1
|
Ashrafizadeh M, Paskeh MDA, Mirzaei S, Gholami MH, Zarrabi A, Hashemi F, Hushmandi K, Hashemi M, Nabavi N, Crea F, Ren J, Klionsky DJ, Kumar AP, Wang Y. Targeting autophagy in prostate cancer: preclinical and clinical evidence for therapeutic response. J Exp Clin Cancer Res 2022; 41:105. [PMID: 35317831 PMCID: PMC8939209 DOI: 10.1186/s13046-022-02293-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/08/2023] Open
Abstract
Prostate cancer is a leading cause of death worldwide and new estimates revealed prostate cancer as the leading cause of death in men in 2021. Therefore, new strategies are pertinent in the treatment of this malignant disease. Macroautophagy/autophagy is a “self-degradation” mechanism capable of facilitating the turnover of long-lived and toxic macromolecules and organelles. Recently, attention has been drawn towards the role of autophagy in cancer and how its modulation provides effective cancer therapy. In the present review, we provide a mechanistic discussion of autophagy in prostate cancer. Autophagy can promote/inhibit proliferation and survival of prostate cancer cells. Besides, metastasis of prostate cancer cells is affected (via induction and inhibition) by autophagy. Autophagy can affect the response of prostate cancer cells to therapy such as chemotherapy and radiotherapy, given the close association between autophagy and apoptosis. Increasing evidence has demonstrated that upstream mediators such as AMPK, non-coding RNAs, KLF5, MTOR and others regulate autophagy in prostate cancer. Anti-tumor compounds, for instance phytochemicals, dually inhibit or induce autophagy in prostate cancer therapy. For improving prostate cancer therapy, nanotherapeutics such as chitosan nanoparticles have been developed. With respect to the context-dependent role of autophagy in prostate cancer, genetic tools such as siRNA and CRISPR-Cas9 can be utilized for targeting autophagic genes. Finally, these findings can be translated into preclinical and clinical studies to improve survival and prognosis of prostate cancer patients. • Prostate cancer is among the leading causes of death in men where targeting autophagy is of importance in treatment; • Autophagy governs proliferation and metastasis capacity of prostate cancer cells; • Autophagy modulation is of interest in improving the therapeutic response of prostate cancer cells; • Molecular pathways, especially involving non-coding RNAs, regulate autophagy in prostate cancer; • Autophagy possesses both diagnostic and prognostic roles in prostate cancer, with promises for clinical application.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey.
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396, Istanbul, Turkey
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, 1417466191, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine University of Tehran, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Francesco Crea
- Cancer Research Group-School of Life Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Jun Ren
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.,Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Daniel J Klionsky
- Life Sciences Institute & Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Yuzhuo Wang
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada.
| |
Collapse
|
2
|
Bataller M, Sánchez-García A, Garcia-Mayea Y, Mir C, Rodriguez I, LLeonart ME. The Role of Sphingolipids Metabolism in Cancer Drug Resistance. Front Oncol 2022; 11:807636. [PMID: 35004331 PMCID: PMC8733468 DOI: 10.3389/fonc.2021.807636] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/07/2021] [Indexed: 12/25/2022] Open
Abstract
Drug resistance continues to be one of the major challenges to cure cancer. As research in this field evolves, it has been proposed that numerous bioactive molecules might be involved in the resistance of cancer cells to certain chemotherapeutics. One well-known group of lipids that play a major role in drug resistance are the sphingolipids. Sphingolipids are essential components of the lipid raft domains of the plasma membrane and this structural function is important for apoptosis and/or cell proliferation. Dysregulation of sphingolipids, including ceramide, sphingomyelin or sphingosine 1-phosphate, has been linked to drug resistance in different types of cancer, including breast, melanoma or colon cancer. Sphingolipid metabolism is complex, involving several lipid catabolism with the participation of key enzymes such as glucosylceramide synthase (GCS) and sphingosine kinase 1 (SPHK1). With an overview of the latest available data on this topic and its implications in cancer therapy, this review focuses on the main enzymes implicated in sphingolipids metabolism and their intermediate metabolites involved in cancer drug resistance.
