1
|
Bordanaba-Florit G, Royo F, Albóniga OE, Clayton A, Falcón-Pérez JM, Webber J. Integration of proteomic and metabolomic analysis reveal distinct metabolic alterations of prostate cancer-associated fibroblasts compared to normal fibroblasts from patient's stroma samples. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167229. [PMID: 38734319 DOI: 10.1016/j.bbadis.2024.167229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
The prostate gland is a complex and heterogeneous organ composed of epithelium and stroma. Whilst many studies into prostate cancer focus on epithelium, the stroma is known to play a key role in disease with the emergence of a cancer-associated fibroblasts (CAF) phenotype associated upon disease progression. In this work, we studied the metabolic rewiring of stromal fibroblasts following differentiation to a cancer-associated, myofibroblast-like, phenotype. We determined that CAFs were metabolically more active compared to normal fibroblasts. This corresponded with a heightened lipogenic metabolism, as both reservoir species and building block compounds. Interestingly, lipid metabolism affects mitochondria functioning yet the mechanisms of lipid-mediated functions are unclear. Data showing oxidised fatty acids and glutathione system are elevated in CAFs, compared to normal fibroblasts, strengthens the hypothesis that increased metabolic activity is related to mitochondrial activity. This manuscript describes mechanisms responsible for the altered metabolic flux and shows that prostate cancer-derived extracellular vesicles can increase basal respiration in normal fibroblasts, mirroring that of the disease-like phenotype. This indicates that extracellular vesicles derived from prostate cancer cells may drive an altered oxygen-dependent metabolism associated to mitochondria in CAFs.
Collapse
Affiliation(s)
| | - Félix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), 28029 Madrid, Spain
| | - Oihane E Albóniga
- Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio, Spain
| | - Aled Clayton
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Juan Manuel Falcón-Pérez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), 28029 Madrid, Spain; Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio, Spain
| | - Jason Webber
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea, UK.
| |
Collapse
|
2
|
Mebarek S, Skafi N, Brizuela L. Targeting Sphingosine 1-Phosphate Metabolism as a Therapeutic Avenue for Prostate Cancer. Cancers (Basel) 2023; 15:2732. [PMID: 37345069 DOI: 10.3390/cancers15102732] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Prostate cancer (PC) is the second most common cancer in men worldwide. More than 65% of men diagnosed with PC are above 65. Patients with localized PC show high long-term survival, however with the disease progression into a metastatic form, it becomes incurable, even after strong radio- and/or chemotherapy. Sphingosine 1-phosphate (S1P) is a bioactive lipid that participates in all the steps of oncogenesis including tumor cell proliferation, survival, migration, invasion, and metastatic spread. The S1P-producing enzymes sphingosine kinases 1 and 2 (SK1 and SK2), and the S1P degrading enzyme S1P lyase (SPL), have been shown to be highly implicated in the onset, development, and therapy resistance of PC during the last 20 years. In this review, the most important studies demonstrating the role of S1P and S1P metabolic partners in PC are discussed. The different in vitro, ex vivo, and in vivo models of PC that were used to demonstrate the implication of S1P metabolism are especially highlighted. Furthermore, the most efficient molecules targeting S1P metabolism that are under preclinical and clinical development for curing PC are summarized. Finally, the possibility of targeting S1P metabolism alone or combined with other therapies in the foreseeable future as an alternative option for PC patients is discussed. Research Strategy: PubMed from INSB was used for article research. First, key words "prostate & sphingosine" were used and 144 articles were found. We also realized other combinations of key words as "prostate cancer bone metastasis" and "prostate cancer treatment". We used the most recent reviews to illustrate prostate cancer topic and sphingolipid metabolism overview topic.
Collapse
Affiliation(s)
- Saida Mebarek
- CNRS UMR 5246, INSA Lyon, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), 69622 Lyon, France
| | - Najwa Skafi
- CNRS, LAGEPP UMR 5007, University of Lyon, Université Claude Bernard Lyon 1, 43 Bd 11 Novembre 1918, 69622 Villeurbanne, France
| | - Leyre Brizuela
- CNRS UMR 5246, INSA Lyon, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), 69622 Lyon, France
| |
Collapse
|
3
|
Pal P, Atilla-Gokcumen GE, Frasor J. Emerging Roles of Ceramides in Breast Cancer Biology and Therapy. Int J Mol Sci 2022; 23:ijms231911178. [PMID: 36232480 PMCID: PMC9569866 DOI: 10.3390/ijms231911178] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
One of the classic hallmarks of cancer is the imbalance between elevated cell proliferation and reduced cell death. Ceramide, a bioactive sphingolipid that can regulate this balance, has long been implicated in cancer. While the effects of ceramide on cell death and therapeutic efficacy are well established, emerging evidence indicates that ceramide turnover to downstream sphingolipids, such as sphingomyelin, hexosylceramides, sphingosine-1-phosphate, and ceramide-1-phosphate, is equally important in driving pro-tumorigenic phenotypes, such as proliferation, survival, migration, stemness, and therapy resistance. The complex and dynamic sphingolipid network has been extensively studied in several cancers, including breast cancer, to find key sphingolipidomic alterations that can be exploited to develop new therapeutic strategies to improve patient outcomes. Here, we review how the current literature shapes our understanding of how ceramide synthesis and turnover are altered in breast cancer and how these changes offer potential strategies to improve breast cancer therapy.
