomédicas Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain
| | | | | | | | | |
Collapse
|
25
|
Morales A, París R, Villanueva A, Llacuna L, García-Ruiz C, Fernández-Checa JC. Pharmacological inhibition or small interfering RNA targeting acid ceramidase sensitizes hepatoma cells to chemotherapy and reduces tumor growth in vivo. Oncogene 2006; 26:905-16. [PMID: 16862171 DOI: 10.1038/sj.onc.1209834] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/14/2023]
Abstract
Ceramidases (CDases) play a key role in cancer therapy through enhanced conversion of ceramide into sphingosine 1-phosphate (S1P), but their involvement in hepatocarcinogenesis is unknown. Here, we report that daunorubicin (DNR) activated acid CDase post-transcriptionally in established human (HepG2 cells) or mouse (Hepa1c1c7) hepatoma cell lines as well as in primary cells from murine liver tumors, but not in cultured mouse hepatocytes. Acid CDase silencing by small interfering RNA (siRNA) or pharmacological inhibition with N-oleoylethanolamine (NOE) enhanced the ceramide to S1P balance compared to DNR alone, sensitizing hepatoma cells (HepG2, Hep-3B, SK-Hep and Hepa1c1c7) to DNR-induced cell death. DNR plus NOE or acid CDase siRNA-induced cell death was preceded by ultrastructural changes in mitochondria, stimulation of reactive oxygen species generation, release of Smac/DIABLO and cytochrome c and caspase-3 activation. In addition, in vivo siRNA treatment targeting acid CDase reduced tumor growth in liver tumor xenografts of HepG2 cells and enhanced DNR therapy. Thus, acid CDase promotes hepatocarcinogenesis and its antagonism may be a promising strategy in the treatment of liver cancer.
Collapse
Affiliation(s)
- A Morales
- Liver Unit, Institut de Malalties Digestives, Hospital Clínic i Provincial, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
26
|
Yang J, Amiri KI, Burke JR, Schmid JA, Richmond A. BMS-345541 targets inhibitor of kappaB kinase and induces apoptosis in melanoma: involvement of nuclear factor kappaB and mitochondria pathways. Clin Cancer Res 2006; 12:950-60. [PMID: 16467110 PMCID: PMC2668250 DOI: 10.1158/1078-0432.ccr-05-1220] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Constitutive activation of inhibitor of kappaB kinase (IKK) confers melanoma resistance to apoptosis and chemotherapy. Whether IKK is able to serve as a therapeutic target in melanoma is unknown. We explored the possibility of exploiting IKK as a therapeutic target in melanoma by using BMS-345541, a novel compound with a highly selective IKKbeta inhibitory activity, to trigger melanoma cell apoptosis. EXPERIMENTAL DESIGN Three human melanoma cell lines (SK-MEL-5, Hs 294T, and A375), all of which have high constitutive IKK activities, served as in vitro and in vivo melanoma models for treatment with BMS-345541. Two known antitumor drugs (temozolomide and bortezomib) were used as parallel controls for evaluation of the therapeutic efficiency and toxicity of BMS-345541. The effects of BMS-345541 on nuclear factor kappaB (NF-kappaB) signaling and on the apoptosis machinery were investigated. RESULTS Inhibition of constitutive IKK activity by BMS-345541 resulted in the reduction of NF-kappaB activity, CXCL1 chemokine secretion by cultured melanoma cells and melanoma cell survival in vitro and in vivo. The effect of BMS-345541 on tumor cell growth was through mitochondria-mediated apoptosis, based on the release of apoptosis-inducing factor, dissipation of mitochondrial membrane potential, and reduced ratio of B cell lymphoma gene-2 (Bcl-2)/Bcl-associated X protein (Bax) in mitochondria. The BMS-345541 execution of apoptosis was apoptosis-inducing factor-dependent, but largely caspase-independent. CONCLUSION BMS-345541 down-regulation of IKK activity results in mitochondria-mediated apoptosis of tumor cells because the programmed cell death machinery in melanoma cells is highly regulated by NF-kappaB signaling. Therefore, IKK may serve as a potential target for melanoma therapy.