Collapse
Affiliation(s)
- Marina Bataller
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain
| | - Almudena Sánchez-García
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain
| | - Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain
| | - Isabel Rodriguez
- Assistant Director of Nursing, Nursing Management Service Hospital Vall d'Hebron, Barcelona, Spain
| | - Matilde Esther LLeonart
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Madrid, Spain
| |
Collapse
|
3
|
Gil-Hernández A, Arroyo-Campuzano M, Simoni-Nieves A, Zazueta C, Gomez-Quiroz LE, Silva-Palacios A. Relevance of Membrane Contact Sites in Cancer Progression. Front Cell Dev Biol 2021; 8:622215. [PMID: 33511135 PMCID: PMC7835521 DOI: 10.3389/fcell.2020.622215] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/10/2020] [Indexed: 01/01/2023] Open
Abstract
Membrane contact sites (MCS) are typically defined as areas of proximity between heterologous or homologous membranes characterized by specific proteins. The study of MCS is considered as an emergent field that shows how crucial organelle interactions are in cell physiology. MCS regulate a myriad of physiological processes such as apoptosis, calcium, and lipid signaling, just to name a few. The membranal interactions between the endoplasmic reticulum (ER)–mitochondria, the ER–plasma membrane, and the vesicular traffic have received special attention in recent years, particularly in cancer research, in which it has been proposed that MCS regulate tumor metabolism and fate, contributing to their progression. However, as the therapeutic or diagnostic potential of MCS has not been fully revisited, in this review, we provide recent information on MCS relevance on calcium and lipid signaling in cancer cells and on its role in tumor progression. We also describe some proteins associated with MCS, like CERT, STIM1, VDAC, and Orai, that impact on cancer progression and that could be a possible diagnostic marker. Overall, these information might contribute to the understanding of the complex biology of cancer cells.
Collapse
Affiliation(s)
- Aurora Gil-Hernández
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Miguel Arroyo-Campuzano
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Arturo Simoni-Nieves
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Luis Enrique Gomez-Quiroz
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
4
|
Vethakanraj HS, Chandrasekaran N, Sekar AK. Acid ceramidase, a double-edged sword in cancer aggression: A minireview. Curr Cancer Drug Targets 2020; 21:CCDT-EPUB-112652. [PMID: 33357194 DOI: 10.2174/1568009620666201223154621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/18/2020] [Accepted: 10/30/2020] [Indexed: 11/22/2022]
Abstract
Acid ceramidase (AC), the key enzyme of the ceramide metabolic pathway hydrolyzes pro-apoptotic ceramide to sphingosine, which by the action of sphingosine-1-kinase is metabolized to mitogenic sphingosine-1-phosphate. The intracellular level of AC determines ceramide/sphingosine-1-phosphate rheostat which in turn decides the cell fate. The upregulated AC expression during cancerous condition acts as a "double-edged sword" by converting pro-apoptotic ceramide to anti-apoptotic sphingosine-1-phosphate, wherein on one end, the level of ceramide is decreased and on the other end, the level of sphingosine-1-phosphate is increased, thus altogether aggravating the cancer progression. In addition, cancer cells with upregulated AC expression exhibited increased cell proliferation, metastasis, chemoresistance, radioresistance and numerous strategies were developed in the past to effectively target the enzyme. Gene silencing and pharmacological inhibition of AC sensitized the resistant cells to chemo/radiotherapy thereby promoting cell death. The core objective of this review is to explore AC mediated tumour progression and the potential role of AC inhibitors in various cancer cell lines/models.
Collapse
|
5
|
White-Gilbertson S, Voelkel-Johnson C. Giants and monsters: Unexpected characters in the story of cancer recurrence. Adv Cancer Res 2020; 148:201-232. [PMID: 32723564 DOI: 10.1016/bs.acr.2020.03.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Polyploid giant cancer cells (PGCC) constitute a dangerous subpopulation of cancer cells and are a driving force in cancer recurrence. These unique cells arise from diploid tumor cells in response to stress encountered in the tumor microenvironment or during cancer therapy. PGCC are greatly dedifferentiated, acquire pluripotency, and are able to replicate through a form of asymmetric division called neosis, which results in new populations that are themselves able to differentiate into new cell types or to re-establish tumors. Progeny tend to be more genetically unstable than the founding population due to the dysregulation required to transition through a PGCC state. Therefore, cancers that escape stressors through this mechanism tend to re-emerge with a more aggressive phenotype that is therapy resistant. This review focuses on the clinical significance of PGCC, the need for standardized nomenclature and molecular markers, as well as possible avenues to develop therapies aimed at PGCC and the process of neosis. The biology underlying the development of PGCC including cell cycle checkpoint dysregulation, stress responses, dedifferentiation, stemness and epithelial-mesenchymal transition is discussed.
Collapse
Affiliation(s)
- Shai White-Gilbertson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
6
|
Voelkel-Johnson C, Norris JS, White-Gilbertson S. Interdiction of Sphingolipid Metabolism Revisited: Focus on Prostate Cancer. Adv Cancer Res 2018; 140:265-293. [PMID: 30060812 PMCID: PMC6460930 DOI: 10.1016/bs.acr.2018.04.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sphingolipid metabolism is known to play a role in cell death, survival, and therapy resistance in cancer. Sphingolipids, particularly dihydroceramide and ceramide, are associated with antiproliferative or cell death responses, respectively, and are central to effective cancer therapy. Within the last decade, strides have been made in elucidating many intricacies of sphingolipid metabolism. New information has emerged on the mechanisms by which sphingolipid metabolism is dysregulated during malignancy and how cancer cells survive and/or escape therapeutic interventions. This chapter focuses on three main themes: (1) sphingolipid enzymes that are dysregulated in cancer, particularly in prostate cancer; (2) inhibitors of sphingolipid metabolism that antagonize prosurvival responses; and (3) sphingolipid-driven escape mechanisms that allow cancer cells to evade therapies. We explore clinical and preclinical approaches to interdict sphingolipid metabolism and provide a rationale for combining strategies to drive the generation of antiproliferative ceramides with prevention of ceramide clearance.