Collapse
Affiliation(s)
- Purab Pal
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - G. Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, The State University of New York (SUNY), Buffalo, NY 14260, USA
- Correspondence: (G.E.A.-G.); (J.F.)
| | - Jonna Frasor
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Correspondence: (G.E.A.-G.); (J.F.)
| |
Collapse
|
4
|
Kyriakou S, Cheung W, Mantso T, Mitsiogianni M, Anestopoulos I, Veuger S, Trafalis DT, Franco R, Pappa A, Tetard D, Panayiotidis MI. A novel methylated analogue of L-Mimosine exerts its therapeutic potency through ROS production and ceramide-induced apoptosis in malignant melanoma. Invest New Drugs 2021; 39:971-986. [PMID: 33624234 PMCID: PMC8280034 DOI: 10.1007/s10637-021-01087-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/11/2021] [Indexed: 01/01/2023]
Abstract
Melanoma is an aggressive and highly metastatic type of skin cancer where the design of new therapies is of utmost importance for the clinical management of the disease. Thus, we have aimed to investigate the mode of action by which a novel methylated analogue of L-Mimosine (e.g., L-SK-4) exerts its therapeutic potency in an in vitro model of malignant melanoma. Cytotoxicity was assessed by the Alamar Blue assay, oxidative stress by commercially available kits, ROS generation, caspase 3/7 activation and mitochondrial membrane depolarisation by flow cytometry, expression of apoptosis-related proteins by western immunoblotting and profiling of lipid biosynthesis by a metabolomic approach. Overall, higher levels of ROS, sphingolipids and apoptosis were induced by L-SK-4 suggesting that the compound's therapeutic potency is mediated through elevated ROS levels which promote the upregulation of sphingolipid (ceramide) biosynthesis thus leading to the activation of both extrinsic and intrinsic apoptosis, in an experimental model of malignant melanoma.
Collapse
Affiliation(s)
- Sotiris Kyriakou
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
- The Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - William Cheung
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Theodora Mantso
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Melina Mitsiogianni
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Ioannis Anestopoulos
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
- The Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Stephany Veuger
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Dimitris T Trafalis
- Department of Pharmacology, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Rodrigo Franco
- Redox Biology Centre, University of Nebraska, Lincoln, USA
- School of Veterinary Medicine & Biomedical Sciences, University of Nebraska, Lincoln, USA
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - David Tetard
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Mihalis I Panayiotidis
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK.
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus.
- The Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus.
| |
Collapse
|
5
|
Mishra R, Upadhyay A, Prajapati VK, Mishra A. Proteasome-mediated proteostasis: Novel medicinal and pharmacological strategies for diseases. Med Res Rev 2018; 38:1916-1973. [DOI: 10.1002/med.21502] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/13/2018] [Accepted: 04/04/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Ribhav Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| | - Vijay Kumar Prajapati
- Department of Biochemistry; School of Life Sciences; Central University of Rajasthan; Rajasthan India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| |
Collapse
|
6
|
Dubois N, Rio E, Ripoche N, Ferchaud-Roucher V, Gaugler MH, Campion L, Krempf M, Carrie C, Mahé M, Mirabel X, Paris F. Plasma ceramide, a real-time predictive marker of pulmonary and hepatic metastases response to stereotactic body radiation therapy combined with irinotecan. Radiother Oncol 2016; 119:229-35. [PMID: 27113798 DOI: 10.1016/j.radonc.2016.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 03/15/2016] [Accepted: 03/18/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSES Early biomarkers of tumour response are needed to discriminate between responders and non-responders to radiotherapy. We evaluated the ability of ceramide, a bioactive sphingolipid, to predict tumour sensitivity in patients treated by hypofractionated stereotactic body radiation therapy (SBRT) combined with irinotecan chemotherapy. MATERIALS AND METHODS Plasma levels of total ceramide and of its subspecies were measured before and during treatment in 35 patients with liver and lung oligometastases of colorectal cancer included in a phase II trial. Cer levels were quantified by LC-ESI-MS/MS and compared to tumour volume response evaluated one year later by CT-scan. RESULTS Pretreatment plasma ceramide levels were not indicative of tumour response. Nevertheless, the levels of total ceramide and of its 4 main subspecies were significantly higher at days 3 and 10 of treatment in objective responders than in non-responders. According to Kaplan-Meier curves, almost complete tumour control was achieved at 1year in patients with increased total ceramide levels whereas 50% of patients with decreased levels experienced an increase in tumour volume. CONCLUSIONS Total plasma ceramide is a promising biomarker of tumour response to SBRT combined with irinotecan that should enable to segregate patients with high risk of tumour escape.