Collapse
Affiliation(s)
- Jinming Yang
- Veterans Affairs Medical Center and Department of Cancer Biology, Vanderbilt University School of Medicine
| | - Katayoun I. Amiri
- Veterans Affairs Medical Center and Department of Cancer Biology, Vanderbilt University School of Medicine
- Meharry Medical College, Nashville, Tennessee
| | - James R. Burke
- Bristol-Myers Squibb Pharmaceutical Research Institute, Princeton, New Jersey
| | - Johannes A. Schmid
- Department of Vascular Biology and Thrombosis Research, University of Vienna Medical School, Vienna, Austria
| | - Ann Richmond
- Veterans Affairs Medical Center and Department of Cancer Biology, Vanderbilt University School of Medicine
| |
Collapse
|
27
|
Dyatlovitskaya EV, Kandyba AG. Role of biologically active sphingolipids in tumor growth. BIOCHEMISTRY (MOSCOW) 2006; 71:10-7. [PMID: 16457613 DOI: 10.1134/s0006297906010020] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/23/2022]
Abstract
This review highlights the literature on the effects of biologically active sphingolipids (sphingosine, ceramide, sphingomyelin, glucosylceramide, gangliosides GM1, GM2, GM3, GD3, etc.) on proliferation, apoptosis, metastases, and invasiveness of tumor cells and the putative role of sphingolipids in chemotherapy of malignant tumors.
Collapse
Affiliation(s)
- E V Dyatlovitskaya
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | | |
Collapse
|
28
|
Basu S, Ma R, Mikulla B, Bradley M, Moulton C, Basu M, Banerjee S, Inokuchi JI. Apoptosis of human carcinoma cells in the presence of inhibitors of glycosphingolipid biosynthesis: I. Treatment of Colo-205 and SKBR3 cells with isomers of PDMP and PPMP. Glycoconj J 2005; 20:157-68. [PMID: 15090729 DOI: 10.1023/b:glyc.0000024254.64450.8b] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/04/2023]
Abstract
Apoptosis, or programmed cell death, plays an important role in many physiological and diseased conditions. Induction of apoptosis in cancer cells by anti-cancer drugs and biosynthetic inhibitors of cells surface glycolipids in the human colon carcinoma cells (Colo-205) are of interest in recent years. In our present studies, we have employed different stereoisomers of PPMP and PDMP (inhibit GlcT-glycosyltransferase (GlcT-GLT)) to initiate apoptosis in Colo-205 cells grown in culture in the presence of (3)H-TdR and (3)H/or (14)C-L-Serine. Our analysis showed that the above reagents (between 1 to 20 microM) initiated apoptosis with induction of Caspase-3 activities and phenotypic morphological changes in a dose-dependent manner. We have observed an increase of radioactive ceramide formation in the presence of a low concentration (1-4 microM) of these reagents in these cell lines. However, high concentrations (4-20 microM) inhibited incorporation of radioactive serine in the higher glycolipids. Colo-205 cells were treated with L-threo-PPMP (0-20 microM) and activities of different GSL: GLTs were estimated in total Golgi-pellets. The cells contained high activity of GalT-4 (UDP-Gal: LcOse3Cer beta 1-4galactosyltransferase), whereas negligible activity of GalT-3 (UDP-Gal: GM2 beta 1-3galactosyltransferase) or GM2-synthase activity of the ganglioside pathway was detected. Previously, GLTs involved in the biosynthetic pathway of SA-Le(x) formation had been detected in these colon carcinoma (or Colo-205) cells (Basu M et al. Glycobiology 1, 527-35 (1991)). However, during progression of apoptosis in Colo-205 cells with increasing concentrations of L-PPMP, the GalT-4 activity was decreased significantly. These changes in the specific activity of GalT-4 in the total Golgi-membranes could be the resultant of decreased gene expression of the enzyme.
Collapse
Affiliation(s)
- Subhash Basu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Liu CY, Liao HF, Shih SC, Lin SC, Chang WH, Chu CH, Wang TE, Chen YJ. Colchicine sensitizes human hepatocellular carcinoma cells to damages caused by radiation. World J Gastroenterol 2005; 11:4237-40. [PMID: 16015697 PMCID: PMC4615450 DOI: 10.3748/wjg.v11.i27.4237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023] Open
Abstract
AIM: We studied the effect of colchicine combined with radiation on the survival of human hepatocellular carcinoma (HCC) HA22T/VGH cells.
METHODS: Twenty-four hours after treatment with 0-8 ng/mL colchicine, HA22T/VGH cells were irradiated at various doses (0, 1, 2, 4, and 8 Gy). Colony assay was performed to assess the surviving cell fraction. Survival curves were fitted by using a linear-quadratic model to estimate the sensitizer enhancement ratio (SER). Flow cytometry was used for cell cycle analysis.