Collapse
Affiliation(s)
- Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - James S. Norris
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Shai White-Gilbertson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
7
|
Molecular Targeting of Acid Ceramidase in Glioblastoma: A Review of Its Role, Potential Treatment, and Challenges. Pharmaceutics 2018; 10:pharmaceutics10020045. [PMID: 29642535 PMCID: PMC6027516 DOI: 10.3390/pharmaceutics10020045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma is the most common, malignant primary tumor of the central nervous system. The average prognosis for life expectancy after diagnosis, with the triad of surgery, chemotherapy, and radiation therapy, is less than 1.5 years. Chemotherapy treatment is mostly limited to temozolomide. In this paper, the authors review an emerging, novel drug called acid ceramidase, which targets glioblastoma. Its role in cancer treatment in general, and more specifically, in the treatment of glioblastoma, are discussed. In addition, the authors provide insights on acid ceramidase as a potential druggable target for glioblastoma.
Collapse
|
8
|
Vujadinovic M, Wunderlich K, Callendret B, Koning M, Vermeulen M, Sanders B, van der Helm E, Gecgel A, Spek D, de Boer K, Stalknecht M, Serroyen J, Grazia Pau M, Schuitemaker H, Zahn R, Custers J, Vellinga J. Adenoviral Type 35 and 26 Vectors with a Bidirectional Expression Cassette in the E1 Region Show an Improved Genetic Stability Profile and Potent Transgene-Specific Immune Response. Hum Gene Ther 2017; 29:337-351. [PMID: 28816084 DOI: 10.1089/hum.2017.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genetic vaccines based on replication-incompetent adenoviral (AdV) vectors are currently in clinical development. Monovalent AdV vectors express one antigen from an expression cassette placed in most cases in the E1 region. For many vaccines, inclusion of several antigens is necessary in order to raise protective immunity and/or target more than one pathogen or pathogen strain. On the basis of the current technology, a mix of several monovalent vectors can be employed. However, a mix of the standard monovalent AdV vectors may not be optimal with respect to manufacturing costs and the final dose per vector in humans. Alternatively, a variety of bivalent recombinant AdV vector approaches is described in the literature. It remains unclear whether all strategies are equally suitable for clinical development while preserving all the beneficial properties of the monovalent AdV (e.g., immunogenic potency). Therefore, a thorough assessment of different bivalent AdV strategies was performed in a head-to-head fashion compared with the monovalent benchmark. The vectors were tested for rescue efficiency, genetic stability, transgene expression, and potency to induce transgene-specific immune responses. We report that the vector expressing multiple antigens from a bidirectional expression cassette in E1 shows a better genetic stability profile and a potent transgene-specific immune response compared with the other tested bivalent vectors.
Collapse
Affiliation(s)
- Marija Vujadinovic
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Kerstin Wunderlich
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Benoit Callendret
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Marina Koning
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Mark Vermeulen
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Barbara Sanders
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Esmeralda van der Helm
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Adile Gecgel
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Dirk Spek
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Karin de Boer
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Masha Stalknecht
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jan Serroyen
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Maria Grazia Pau
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Hanneke Schuitemaker
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Roland Zahn
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jerome Custers
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jort Vellinga
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| |
Collapse
|
9
|
Roh JL, Park JY, Kim EH, Jang HJ. Targeting acid ceramidase sensitises head and neck cancer to cisplatin. Eur J Cancer 2015; 52:163-72. [PMID: 26687835 DOI: 10.1016/j.ejca.2015.10.056] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 10/20/2015] [Accepted: 10/23/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Acid ceramidase (AC), a key enzyme in ceramide metabolism, plays a role in cancer progression and resistance to therapy. However, the role of AC in head and neck cancer (HNC) has not been addressed. Here, we investigate the effect of AC inhibition on the response to cisplatin-based chemotherapy for HNC. METHODS AC protein and messenger RNA (mRNA) expression were examined in primary tumours and paired normal tissues, and in HNC cell lines. The effects of genetic and pharmacological AC inhibition using small hairpin RNA (shRNA) and N-oleoyl-ethanolamine (NOE), alone and in combination with cisplatin, were assessed in human HNC cells by measuring cell viability, cell cycle progression, apoptosis, mRNA, and protein expression, and in preclinical tumour xenograft mouse models. FINDINGS AC overexpression was observed in four of six primary tumour tissues and six of nine HNC cell lines. Cisplatin sensitivity was significantly decreased by AC overexpression and significantly increased by AC downregulation in HNC cells (P<0.01). NOE or AC shRNA-mediated AC inhibition enhanced cisplatin-induced HNC cell death by increasing ceramide production and activating pro-apoptotic proteins, and these effects were abrogated by PUMA small interfering RNA transfection. AC inhibition promoted cisplatin-induced apoptosis of HNC cells in vitro and in vivo. INTERPRETATIONS AC overexpression is associated with cisplatin sensitivity, suggesting its potential role as a chemotherapeutic target for HNC. Genetic or pharmacological AC inhibition promotes cisplatin cytotoxicity in HNC cells.