Collapse
Affiliation(s)
- Nolwenn Dubois
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France; Inserm, UMR892, Nantes, France; Université de Nantes, France; CNRS, UMR 6299, Nantes, France
| | - Emmanuel Rio
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Natacha Ripoche
- Inserm, UMR892, Nantes, France; Université de Nantes, France; CNRS, UMR 6299, Nantes, France
| | | | - Marie-Hélène Gaugler
- Inserm, UMR892, Nantes, France; Université de Nantes, France; CNRS, UMR 6299, Nantes, France
| | - Loic Campion
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Michel Krempf
- Université de Nantes, France; INRA, UMR 1280, Nantes, France
| | | | - Marc Mahé
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | | | - François Paris
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France; Inserm, UMR892, Nantes, France; Université de Nantes, France; CNRS, UMR 6299, Nantes, France.
| |
Collapse
|
7
|
Abstract
Studies over the past two decades have identified ceramide as a multifunctional central molecule in the sphingolipid biosynthetic pathway. Given its diverse tumor suppressive activities, molecular understanding of ceramide action will produce fundamental insights into processes that limit tumorigenesis and may identify key molecular targets for therapeutic intervention. Ceramide can be activated by a diverse array of stresses such as heat shock, genotoxic damage, oxidative stress and anticancer drugs. Ceramide triggers a variety of tumor suppressive and anti-proliferative cellular programs such as apoptosis, autophagy, senescence, and necroptosis by activating or repressing key effector molecules. Defects in ceramide generation and metabolism in cancer contribute to tumor cell survival and resistance to chemotherapy. The potent and versatile anticancer activity profile of ceramide has motivated drug development efforts to (re-)activate ceramide in established tumors. This review focuses on our current understanding of the tumor suppressive functions of ceramide and highlights the potential downstream targets of ceramide which are involved in its tumor suppressive action.
Collapse
|
8
|
The Emerging Role of Extracellular Vesicle-Mediated Drug Resistance in Cancers: Implications in Advanced Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:454837. [PMID: 26587537 PMCID: PMC4637461 DOI: 10.1155/2015/454837] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 07/05/2015] [Indexed: 01/07/2023]
Abstract
Emerging evidence has shown that the extracellular vesicles (EVs) regulate various biological processes and can control cell proliferation and survival, as well as being involved in normal cell development and diseases such as cancers. In cancer treatment, development of acquired drug resistance phenotype is a serious issue. Recently it has been shown that the presence of multidrug resistance proteins such as Pgp-1 and enrichment of the lipid ceramide in EVs could have a role in mediating drug resistance. EVs could also mediate multidrug resistance through uptake of drugs in vesicles and thus limit the bioavailability of drugs to treat cancer cells. In this review, we discussed the emerging evidence of the role EVs play in mediating drug resistance in cancers and in particular the role of EVs mediating drug resistance in advanced prostate cancer. The role of EV-associated multidrug resistance proteins, miRNA, mRNA, and lipid as well as the potential interaction(s) among these factors was probed. Lastly, we provide an overview of the current available treatments for advanced prostate cancer, considering where EVs may mediate the development of resistance against these drugs.
Collapse
|
9
|
Cheng JC, Bai A, Beckham TH, Marrison ST, Yount CL, Young K, Lu P, Bartlett AM, Wu BX, Keane BJ, Armeson KE, Marshall DT, Keane TE, Smith MT, Jones EE, Drake RR, Bielawska A, Norris JS, Liu X. Radiation-induced acid ceramidase confers prostate cancer resistance and tumor relapse. J Clin Invest 2013; 123:4344-58. [PMID: 24091326 DOI: 10.1172/jci64791] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 07/11/2013] [Indexed: 01/06/2023] Open
Abstract
Escape of prostate cancer (PCa) cells from ionizing radiation-induced (IR-induced) killing leads to disease progression and cancer relapse. The influence of sphingolipids, such as ceramide and its metabolite sphingosine 1-phosphate, on signal transduction pathways under cell stress is important to survival adaptation responses. In this study, we demonstrate that ceramide-deacylating enzyme acid ceramidase (AC) was preferentially upregulated in irradiated PCa cells. Radiation-induced AC gene transactivation by activator protein 1 (AP-1) binding on the proximal promoter was sensitive to inhibition of de novo ceramide biosynthesis, as demonstrated by promoter reporter and ChIP-qPCR analyses. Our data indicate that a protective feedback mechanism mitigates the apoptotic effect of IR-induced ceramide generation. We found that deregulation of c-Jun induced marked radiosensitization in vivo and in vitro, which was rescued by ectopic AC overexpression. AC overexpression in PCa clonogens that survived a fractionated 80-Gy IR course was associated with increased radioresistance and proliferation, suggesting a role for AC in radiotherapy failure and relapse. Immunohistochemical analysis of human PCa tissues revealed higher levels of AC after radiotherapy failure than those in therapy-naive PCa, prostatic intraepithelial neoplasia, or benign tissues. Addition of an AC inhibitor to an animal model of xenograft irradiation produced radiosensitization and prevention of relapse. These data indicate that AC is a potentially tractable target for adjuvant radiotherapy.
Collapse
|
10
|
Mukhopadhyay A, Tabanor K, Chaguturu R, Aldrich JV. Targeting inhibitor 2 of protein phosphatase 2A as a therapeutic strategy for prostate cancer treatment. Cancer Biol Ther 2013; 14:962-72. [PMID: 24025258 DOI: 10.4161/cbt.25943] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inhibitor 2 of protein phosphatase 2A (I2PP2A), a biological inhibitor of the cellular serine/threonine protein phosphatase PP2A, is associated with numerous cellular processes that often lead to the formation and progression of cancer. In this study we hypothesized that targeting the inhibition of I2PP2A's multiple functions in prostate cancer cells might prevent cancer progression. We have investigated the effect of the small chain C6-ceramide, known to be a bioactive tumor suppressor lipid, on I2PP2A function, thereby affecting c-Myc signaling and histone acetylation in cells. Our data indicated that C6-ceramide treatment of prostate cancer cells induces cell death in PC-3, DU145, and LNCaP cells, but not normal prostate epithelial cells. C6-ceramide was able to disrupt the association between PP2A and I2PP2A. C6-ceramide inhibits I2PP2A's upregulation of c-Myc and downregulation of histone acetylation in prostate cancer cells. Our data indicated that targeting cancer related signaling pathways through I2PP2A using ceramide as an anti-I2PP2A agent could have beneficial effects as a therapeutic approach to prevent prostate cancer.