RESULTS: Colchicine at lower concentrations (1 and 2 ng/mL) had obvious synergy with radiation to inhibit HCC cell growth, whereas higher concentrations (4 and 8 ng/mL) had only additive effect to radiation. Pretreatment with 1 and 2 ng/mL colchicine for 24-h enhanced cell killing by radiation with SERs of 1.21 and 1.53, respectively. G2/M arrest was only observed with higher colchicine doses (8 and 16 ng/mL) after 24-h treatment; this effect was neither seen with lower doses (1, 2, and 4 ng/mL) nor with any dose after only 1 h of treatment.
CONCLUSION: Our results suggest that colchicine has potential as an adjunct to radiotherapy for HCC treatment. Lower doses of colchicine possess radiosensitizing effects via some mechanism other than G2/M arrest. Further study is necessary to elucidate the mechanism.
Collapse
Affiliation(s)
- Chia-Yuan Liu
- Division of Gastroenterology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104, Taiwan, China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Malisan F, Testi R. The Ganglioside GD3 as the Greek Goddess Hecate: Several Faces Turned Towards as Many Directions. IUBMB Life 2005; 57:477-82. [PMID: 16081368 DOI: 10.1080/15216540500167179] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/25/2022]
Abstract
The disialoganglioside GD3 can mediate biological functions as diverse as proliferation, differentiation, and apoptosis. Since intracellular level of GD3 is crucial for the cell, understanding the mechanisms by which GD3 metabolism is tightly regulated seems of particular importance. GD3 can be enlisted among the most potent natural inducers of mitochondrial damage and apoptosis. However, some cell types resist GD3-mediated mitochondrial damage through complex mechanisms which are beginning to be unveiled.
Collapse
Affiliation(s)
- Florence Malisan
- Laboratory of Immunology and Signal Transduction, Department of Experimental Medecine and Biochemical sciences, University "Tor Vergata", Rome, Italy.
| | | |
Collapse
|
31
|
Brønnum H, Seested T, Hellgren LI, Brix S, Frøkiaer H. Milk-Derived GM3 and GD3 Differentially Inhibit Dendritic Cell Maturation and Effector Functionalities. Scand J Immunol 2005; 61:551-7. [PMID: 15963050 DOI: 10.1111/j.1365-3083.2005.01566.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
Gangliosides are complex glycosphingolipids, which exert immune-modulating effects on various cell types. Ganglioside GD(3) and GM(3) are the predominant gangliosides of human breast milk but during the early phase of lactation, the content of GD(3) decreases while GM(3) increases. The biological value of gangliosides in breast milk has yet to be elucidated but when milk is ingested, dietary gangliosides might conceptually affect immune cells, such as dendritic cells (DCs). In this study, we address the in vitro effect of GD(3) and GM(3) on DC effector functionalities. Treatment of bone marrow-derived DCs with GD(3) before lipopolysaccharide-induced maturation decreased the production of interleukin-6 (IL-6), IL-10, IL-12 and tumor necrosis factor-alpha as well as reduced the alloreactivity in mixed leucocyte reaction (MLR). In contrast, only IL-10 and IL-12 productions were significantly inhibited by GM(3,) and the potency of DCs to activate CD4(+) cells in MLR was unaffected by GM(3). However, both gangliosides suppressed expression of CD40, CD80, CD86 and major histocompatibility complex class II on DCs. Because GD(3) overall inhibits DC functionalities more than GM(3), the immune modulating effect of the ganglioside fraction of breast milk might be more prominent in the commencement of lactation during which the milk contains the most GD(3).
Collapse
Affiliation(s)
- H Brønnum
- Biochemistry and Nutrition Group, Centre for Advanced Food Studies and Biocentrum-DTU, Technical University of Denmark, Denmark
| | | | | | | | | |
Collapse
|
32
|
Morales A, Colell A, Mari M, Garcia-Ruiz C, Fernandez-Checa JC. Glycosphingolipids and mitochondria: role in apoptosis and disease. Glycoconj J 2005; 20:579-88. [PMID: 15454696 DOI: 10.1023/b:glyc.0000043294.62504.2c] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/12/2023]
Abstract
Glycosphingolipids (GSLs) comprise a class of lipids with important structural and signaling functions. Synthesized from ceramide in the Golgi, they are subsequently distributed to different compartments, most predominantly in the plasma membrane where they integrate signaling platforms. A recently characterized trafficking of ganglioside GD3 (GD3), a GSLs with two sialic-acid residues, to mitochondria has revealed a novel function of this lipid as a death effector. In addition to the interaction of GD3 with mitochondria recruiting these organelles to apoptotic pathways, GD3 disables survival paths dependent on NF-kappaB, thus favoring the balance towards cell death. The present review gathers the evidence documenting this emerging function of GSLs in cell death and their involvement in pathological states.