Collapse
Affiliation(s)
- Jong-Lyel Roh
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Jin Young Park
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Hye Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hye Jin Jang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Bach A, Pizzirani D, Realini N, Vozella V, Russo D, Penna I, Melzig L, Scarpelli R, Piomelli D. Benzoxazolone Carboxamides as Potent Acid Ceramidase Inhibitors: Synthesis and Structure-Activity Relationship (SAR) Studies. J Med Chem 2015; 58:9258-72. [PMID: 26560855 DOI: 10.1021/acs.jmedchem.5b01188] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ceramides are lipid-derived intracellular messengers involved in the control of senescence, inflammation, and apoptosis. The cysteine amidase, acid ceramidase (AC), hydrolyzes these substances into sphingosine and fatty acid and, by doing so, regulates their signaling activity. AC inhibitors may be useful in the treatment of pathological conditions, such as cancer, in which ceramide levels are abnormally reduced. Here, we present a systematic SAR investigation of the benzoxazolone carboxamides, a recently described class of AC inhibitors that display high potency and systemic activity in mice. We examined a diverse series of substitutions on both benzoxazolone ring and carboxamide side chain. Several modifications enhanced potency and stability, and one key compound with a balanced activity-stability profile (14) was found to inhibit AC activity in mouse lungs and cerebral cortex after systemic administration. The results expand our arsenal of AC inhibitors, thereby facilitating the use of these compounds as pharmacological tools and their potential development as drug leads.
Collapse
Affiliation(s)
- Anders Bach
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Daniela Pizzirani
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Natalia Realini
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Valentina Vozella
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Debora Russo
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Ilaria Penna
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Laurin Melzig
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Rita Scarpelli
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Daniele Piomelli
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy.,Departments of Anatomy and Neurobiology, Biological Chemistry, and Pharmacology, University of California , Irvine, California 92697-4625, United States
| |
Collapse
|
11
|
Modiano JF, Lindborg BA, McElmurry RT, Lewellen M, Forster CL, Zamora EA, Schaack J, Bellgrau D, O'Brien TD, Tolar J. Mesenchymal stromal cells inhibit murine syngeneic anti-tumor immune responses by attenuating inflammation and reorganizing the tumor microenvironment. Cancer Immunol Immunother 2015; 64:1449-60. [PMID: 26250807 DOI: 10.1007/s00262-015-1749-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 07/30/2015] [Indexed: 12/29/2022]
Abstract
The potential of mesenchymal stromal cells (MSCs) to inhibit anti-tumor immunity is becoming increasingly well recognized, but the precise steps affected by these cells during the development of an anti-tumor immune response remain incompletely understood. Here, we examined how MSCs affect the steps required to mount an effective anti-tumor immune response following administration of adenovirus Fas ligand (Ad-FasL) in the Lewis lung carcinoma (LL3) model. Administration of bone marrow-derived MSCs with LL3 cells accelerated tumor growth significantly. MSCs inhibited the inflammation induced by Ad-FasL in the primary tumors, precluding their rejection; MSCs also reduced the consequent expansion of tumor-specific T cells in the treated hosts. When immune T cells were transferred to adoptive recipients, MSCs impaired, but did not completely abrogate the ability of these T cells to promote elimination of secondary tumors. This impairment was associated with a modest reduction in tumor-infiltrating T cells, with a significant reduction in tumor-infiltrating macrophages, and with a reorganization of the stromal environment. Our data indicate that MSCs in the tumor environment reduce the efficacy of immunotherapy by creating a functional and anatomic barrier that impairs inflammation, T cell priming and expansion, and T cell function-including recruitment of effector cells.
Collapse
Affiliation(s)
- Jaime F Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Masonic Cancer Center, University of Minnesota, 1365 Gortner Avenue, St. Paul, MN, 55108, USA. .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA. .,Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA. .,Center for Immunology, University of Minnesota, Minneapolis, MN, USA.