Collapse
|
11
|
Abstract
There are several well-established mechanisms involved in radiation-induced cell death in mammalian cell systems. The p53-mediated apoptotic pathway is the most widely recognized mechanism (Lowe et al. Nature 362:847-849, 1993), although apoptosis has long been considered a less relevant mechanism of radiation-induced cell death (Steel, Acta Oncol 40:968-975, 2001; Brown and Wouters, Cancer Res 59:1391-1399, 1999; Olive and Durand, Int J Radiat Biol 71:695-707, 1997). We and others have recently focused instead on the emerging links between radiation, apoptosis, and ceramide and showed that ceramide is a sphingolipid-derived second messenger capable of initiating apoptotic cascades in response to various stress stimuli, including radiation.Ceramide, the backbone of all sphingolipids, is synthesized by a family of ceramide synthases (CerS), each using acyl-CoAs of defined chain length for N-acylation of the sphingoid long-chain base. Six mammalian CerS homologs have been cloned that demonstrated high selectivity towards acyl-CoAs (Lahiri et al. FEBS Lett 581:5289-5294, 2007), and more recently, it was shown that their activity can be modulated by dimer formation (Mesicek et al. Cell Signal 22:1300-1307, 2010; Laviad et al. J Biol Chem 283:5677-5684, 2008).This de novo ceramide synthesis has been observed in irradiated cells through a pathway normally suppressed by ataxia telangiectasia-mutated (ATM) protein, a key component of the cellular response to DNA double-strand breaks (Liao et al. J Biol Chem 274:17908-17917, 1999). ATM is not the sole factor known to affect apoptotic potential by modulating CerS activity. Recent work has also implicated protein kinase Cα (PKCα) as a potential CerS activator (Truman et al. Cancer Biol Ther 8:54-63, 2009).In this review, we summarize involvement of CerS in sphingolipid-mediated apoptosis in irradiated human prostate cancer cells and discuss future directions in this field.
Collapse
Affiliation(s)
- Carla Hajj
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | | |
Collapse
|
12
|
Abstract
One crucial barrier to progress in the treatment of cancer has been the inability to control the balance between cell proliferation and apoptosis: enter ceramide. Discoveries over the past 15 years have elevated this sphingolipid to the lofty position of a regulator of cell fate. Ceramide, it turns out, is a powerful tumour suppressor, potentiating signalling events that drive apoptosis, autophagic responses and cell cycle arrest. However, defects in ceramide generation and metabolism in cancer cells contribute to tumour cell survival and resistance to chemotherapy. This Review focuses on ceramide signalling and the targeting of specific metabolic junctures to amplify the tumour suppressive activities of ceramide. The potential of ceramide-based therapeutics in the treatment of cancer is also discussed.
Collapse
Affiliation(s)
- Samy A F Morad
- Department of Experimental Therapeutics, John Wayne Cancer Institute at Saint John's Health Center, 2200 Santa Monica Boulevard, Santa Monica, California 90404, USA.
| | | |
Collapse
|
13
|
Qin JD, Weiss L, Slavin S, Gatt S, Dagan A. Synthetic, non-natural analogs of ceramide elevate cellular ceramide, inducing apoptotic death to prostate cancer cells and eradicating tumors in mice. Cancer Invest 2010; 28:535-43. [PMID: 20014940 DOI: 10.3109/07357900903478915] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The anticancer effects of synthetic, non-natural analogs of ceramide were tested using human TSU-Pr1 prostate cancer cells in-vitro as well as in-vivo, following their effects on tumors development in mice. When incubated with the cultured cancer cells, the analogs elevated cellular ceramide and induced a cytotoxicity and death by apoptosis. When a ceramide analog was injected intradermally or intraperitoneally into BALB/c-Nude or NOD-SCID mice bearing a human prostate tumor, a considerable regression of the tumor was observed. The synthetic ceramide analogs should thus be further investigated as potential anticancer drugs.
Collapse
Affiliation(s)
- Jing Dong Qin
- Department of Biochemistry, Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
14
|
Mahdy AEM, Cheng JC, Li J, Elojeimy S, Meacham WD, Turner LS, Bai A, Gault CR, McPherson AS, Garcia N, Beckham TH, Saad A, Bielawska A, Bielawski J, Hannun YA, Keane TE, Taha MI, Hammouda HM, Norris JS, Liu X. Acid ceramidase upregulation in prostate cancer cells confers resistance to radiation: AC inhibition, a potential radiosensitizer. Mol Ther 2008; 17:430-8. [PMID: 19107118 DOI: 10.1038/mt.2008.281] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Radiation resistance in a subset of prostate tumors remains a challenge to prostate cancer radiotherapy. The current study on the effects of radiation on prostate cancer cells reveals that radiation programs an unpredicted resistance mechanism by upregulating acid ceramidase (AC). Irradiated cells demonstrated limited changes of ceramide levels while elevating levels of sphingosine and sphingosine-1-phosphate. By genetically downregulating AC with small interfering RNA (siRNA), we observed radiosensitization of cells using clonogenic and cytotoxicity assays. Conversely, AC overexpression further decreased sensitivity to radiation. We also observed that radiation-induced AC upregulation was sufficient to create cross-resistance to chemotherapy as demonstrated by decreased sensitivity to Taxol and C(6) ceramide compared to controls. Lower levels of caspase 3/7 activity were detected in cells pretreated with radiation, also indicating increased resistance. Finally, utilization of the small molecule AC inhibitor, LCL385, sensitized PPC-1 cells to radiation and significantly decreased tumor xenograft growth. These data suggest a new mechanism of cancer cell resistance to radiation, through upregulation of AC that is, in part, mediated by application of the therapy itself. An improved understanding of radiotherapy and the application of combination therapy achieved in this study offer new opportunities for the modulation of radiation effects in the treatment of cancer.