Collapse
Affiliation(s)
- Albert Morales
- Liver Unit, Instituto de Malalties Digestives, Hospital Clinic i Provincial, Instituto Investigaciones Biomédicas August Pi i Sunyer, Barcelona 08036, Spain
| | | | | | | | | |
Collapse
|
33
|
Basu S, Ma R, Boyle PJ, Mikulla B, Bradley M, Smith B, Basu M, Banerjee S. Apoptosis of human carcinoma cells in the presence of potential anti-cancer drugs: III. Treatment of Colo-205 and SKBR3 cells with: cis -platin, Tamoxifen, Melphalan, Betulinic acid, L-PDMP, L-PPMP, and GD3 ganglioside. Glycoconj J 2005; 20:563-77. [PMID: 15454695 DOI: 10.1023/b:glyc.0000043293.46845.07] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022]
Abstract
Breast cancer is the most common type of cancer, predominantly among women over 20, whereas colo-rectal cancer occurs in both men and women over the age of 50. Chemotherapy of both cancers affect rapidly growing normal as well as cancer cells. Cancer cells are non-apoptotic. Seven anti-cancer agents (cis -platin, Tamoxifen, Melphalan, Betulinic acid, D-PDMP, L-PPMP, and GD3) have been tested with human breast (SKBR3) and colon (Colo-205) carcinoma cells for their apoptotic effect and found to be positive by several assay systems. Colo-205 cells were obtained from ATCC, and the SKBR3 cells were a gift from the Cleveland Clinic. All of these six agents killed those two cell lines in a dose-dependent manner. In the early apoptotic stage (6 h), these cells showed only a flopping of phosphatidylserine on the outer lamella of the plasma membranes as evidenced by the binding of a novel fluorescent dye PSS-380. After 24 h of the treatment, those apoptotic cells showed damage of the plasma as well as the nuclear membrane as evidenced by binding of propidium iodide to the nuclear DNA. DNA laddering assay viewed further breakdown of DNA by 1% agarose gel electrophoresis analysis. It is concluded that during apoptosis the signaling by Mitochondrial Signaling Pathway (MSP) is stimulated by some of these agents. Caspase 3 was activated with the concomitant appearance of its p17 polypeptide as viewed by Westernblot analyses. Incorporation of radioactivity from [U-(14)C]-L-serine in total sphingolipid mixture was observed between 2 and 4 micromolar concentrations of most of the agents except ci s-platin. However, apoptosis in carcinoma cells in the presence of cis -platin is induced by a caspase 3 activation pathway without any increase in synthesis of ceramide.
Collapse
Affiliation(s)
- Subhash Basu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Fantappiè O, Lodovici M, Fabrizio P, Marchettia S, Fabbroni V, Solazzo M, Lasagna N, Pantaleo P, Mazzanti R. Vitamin E protects DNA from oxidative damage in human hepatocellular carcinoma cell lines. Free Radic Res 2005; 38:751-9. [PMID: 15453640 DOI: 10.1080/10715760410001705140] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/26/2022]
Abstract
Expression of multiple drug resistant (MDR) phenotype and over-expression of P-glycoprotein (P-gp) in the human hepatocellular carcinoma (HCC) cell clone P1(0.5), derived from the PLC/PRF/5 cell line (P5), are associated with strong resistance to oxidative stress and a significant (p < 0.01) increase in intracellular vitamin E content as compared with the parental cell line. This study evaluates the role of vitamin E in conferring resistance to drugs and oxidative stress in P1(0.5) cells. Parental drug-sensitive cells, P5, were incubated in alpha-tocopherol succinate (alpha-TS, 5 microM for 24 h) enriched medium to increase intracellular vitamin E content to levels comparable to those observed in P1(0.5) cells at basal conditions. Susceptibility to lipid peroxidation and oxidative DNA damage were assessed by measuring the concentration of thiobarbituric-reactive substances (TBARS) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) at basal and after experimental conditions. Cell capacity to form colonies and resistance to doxorubicin were also studied. P5 cells, treated with alpha-TS, became resistant to ADP-Fe3+ and to ionizing radiation-induced lipid peroxidation as P1(0.5) cells. Exposure to ADP-Fe3+ or ionizing radiation increased TBARS and the 8-OHdG content in the P5 cells, while vitamin E enrichment abolished these effects. Irradiation doses at 5 cGy increased TBARS and 8-OHdG. They also inhibited cell capacity to form colonies in the untreated P5 cells. Incubation with alpha-TS fully reverted this effect and significantly (p < 0.01) reduced the inhibitory effect of cell proliferation induced by irradiation doses at >500 cGy. Resistance to doxorubicin was not affected by alpha-TS. These observations demonstrate the role of vitamin E in conferring protection from lipid peroxidation, ionizing radiation and oxidative DNA damage on the human HCC cell line. They also rule out any role of P-gp over-expression as being responsible for these observations in cells with MDR phenotype expression.