| | - Beth A Lindborg
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.,Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA.,BRTI Life Sciences, Two Harbors, MN, USA
| | - Ron T McElmurry
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Mitzi Lewellen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Masonic Cancer Center, University of Minnesota, 1365 Gortner Avenue, St. Paul, MN, 55108, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Colleen L Forster
- BioNet Histology Research Laboratory, Academic Health Center, University of Minnesota, Minneapolis, MN, USA
| | - Edward A Zamora
- Microbiology, Immunology, and Cancer Biology Graduate Group, University of Minnesota, Minneapolis, MN, USA
| | - Jerome Schaack
- Department of Microbiology, School of Medicine, University of Colorado, Aurora, CO, USA.,University of Colorado Cancer Center, Aurora, CO, USA
| | - Donald Bellgrau
- University of Colorado Cancer Center, Aurora, CO, USA.,Integrated Department of Immunology, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Timothy D O'Brien
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.,Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA
| | - Jakub Tolar
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.,Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
12
|
Abstract
The topic of ceramidases has experienced an enormous boost during the last few years. Ceramidases catalyze the degradation of ceramide to sphingosine and fatty acids. Ceramide is not only the central hub of sphingolipid biosynthesis and degradation, it is also a key molecule in sphingolipid signaling, promoting differentiation or apoptosis. Acid ceramidase inhibition sensitizes certain types of cancer to chemo- and radio-therapy and this is suggestive of a role of acid ceramidase inhibitors as chemo-sensitizers which can act synergistically with chemo-therapeutic drugs. In this review, we summarize the development of ceramide analogues as first-generation ceramidase inhibitors together with data on their activity in cells and disease models. Furthermore, we describe the recent developments that have led to highly potent second-generation ceramidase inhibitors that act at nanomolar concentrations. In the third part, various assays of ceramidases are described and their relevance for accurately measuring ceramidase activities and for the development of novel inhibitors is highlighted. Besides potential clinical implications, the recent improvements in ceramidase inhibition and assaying may help to better understand the mechanisms of ceramide biology.
Collapse
Affiliation(s)
- Essa M Saied
- Humboldt Universität zu Berlin, Institute for Chemistry, Berlin, Germany; Suez Canal University, Chemistry Department, Faculty of Science, Ismailia, Egypt
| | - Christoph Arenz
- Humboldt Universität zu Berlin, Institute for Chemistry, Berlin, Germany.
| |
Collapse
|
13
|
Kus G, Kabadere S, Uyar R, Kutlu HM. Induction of apoptosis in prostate cancer cells by the novel ceramidase inhibitor ceranib-2. In Vitro Cell Dev Biol Anim 2015; 51:1056-63. [PMID: 26170224 DOI: 10.1007/s11626-015-9932-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 06/12/2015] [Indexed: 12/22/2022]
Abstract
Ceramidases are key enzymes that decrease ceramide levels in cells. A reduction in ceramide concentration impairs ceramide signalling, and results in apoptosis resistance in cancer cells. This study investigates the potential for ceranib-2, a novel ceramidase inhibitor, to affect the survival and/or promote apoptosis of prostate cancer cells (LNCaP and DU145) in vitro. Cell viability was determined using MTT, and apoptosis assessed via flow cytometry. We examined structural changes with both confocal and transmission electron microscopes. Ceranib-2 concentrations of 0.1, 1, 5, 10, 25 and 50 μM were applied to LNCaP and DU145 cell lines. The corresponding reduction in LNCaP cell viability (against the control) was 84%, 80%, 64%, 56%, 40% and 15% after 24 h, and 81%, 74%, 60%, 55%, 27% and 11% after 48 h. For DU145 cells, viability was reduced to 84%, 82%, 63%, 50%, 41% and 18% after 24 h, and 64%, 42%, 30%, 20%, 8% and 5% after 48 h. Following treatment with 25 and 50 μM ceranib-2, the respective observed rates of early apoptosis in LNCaP cells were 23% and 36% after 24 h and 27% and 58% after 48 h. The morphological and ultrastructural signs of apoptosis detected were fragmented nuclei, chromatin condensations and cytoskeleton laceration. The inhibitory effects of ceranib-2 on prostate cancer cell survival are dose and time dependent. For LNCaP cells, ceranib-2 toxicity was predominately apoptotic in nature, while for DU145 cells, cell death may be related to non-apoptotic mechanisms.
Collapse
Affiliation(s)
- Gokhan Kus
- Department of Health Programme, Open Faculty, Anadolu University, 26480, Eskisehir, Turkey.