Collapse
Affiliation(s)
- Ayman E M Mahdy
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Leroux ME, Auzenne E, Evans R, Hail N, Spohn W, Ghosh SC, Farquhar D, McDonnell T, Klostergaard J. Sphingolipids and the sphingosine kinase inhibitor, SKI II, induce BCL-2-independent apoptosis in human prostatic adenocarcinoma cells. Prostate 2007; 67:1699-717. [PMID: 17879964 DOI: 10.1002/pros.20645] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Elevated BCL-2 is one mechanism of therapeutic resistance in prostate cancer (PC), and new approaches are needed to overcome such resistance. METHODS We evaluated the effects of BCL-2 over-expression in human prostatic adenocarcinoma cells on their susceptibility to sphingolipids (SLs) and to the sphingosine kinase (SpK) inhibitor, SKI II. RESULTS In survival assays, no significant differences were observed in the responses to sphingosine or ceramide among parental PC-3 cells lacking detectable BCL-2 and BCL-2 over-expressing PC-3 transfectants; similarly, the responses to dimethyl-sphingosine (DMSP) of parental LNCaP cells and a BCL-2 over-expressing LNCaP transfectant were equivalent. SKI II induced protracted, BCL-2-independent survival loss in both PC-3 and LNCaP parental/transfectant pairs; in contrast, DMSP induced rapid cell shrinkage, caspase activation and caspase-dependent DNA fragmentation. DMSP-induced DNA fragmentation and loss of mitochondrial membrane potential were equivalent in BCL-2 transfectants and parental PC-3 cells and were not associated with BCL-2 downregulation. DMSP-mediated cytotoxicity was not associated with the enhanced production of reactive oxygen intermediates. SL analyses of parental and transfectant PC-3 cells did not reveal increased levels of sphingosine-1-phosphate in the BCL-2 transfectants; further, there only a modest early shift, corresponding to apoptotic onset, in pro- versus anti-apoptotic SLs in response to DMSP treatment. CONCLUSIONS Thus, in contrast to the inhibitory effects of BCL-2 on apoptosis induced by various agents in tumor cells, SKI II and selected pro-apoptotic SLs appear atypical in their independence from such inhibition, and may have merits as new candidates for treatment of AI PC.
Collapse
Affiliation(s)
- M E Leroux
- Department of Molecular and Cellular Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Lin CF, Chen CL, Chiang CW, Jan MS, Huang WC, Lin YS. GSK-3beta acts downstream of PP2A and the PI 3-kinase-Akt pathway, and upstream of caspase-2 in ceramide-induced mitochondrial apoptosis. J Cell Sci 2007; 120:2935-43. [PMID: 17666435 DOI: 10.1242/jcs.03473] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The signaling of glycogen synthase kinase-3beta (GSK-3beta) has been implicated in stress-induced apoptosis. However, the pro-apoptotic role of GSK-3beta is still unclear. Here, we show the involvement of GSK-3beta in ceramide-induced mitochondrial apoptosis. Ceramide induced GSK-3beta activation via protein dephosphorylation at serine 9. We previously reported that ceramide induced caspase-2 and caspase-8 activation, Bid cleavage, mitochondrial damage, and apoptosis. In this study, we found that caspase-2 activation and the subsequent apoptotic events were abolished by the GSK-3beta inhibitors lithium chloride and SB216763, and by GSK-3beta knockdown using short interfering RNA. We also found that ceramide-activated protein phosphatase 2A (PP2A) indirectly caused GSK-3beta activation, and that the PP2A-regulated PI 3-kinase-Akt pathway was involved in GSK-3beta activation. These results indicate a role for GSK-3beta in ceramide-induced apoptosis, in which GSK-3beta acts downstream of PP2A and the PI 3-kinase-Akt pathway, and upstream of caspase-2 and caspase-8.