Collapse
Affiliation(s)
- Ornella Fantappiè
- Department of Internal Medicine, U.A. Oncologia, Azienda Ospedaliero-Universitaria Careggi, Viale G.B. Morgagni 85, I-50134 Florence, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Lluis JM, Morales A, Blasco C, Colell A, Mari M, Garcia-Ruiz C, Fernandez-Checa JC. Critical role of mitochondrial glutathione in the survival of hepatocytes during hypoxia. J Biol Chem 2004; 280:3224-32. [PMID: 15548523 DOI: 10.1074/jbc.m408244200] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
Hypoxia is known to stimulate reactive oxygen species (ROS) generation. Because reduced glutathione (GSH) is compartmentalized in cytosol and mitochondria, we examined the specific role of mitochondrial GSH (mGSH) in the survival of hepatocytes during hypoxia (5% O2). 5% O2 stimulated ROS in HepG2 cells and cultured rat hepatocytes. Mitochondrial complex I and II inhibitors prevented this effect, whereas inhibition of nitric oxide synthesis with Nomega-nitro-L-arginine methyl ester hydrochloride or the peroxynitrite scavenger uric acid did not. Depletion of GSH stores in both cytosol and mitochondria enhanced the susceptibility of HepG2 cells or primary rat hepatocytes to 5% O2 exposure. However, this sensitization was abrogated by preventing mitochondrial ROS generation by complex I and II inhibition. Moreover, selective mGSH depletion by (R,S)-3-hydroxy-4-pentenoate that spared cytosol GSH levels sensitized rat hepatocytes to hypoxia because of enhanced ROS generation. GSH restoration by GSH ethyl ester or by blocking mitochondrial electron flow at complex I and II rescued (R,S)-3-hydroxy-4-pentenoate-treated hepatocytes to hypoxia-induced cell death. Thus, mGSH controls the survival of hepatocytes during hypoxia through the regulation of mitochondrial generation of oxidative stress.
Collapse
Affiliation(s)
- Josep M Lluis
- Liver Unit, Instituto de Malalties Digestives, Hospital Clinic i Provincial, Instituto Investigaciones Biomédicas August Pi i Sunyer
| | | | | | | | | | | | | |
Collapse
|
36
|
Ardite E, Barbera JA, Roca J, Fernández-Checa JC. Glutathione depletion impairs myogenic differentiation of murine skeletal muscle C2C12 cells through sustained NF-kappaB activation. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:719-28. [PMID: 15331397 PMCID: PMC1618592 DOI: 10.1016/s0002-9440(10)63335-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/23/2022]
Abstract
Skeletal muscle differentation is a complex process regulated at multiple levels. This study addressed the effect of glutathione (GSH) depletion on the transition of murine skeletal muscle C2C12 myoblasts into myocytes induced by growth factor inactivation. Cellular GSH levels increased within 24 hours on myogenic stimulation of myoblasts due to enhanced GSH synthetic rate accounted for by stimulated glutamate-L-cysteine ligase (also known as gamma-glutamylcysteine synthetase) activity. In contrast, the synthesis rate of GSH using gamma-glutamylcysteine and glutamate as precursors, which reflects the activity of the GSH synthetase, did not change during differentiation. The stimulation of GSH stores preceded the myogenic differentiation of C2C12 myoblasts monitored by expression of muscle-specific genes, creatine kinase (CK), myosin heavy chain (MyHC), and MyoD. The pattern of DNA binding activity of NF-kappaB and AP-1 in differentiating cells was similar both displaying an activation peak at 24 hours after myogenic stimulation. Depletion of cellular GSH levels 24 hours after stimulation of differentiation abrogated myogenesis as reflected by lower CK activity, MyHC levels, MyoD expression, and myotubes formation, effects that were reversible on GSH replenishment by GSH ethyl ester (GHSEE). Moreover, GSH depletion led to sustained activation of NF-kappaB, while GSHEE prevented it. Furthermore, inhibition of NF-kappaB activation restored myogenesis despite GSH depletion. Thus, GSH contributes to the formation of myotubes from satellite myoblasts by ensuring inactivation of NF-kappaB, and hence maintaining optimal GSH levels may be beneficial in restoring muscle mass in chronic inflammatory disorders.