| | - Selda Kabadere
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Ruhi Uyar
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Hatice Mehtap Kutlu
- Department of Biology, Faculty of Science, Anadolu University, Eskisehir, Turkey
| |
Collapse
|
14
|
Mizutani N, Inoue M, Omori Y, Ito H, Tamiya-Koizumi K, Takagi A, Kojima T, Nakamura M, Iwaki S, Nakatochi M, Suzuki M, Nozawa Y, Murate T. Increased acid ceramidase expression depends on upregulation of androgen-dependent deubiquitinases, USP2, in a human prostate cancer cell line, LNCaP. J Biochem 2015; 158:309-19. [PMID: 25888580 DOI: 10.1093/jb/mvv039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/25/2015] [Indexed: 01/03/2023] Open
Abstract
Acid ceramidase (ACDase) metabolizes ceramide to sphingosine, leading to sphingosine 1-phosphate production. Reportedly, ACDase has been upregulated in prostate cancer. However, its regulatory mechanism remains unclear. LNCaP (androgen-sensitive prostate cancer cell line) but not PC3 and DU-145, (androgen-unresponsive cell lines) exhibited the highest ACDase protein. Among three cell lines, ASAH1 mRNA level was not correlated with ACDase protein expression, and the 5'-promoter activity did not show androgen dependency, suggesting the post-transcriptional regulation of ACDase in LNCaP cells. Based on these results, LNCaP was analysed further. Casodex, androgen receptor antagonist, and charcoal-stripped FCS (CS-FCS) decreased ACDase protein and activity, whereas dihydrotestosterone in CS-FCS culture increased ACDase protein and enzyme activity. MG132, a proteasome inhibitor, prevented the decrease of ACDase protein when cultured in CS-FCS, suggesting the involvement of ubiquitin/proteasome system. Reportedly, USP2, a deubiquitinase, plays an important role in LNCaP cells. USP2 siRNA decreased ACDase protein, whereas USP2 overexpression increased ACDase protein of LNCaP cells. However, SKP2, an ubiquitin E3 ligase known to be active in prostate cancer, did not affect androgen-dependent ACDase expression in LNCaP cells. Thus, ACDase regulation by androgen in androgen-sensitive LNCaP cells is mainly due to its prolonged protein half-life by androgen-stimulated USP2 expression.
Collapse
Affiliation(s)
- Naoki Mizutani
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Minami Inoue
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Yukari Omori
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Hiromi Ito
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Keiko Tamiya-Koizumi
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Akira Takagi
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Tetsuhito Kojima
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Mitsuhiro Nakamura
- Department of Drug Information, Gifu Pharmaceutical University, Gifu 501-1196
| | - Soichiro Iwaki
- Department of Molecular and Cellular Pathophysiology and Therapeutics, Graduate School of Pharmaceutical Science, Nagoya City University, Nagoya 467-8603
| | - Masahiro Nakatochi
- Bioinformatics Section, Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya 466-8550
| | - Motoshi Suzuki
- Division of Molecular Carcinogenesis, Nagoya University Graduate School of Medicine, Nagoya 466-8560; and
| | - Yoshinori Nozawa
- Department of Food and Health Science, Tokai Gakuin University, Kakamigahara 504-8511, Japan
| | - Takashi Murate
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673;
| |
Collapse
|
15
|
Sänger N, Ruckhäberle E, Györffy B, Engels K, Heinrich T, Fehm T, Graf A, Holtrich U, Becker S, Karn T. Acid ceramidase is associated with an improved prognosis in both DCIS and invasive breast cancer. Mol Oncol 2015; 9:58-67. [PMID: 25131496 PMCID: PMC5528695 DOI: 10.1016/j.molonc.2014.07.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/02/2014] [Accepted: 07/22/2014] [Indexed: 11/26/2022] Open
Abstract
Acid ceramidase (ASAH1) a key enzyme of sphingolipid metabolism converting pro-apoptotic ceramide to sphingosine has been shown to be overexpressed in various cancers. We previously demonstrated higher expression of ASAH1 in ER positive compared to ER negative breast cancer. In the current study we performed subtype specific analyses of ASAH1 gene expression in invasive and non invasive breast cancer. We show that expression of ASAH1 is mainly associated with luminal A - like cancers which are known to have the best prognosis of all breast cancer subtypes. Moreover tumors with high ASAH1 expression among the other subtypes are also characterized by an improved prognosis. The good prognosis of tumors with high ASAH1 is independent of the type of adjuvant treatment in breast cancer and is also detected in non small cell lung cancer patients. Moreover, even in pre-invasive DCIS of the breast ASAH1 is associated with a luminal phenotype and a reduced frequency of recurrences. Thus, high ASAH1 expression is generally associated with an improved prognosis in invasive breast cancer independent of adjuvant treatment and could also be valuable as prognostic factor for pre-invasive DCIS.
Collapse
Affiliation(s)
- Nicole Sänger
- Department of Obstetrics and Gynecology, University Hospital Frankfurt, Germany
| | - Eugen Ruckhäberle
- Department of Obstetrics and Gynecology, Heinrich-Heine-University Duesseldorf, Germany
| | - Balazs Györffy
- MTA TTK Lendület Cancer Biomarker Research Group, Budapest, Hungary; 2(nd) Dept. of Pediatrics, Semmelweis University, Budapest, Hungary; MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Knut Engels
- Zentrum für Pathologie, Zytologie und Molekularpathologie Neuss, Germany
| | - Tomas Heinrich
- Department of Obstetrics and Gynecology, University Hospital Frankfurt, Germany
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, Heinrich-Heine-University Duesseldorf, Germany
| | - Anna Graf
- Department of Obstetrics and Gynecology, University Hospital Frankfurt, Germany
| | - Uwe Holtrich
- Department of Obstetrics and Gynecology, University Hospital Frankfurt, Germany
| | - Sven Becker
- Department of Obstetrics and Gynecology, University Hospital Frankfurt, Germany
| | - Thomas Karn
- Department of Obstetrics and Gynecology, University Hospital Frankfurt, Germany.
| |
Collapse
|
16
|
Carroll B, Donaldson JC, Obeid L. Sphingolipids in the DNA damage response. Adv Biol Regul 2014; 58:38-52. [PMID: 25434743 DOI: 10.1016/j.jbior.2014.11.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 12/16/2022]
Abstract
Recently, sphingolipid metabolizing enzymes have emerged as important targets of many chemotherapeutics and DNA damaging agents and therefore play significant roles in mediating the physiological response of the cell to DNA damage. In this review we will highlight points of connection between the DNA damage response (DDR) and sphingolipid metabolism; specifically how certain sphingolipid enzymes are regulated in response to DNA damage and how the bioactive lipids produced by these enzymes affect cell fate.