Collapse
Affiliation(s)
- Chiou-Feng Lin
- Department of Microbiology and Immunology, National Cheng Kung University Medical College, Tainan 701, Taiwan
| | | | | | | | | | | |
Collapse
|
17
|
Scarlatti F, Sala G, Ricci C, Maioli C, Milani F, Minella M, Botturi M, Ghidoni R. Resveratrol sensitization of DU145 prostate cancer cells to ionizing radiation is associated to ceramide increase. Cancer Lett 2007; 253:124-30. [PMID: 17321671 DOI: 10.1016/j.canlet.2007.01.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 01/18/2007] [Accepted: 01/19/2007] [Indexed: 02/06/2023]
Abstract
Radiotherapy is an established therapeutic modality for prostate cancer. Since it is well known that radiotherapy is limited due to its severe toxicity towards normal cells at high dose and minimal effect at low dose, the search for biological compounds that increase the sensitivity of tumors cells to radiation may improve the efficacy of therapy. Resveratrol, a natural antioxidant, was shown to inhibit carcinogenesis in animal models, and to block the process of tumor initiation and progression. The purpose of this study was to examine whether or not resveratrol can sensitize DU145, an androgen-independent human prostate cancer cell line, to ionizing radiation. We report here that DU145 cells are resistant to ionizing radiation-induced cell death, but pretreatment with resveratrol significantly enhances cell death. Resveratrol acts synergistically with ionizing radiation to inhibit cell survival in vitro. Resveratrol also potentiates ionizing radiation-induced ceramide accumulation, by promoting its de novo biosynthesis. This confirms ceramide as an effective mediator of the anticancer potential induced by resveratrol.
Collapse
Affiliation(s)
- Francesca Scarlatti
- Laboratory of Biochemistry & Mol Biology, San Paolo Medical School, University of Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Han Y, Kim Y, Kang H, Hong SH, Kim YH, Lim DS, Park C, Yun YS, Song JY. N-acetylphytosphingosine-induced apoptosis of Jurkat cells is mediated by the conformational change in Bak. Apoptosis 2006; 11:581-8. [PMID: 16528476 DOI: 10.1007/s10495-006-4569-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
N-acetylphytosphingosine (NAPS), a sphingolipid derivative, is one of the well-known signal molecules that mediates various cellular functions, including cell growth, differentiation, and apoptosis. In this study, we demonstrated that NAPS induces apoptosis of Jurkat cells by activating Bak, but not Bax, which are both members of a proapoptotic subfamily of the Bcl-2 proteins. NAPS activated caspase-8 in a FADD-independent manner, but the lack of caspase-8 did not suppress the activation of caspase-3 and -9 and cell death, indicating that caspase-8 activation does not play an important role in NAPS-induced cell death. The overexpression of Bcl-xL, an anti-apoptotic protein, completely inhibited the activation of the caspases and apoptosis, assuming that NAPS-induced apoptosis was initiated by the mitochondria. The expression levels of pro- and anti-apoptotic Bcl-2 family members were not changed by the NAPS treatment. However, Bad was translocated from the cytosol into the mitochondria, where it bound to Bcl-xL, and Bak was dissociated from Bcl-xL and conformationally changed. Taken together, these findings indicate that NAPS induced apoptosis of Jurkat cells in a mitochondria-dependent manner that was controlled by the translocation of Bad and the conformational change in Bak.
Collapse
Affiliation(s)
- Y Han
- Laboratory of Radiation Immunology, Korea Institute of Radiological and Medical Sciences, KAERI, Gongneung-Dong, Nowon-Gu, Seoul, 139-706, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Schmelz EM, Symolon H. Sphingolipids and Cancer. SPHINGOLIPID BIOLOGY 2006:363-381. [DOI: 10.1007/4-431-34200-1_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
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] [Scholar 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
|
21
|
Mimeault M, Batra SK. Recent advances on multiple tumorigenic cascades involved in prostatic cancer progression and targeting therapies. Carcinogenesis 2005; 27:1-22. [PMID: 16195239 DOI: 10.1093/carcin/bgi229] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recent advances on differently-expressed gene products and their functions during the progression from localized androgen-dependent states into androgen-independent and metastatic forms of prostate cancer are reported. The expression levels of numerous oncogenes and tumor suppressor genes in distinct prostatic cancer epithelial cell lines and tissues relative to normal prostate cells are described. This is carried out to identify the signaling elements that are altered during the initiation, progression and metastatic process of prostate cancer. Additional information on the interactions between certain deregulated signaling pathways such as androgen receptor (AR), estrogen receptors, epidermal growth factor receptor (EGFR), hedgehog and Wnt/beta-catenin cascades in controlling the proliferation, survival and invasion of tumor prostate epithelial cells during the disease progression is described. The emphasis is on the critical functions of the AR and EGF-EGFR systems at all stages during prostate carcinogenesis. Of therapeutic interest, new strategies for the diagnosis and treatment of localized and metastatic forms of prostate cancer by targeting multiple tumorigenic signaling elements are also reported.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | |
Collapse
|
22
|
Gonzalez-Guerrico AM, Kazanietz MG. Phorbol ester-induced apoptosis in prostate cancer cells via autocrine activation of the extrinsic apoptotic cascade: a key role for protein kinase C delta. J Biol Chem 2005; 280:38982-91. [PMID: 16183650 DOI: 10.1074/jbc.m506767200] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