Collapse
Affiliation(s)
- Esther Ardite
- Servei de Pneumologia, Instituto Clinic de Pneumologia y Cirugía Torácica, Barcelona, Spain
| | | | | | | |
Collapse
|
37
|
Abstract
Gangliosides, sialic acid-containing glycosphingolipids, have engendered great interest for more than 20 years in the search for target molecules of relevance for tumour growth and formation of metastases and as potential targets for immunotherapy. These molecules show large quantitative and structural variability, which is related to cell type and developmental stage. Their potential role in the formation of tumour metastases was suggested from data supporting that they are involved in cell growth regulation and in cell-cell and cell-matrix adhesion. Moreover, gangliosides are expressed on the cell surface and thereby are accessible for antibodies or other ganglioside-binding molecules to induce cell death, inhibit cell growth and/or inhibit formation of tumour metastasis. All tumours exhibit aberrant ganglioside expression. This includes overexpression of normal ganglioside constituents, which appears to be common among various tumours, and expression of gangliosides not found in normal adult tissue but often found during fetal development. The ganglioside composition of melanoma cells has been found to correlate with their metastatic potential and also to be selectively expressed in cells of a tumour mass and invading tumour cells. Passive immunotherapy using murine or murine/human chimeric monoclonal antiganglioside antibodies in their native form or combined with various effector molecules has been investigated. However, the vaccination strategy using native or structurally modified tumour-associated gangliosides in combination with adjuvants is currently the dominant method in clinical trials. The outcomes reported so far vary between type of tumour and treatment strategies. However, we believe that targeting gangliosides is as promising as any other immune therapeutic strategy, and basic research as well as clinical trials utilising new aspects is encouraged.
Collapse
Affiliation(s)
- Pam Fredman
- Experimental Neuroscience Section, Institute of Clinical Neuroscience, The Sahlgrenska Institute at Göteborg University, Sahlgrenska University Hospital, SE 43180 Molndal, Sweden.
| | | | | |
Collapse
|
38
|
Abstract
Apoptosis can be regulated at multiple levels. A number of proteins with regulatory function in cell death are sensitive to cellular redox environment. The antioxidant glutathione (GSH) and redox-sensitive proteins, thioredoxin and glutathione S-transferase, thus regulate cell death pathways by modulating the redox state of specific thiol residues of target proteins including stress kinases, transcription factors, and caspases. GSH in mitochondria plays an important role in the integrity of mitochondrial proteins and lipids known to play a vital role in the permeabilization of mitochondrial membranes and release of proapoptotic factors. The regulation of mitochondrial GSH (mGSH) is determined by its uptake from the cytosol which is dependent on appropriate membrane dynamics. The deposition of cholesterol in mitochondria induced by alcohol intake impairs this translocation, resulting in severe depletion of mGSH and in sensitization to apoptosis stimuli. Although the interaction of proapoptotic proteins with mitochondria initiates apoptotic pathways, recent data indicate that the mitochondrial trafficking of glycosphingolipids, e.g., ganglioside GD3, induced by apoptotic stimuli is a key event that sets off mitochondrial-dependent apoptotic cascades.
Collapse
Affiliation(s)
- José C Fernández-Checa
- Liver Unit, Instituto Malalties Digestives, Hospital Clinic i Provincial, Instituto Investigaciones Biomedicas August Pi I Sunyer, Consejo Superior Investigaciones Científicas, 08036 Barcelona, Spain.
| |
Collapse
|