Collapse
Affiliation(s)
- Brittany Carroll
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jane Catalina Donaldson
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Lina Obeid
- Northport VA Medical Center, Northport, NY 11768, USA; Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
17
|
Don AS, Lim XY, Couttas TA. Re-configuration of sphingolipid metabolism by oncogenic transformation. Biomolecules 2014; 4:315-53. [PMID: 24970218 PMCID: PMC4030989 DOI: 10.3390/biom4010315] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 02/11/2014] [Accepted: 02/27/2014] [Indexed: 12/15/2022] Open
Abstract
The sphingolipids are one of the major lipid families in eukaryotes, incorporating a diverse array of structural variants that exert a powerful influence over cell fate and physiology. Increased expression of sphingosine kinase 1 (SPHK1), which catalyses the synthesis of the pro-survival, pro-angiogenic metabolite sphingosine 1-phosphate (S1P), is well established as a hallmark of multiple cancers. Metabolic alterations that reduce levels of the pro-apoptotic lipid ceramide, particularly its glucosylation by glucosylceramide synthase (GCS), have frequently been associated with cancer drug resistance. However, the simple notion that the balance between ceramide and S1P, often referred to as the sphingolipid rheostat, dictates cell survival contrasts with recent studies showing that highly potent and selective SPHK1 inhibitors do not affect cancer cell proliferation or survival, and studies demonstrating higher ceramide levels in some metastatic cancers. Recent reports have implicated other sphingolipid metabolic enzymes such as acid sphingomyelinase (ASM) more strongly in cancer pathogenesis, and highlight lysosomal sphingolipid metabolism as a possible weak point for therapeutic targeting in cancer. This review describes the evidence implicating different sphingolipid metabolic enzymes and their products in cancer pathogenesis, and suggests how newer systems-level approaches may improve our overall understanding of how oncogenic transformation reconfigures sphingolipid metabolism.
Collapse
Affiliation(s)
- Anthony S Don
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Xin Y Lim
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Timothy A Couttas
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
18
|
Truman JP, García-Barros M, Obeid LM, Hannun YA. Evolving concepts in cancer therapy through targeting sphingolipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:1174-88. [PMID: 24384461 DOI: 10.1016/j.bbalip.2013.12.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/18/2013] [Accepted: 12/21/2013] [Indexed: 12/29/2022]
Abstract
Traditional methods of cancer treatment are limited in their efficacy due to both inherent and acquired factors. Many different studies have shown that the generation of ceramide in response to cytotoxic therapy is generally an important step leading to cell death. Cancer cells employ different methods to both limit ceramide generation and to remove ceramide in order to become resistant to treatment. Furthermore, sphingosine kinase activity, which phosphorylates sphingosine the product of ceramide hydrolysis, has been linked to multidrug resistance, and can act as a strong survival factor. This review will examine several of the most frequently used cancer therapies and their effect on both ceramide generation and the mechanisms employed to remove it. The development and use of inhibitors of sphingosine kinase will be focused upon as an example of how targeting sphingolipid metabolism may provide an effective means to improve treatment response rates and reduce associated treatment toxicity. This article is part of a Special Issue entitled Tools to study lipid functions.
Collapse
Affiliation(s)
- Jean-Philip Truman
- Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794 Stony Brook, NY, USA.
| | - Mónica García-Barros
- Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794 Stony Brook, NY, USA.
| | - Lina M Obeid
- Northport Veterans Affairs Medical Center, Northport, NY 11768, USA; Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 178, 11794 Stony Brook, NY, USA.
| | - Yusuf A Hannun
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 178, 11794 Stony Brook, NY, USA.