It is well established that activation of protein kinase C (PKC) by phorbol esters promotes apoptosis in androgen-dependent prostate cancer cells. However, there is limited information regarding the cellular mechanisms involved in this effect. In this report we identified a novel autocrine pro-apoptotic loop triggered by PKCdelta activation in prostate cancer cells that is mediated by death receptor ligands. The apoptotic effect of phorbol 12-myristate 13-acetate in LNCaP cells was impaired by inhibition or depletion of tumor necrosis factor alpha-converting enzyme, the enzyme responsible for tumor necrosis factor alpha (TNFalpha) shedding. Moreover, the apoptogenic effect of conditioned medium collected after phorbol 12-myristate 13-acetate treatment could be inhibited by blocking antibodies against TNFalpha and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), but not FasL, as well as by RNA interference depletion of TNFalpha and TRAIL receptors. Moreover, depletion or inhibition of death receptor downstream effectors, including caspase-8, FADD, p38 MAPK, and JNK, significantly reduced the apoptogenic effect of the conditioned medium. PKCdelta played a major role in this autocrine loop, both in the secretion of autocrine factors as well as a downstream effector. Taken together, our results demonstrate that activation of PKCdelta in prostate cancer cells causes apoptosis via the release of death receptor ligands and the activation of the extrinsic apoptotic cascade.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Apoptosis/drug effects
- Apoptosis Regulatory Proteins/antagonists & inhibitors
- Apoptosis Regulatory Proteins/genetics
- Base Sequence
- Cell Line, Tumor
- Enzyme Activation/drug effects
- Humans
- MAP Kinase Signaling System/drug effects
- Male
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/genetics
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Protein Kinase C-delta/metabolism
- RNA Interference
- RNA, Neoplasm/genetics
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type II/genetics
- TNF-Related Apoptosis-Inducing Ligand
- Tetradecanoylphorbol Acetate/pharmacology
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Anatilde M Gonzalez-Guerrico
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6160, USA
| | | |
Collapse
|
23
|
Abstract
In most cell types, a key event in apoptosis is the release of proapoptotic intermembrane space proteins from mitochondria to the cytoplasm. In general, it is the release of these intermembrane space proteins that is responsible for the activation of caspases and DNases that are responsible for the execution of apoptosis. The mechanism for the increased permeability of the mitochondrial outer membrane during the induction phase of apoptosis is currently unknown and highly debated. This review will focus on one such proposed mechanism, namely, the formation of ceramide channels in the mitochondrial outer membrane. Ceramides are known to play a major regulatory role in apoptosis by inducing the release of proapoptotic proteins from the mitochondria. As mitochondria are known to contain the enzymes responsible for the synthesis and hydrolysis of ceramide, there exists a mechanism for regulating the level of ceramide in mitochondria. In addition, mitochondrial ceramide levels have been shown to be elevated prior to the induction phase of apoptosis. Ceramide has been shown to form large protein permeable channels in planar phospholipid and mitochondrial outer membranes. Thus, ceramide channels are good candidates for the pathway with which proapoptotic proteins are released from mitochondria during the induction phase of apoptosis.
Collapse
Affiliation(s)
- Leah J Siskind
- Department of Biology, University of Maryland-College Park, College Park, MD 20742, USA.
| |
Collapse
|
24
|
Hara S, Nakashima S, Kiyono T, Sawada M, Yoshimura S, Iwama T, Banno Y, Shinoda J, Sakai N. p53-Independent ceramide formation in human glioma cells during gamma-radiation-induced apoptosis. Cell Death Differ 2005; 11:853-61. [PMID: 15088070 DOI: 10.1038/sj.cdd.4401428] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Although the p53 tumor-suppressor gene product plays a critical role in apoptotic cell death induced by DNA-damaging chemotherapeutic agents, human glioma cells with functional p53 were more resistant to gamma-radiation than those with mutant p53. U-87 MG cells with wild-type p53 were resistant to gamma-radiation. U87-W E6 cells that lost functional p53, by the expression of type 16 human papillomavirus E6 oncoprotein, became susceptible to radiation-induced apoptosis. The formation of ceramide by acid sphingomyelinase (A-SMase), but not by neutral sphingomyelinase, was associated with p53-independent apoptosis. SR33557 (2-isopropyl-1-(4-[3-N-methyl-N-(3,4-dimethoxybphenethyl)amino]propyloxy)benzene-sulfonyl) indolizine, an inhibitor of A-SMase, suppressed radiation-induced apoptotic cell death. In contrast, radiation-induced A-SMase activation was blocked in glioma cells with endogenous functional p53. The expression of acid ceramidase was induced by gamma-radiation, and was more evident in cells with functional p53. N-oleoylethanolamine, which is known to inhibit ceramidase activity, unexpectedly downregulated acid ceramidase and accelerated radiation-induced apoptosis in U87-W E6 cells. Moreover, cells with functional p53 could be sensitized to gamma-radiation by N-oleoylethanolamine, which suppressed radiation-induced acid ceramidase expression and then enhanced ceramide formation. Sensitization to gamma-radiation was also observed in U87-MG cells depleted of functional p53 by retroviral expression of small interfering RNA. These results indicate that ceramide may function as a mediator of p53-independent apoptosis in human glioma cells in response to gamma-radiation, and suggest that p53-dependent expression of acid ceramidase and blockage of A-SMase activation play pivotal roles in protection from gamma-radiation of cells with endogenous functional p53.
Collapse
Affiliation(s)
- S Hara
- Department of Neurosurgery, Gifu University School of Medicine, Tsukasamachi-40, Gifu 500-8705, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Schultz A, Larsson C. Ceramide influences neurite outgrowth and neuroblastoma cell apoptosis regulated by novel protein kinase C isoforms. J Neurochem 2004; 89:1427-35. [PMID: 15189345 DOI: 10.1111/j.1471-4159.2004.02431.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have previously seen that protein kinase C (PKC) epsilon induces neurite outgrowth and that PKCdelta and PKCtheta elicit apoptosis in neuroblastoma cells. In this study we investigate the effects of cell-permeable C(2)-ceramide on these events in SK-N-BE(2) neuroblastoma cells. C(2)-ceramide abolishes neurite formation induced by overexpression of PKCepsilon and, in cells overexpressing PKCdelta or PKCtheta, ceramide treatment leads to apoptosis. Exposure to C(2)-ceramide also suppressed neurite outgrowth induced by retinoic acid, but ceramide did not abrogate neurite induction by treatment with the ROCK inhibitor Y-27632, demonstrating that C(2)-ceramide is not a general inhibitor of neurite outgrowth. The neurite-suppressing effect occurs independently of cell-death. Furthermore, C(2)-ceramide relocated PKCepsilon and the isolated regulatory domain of PKCepsilon from the cytosol to the perinuclear region. In contrast, neither the localization of PKCdelta nor of PKCtheta was affected by C(2)-ceramide. Taken together, the data indicate that the neurite-inhibiting effect of C(2)-ceramide treatment may be caused by a re-localization of PKCepsilon and thus identify a functional consequence of ceramide effects on PKCepsilon localization.
Collapse
Affiliation(s)
- Anna Schultz
- Molecular Medicine, Lund University, 205-02 Malmö, Sweden
| | | |
Collapse
|
26
|
Alphonse G, Bionda C, Aloy MT, Ardail D, Rousson R, Rodriguez-Lafrasse C. Overcoming resistance to gamma-rays in squamous carcinoma cells by poly-drug elevation of ceramide levels. Oncogene 2004; 23:2703-15. [PMID: 15048093 DOI: 10.1038/sj.onc.1207357] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent strategies to sensitize radioresistant tumours are based on combining gamma-irradiation with inducers of apoptosis. We report that the combination of three inhibitors of sphingolipid metabolism, DL-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol.HCl(DL-PDMP)+imipramine +/- D-erythro-2-(N-myristoylamino)-1-phenyl-1-propanol (D-MAPP), with 10-Gy irradiation triggers both mitotic and apoptotic killing in radioresistant SQ20B squamous carcinoma cells. In these cells, apoptosis is defective due to a lack of ceramide generation upstream, which cannot be explained by sphingomyelinase (neutral and acidic) deficiency or rapid derivation to the sphingolipid pathway. We present evidence of a functional transduction death pathway when ceramide generation is restored, which involves the mitochondrial-mediated pathway coupled to alterations in redox status and to executive caspases activation. The poly-drug treatment restored apoptosis to levels similar to those observed in radiosensitive SCC61 squamous carcinoma cells. Simultaneous exposure to gamma-irradiation and poly-drug treatment acted synergistically in SQ20B cells to produce a marked increase in both mitochondrial dysfunction and caspase cleavage, which led to a 7.8-fold increase in apoptosis within 48 h, relative to irradiated cells. Moreover, the results suggest that the ceramide released by irradiation or poly-drug treatment converges upon common cellular targets. Modulation of endogenous ceramide levels by inhibitors of sphingolipid metabolism may represent a new cellular target for the sensitization of radioresistant tumours to gamma-ray therapy.
Collapse
Affiliation(s)
- Gersende Alphonse
- Department of Biochemistry, INSERM U189, Lyon-Sud Medical School, BP12, 69921 Oullins Cedex, France
| | | | | | | | | | | |
Collapse
|
27
|
Reynolds CP, Maurer BJ, Kolesnick RN. Ceramide synthesis and metabolism as a target for cancer therapy. Cancer Lett 2004; 206:169-80. [PMID: 15013522 DOI: 10.1016/j.canlet.2003.08.034] [Citation(s) in RCA: 245] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2003] [Accepted: 08/14/2003] [Indexed: 12/29/2022]
Abstract
Sphingolipids, which include ceramides and sphingosine, are essential structural components of cell membranes that also have messenger functions that regulate the proliferation, survival, and death of cells. Exogenous application of ceramide is cytotoxic, and exposure of cells to radiation or chemotherapy is associated with increased ceramide levels due to enhanced de novo synthesis, catabolism of sphingomyelin, or both. Ceramide can be metabolized to less toxic forms by glycosylation, acylation, or by catabolism to sphingosine, which is then phosphorylated to the anti-apoptotic sphingosine 1-phosphate. Glucosylceramide synthase overexpression has been shown to enhance resistance to doxorubicin, suggesting that inhibition of ceramide metabolism or catabolism might enhance cancer chemotherapy. Several anticancer agents, including the cytotoxic retinoid, fenretinide (4-HPR), have been shown to act, at least in part, by increasing tumor cell ceramide via de novo synthesis. Combinations of 4-HPR and modulators of ceramide action and/or metabolism demonstrated increased anti-tumor activity in pre-clinical models with minimal toxicity for non-malignant cells, and were effective in a p53-independent manner against tumor cell lines resistant to standard cytotoxic agents. Phase I trials of ceramide metabolism inhibitors in combination with 4-HPR and with other cytotoxic agents are in development. Thus, pharmacological manipulation of sphingolipid metabolism to enhance tumor cell ceramide is being realized and offers a novel approach to cancer chemotherapy.
Collapse
Affiliation(s)
- C Patrick Reynolds
- Division of Hematology-Oncology MS 57, Children's Hospital of Los Angeles, The University of Southern California Keck School of Medicine, 4650 Sunset Blvd., Los Angeles, CA 90054-0700, USA.
| | | | | |
Collapse
|