| |
Collapse
|
19
|
Beckham TH, Cheng JC, Lu P, Marrison ST, Norris JS, Liu X. Acid ceramidase promotes nuclear export of PTEN through sphingosine 1-phosphate mediated Akt signaling. PLoS One 2013; 8:e76593. [PMID: 24098536 PMCID: PMC3788144 DOI: 10.1371/journal.pone.0076593] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 09/02/2013] [Indexed: 11/19/2022] Open
Abstract
The tumor suppressor PTEN is now understood to regulate cellular processes at the cytoplasmic membrane, where it classically regulates PI3K signaling, as well as in the nucleus where multiple roles in controlling cell cycle and genome stability have been elucidated. Mechanisms that dictate nuclear import and, less extensively, nuclear export of PTEN have been described, however the relevance of these processes in disease states, particularly cancer, remain largely unknown. We investigated the impact of acid ceramidase on the nuclear-cytoplasmic trafficking of PTEN. Immunohistochemical analysis of a human prostate tissue microarray revealed that nuclear PTEN was lost in patients whose tumors had elevated acid ceramidase. We found that acid ceramidase promotes a reduction in nuclear PTEN that is dependent upon sphingosine 1-phosphate-mediated activation of Akt. We were further able to show that sphingosine 1-phosphate promotes formation of a complex between Crm1 and PTEN, and that leptomycin B prevents acid ceramidase and sphingosine 1-phosphate mediated loss of nuclear PTEN, suggesting an active exportin-mediated event. To investigate whether the tumor promoting aspects of acid ceramidase in prostate cancer depend upon its ability to export PTEN from the nucleus, we used enforced nuclear expression of PTEN to study docetaxel-induced apoptosis and cell killing, proliferation, and xenoengraftment. Interestingly, while acid ceramidase was able to protect cells expressing wild type PTEN from docetaxel, promote proliferation and xenoengraftment, acid ceramidase had no impact in cells expressing PTEN-NLS. These findings suggest that acid ceramidase, through sphingosine 1-phosphate, promotes nuclear export of PTEN as a means of promoting tumor formation, cell proliferation, and resistance to therapy.
Collapse
Affiliation(s)
- Thomas H. Beckham
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Joseph C. Cheng
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Ping Lu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - S. Tucker Marrison
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - James S. Norris
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Xiang Liu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|
20
|
Acid ceramidase (AC)--a key enzyme of sphingolipid metabolism--correlates with better prognosis in epithelial ovarian cancer. Int J Gynecol Pathol 2013; 32:249-57. [PMID: 23518908 DOI: 10.1097/pgp.0b013e3182673982] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acid ceramidase (AC), a key enzyme of sphingolipid metabolism, seems to play an important role in cancer progression. The objective of this study was to explore the expression of AC in ovarian cancer and its impact on prognosis. Expression analysis of AC in n=112 ovarian cancer patients was performed by immunohistochemical analysis of primary paraffin-embedded tumor samples. The results were scored on the basis of the staining intensity and percentage of positive tumor cells, resulting in an immunoreactive score from 0 to 12. These results were correlated to clinical and pathologic characteristics and survival. AC expression correlated significantly only with FIGO stage (0.047). In serous carcinoma, low level of AC was independently associated with reduced progression-free survival and overall survival of 12.0 mo [95% confidence interval (CI), 5.78-18.23] versus 18.1 mo (95% CI, 11.61-24.59; P=0.008) and 35.7 mo (95% CI, 22.24-47.16) versus 58.7 mo (95% CI, 36.48-80.91; P=0.032), respectively. In multivariate analysis, AC presents as an independent prognostic factor for progression-free survival (hazard ratio 1.88; 95% CI, 1.13-3.11; P=0.015). AC is a prognostic factor in epithelial ovarian cancer. Low AC expression can be associated with tumor progression in carcinoma of the ovaries. These results are in contrast to the concept of AC as a promoter for cancer progression. Nevertheless, they are supported by the lately discovered tumor-suppressing function of sphingosine, the enzymatic product of AC.
Collapse
|
21
|
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
|
22
|
The impact of sphingosine kinase-1 in head and neck cancer. Biomolecules 2013; 3:481-513. [PMID: 24970177 PMCID: PMC4030949 DOI: 10.3390/biom3030481] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/02/2013] [Accepted: 08/03/2013] [Indexed: 12/15/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has a high reoccurrence rate and an extremely low survival rate. There is limited availability of effective therapies to reduce the rate of recurrence, resulting in high morbidity and mortality of advanced cases. Late presentation, delay in detection of lesions, and a high rate of metastasis make HNSCC a devastating disease. This review offers insight into the role of sphingosine kinase-1 (SphK1), a key enzyme in sphingolipid metabolism, in HNSCC. Sphingolipids not only play a structural role in cellular membranes, but also modulate cell signal transduction pathways to influence biological outcomes such as senescence, differentiation, apoptosis, migration, proliferation, and angiogenesis. SphK1 is a critical regulator of the delicate balance between proliferation and apoptosis. The highest expression of SphK1 is found in the advanced stage of disease, and there is a positive correlation between SphK1 expression and recurrent tumors. On the other hand, silencing SphK1 reduces HNSCC tumor growth and sensitizes tumors to radiation-induced death. Thus, SphK1 plays an important and influential role in determining HNSCC proliferation and metastasis. We discuss roles of SphK1 and other sphingolipids in HNSCC development and therapeutic strategies against HNSCC.
Collapse
|
23
|
Abstract
Non-surgical therapies for human malignancies must negotiate complex cell signaling pathways to impede cancer cell growth, ideally promoting death of cancer cells while sparing healthy tissue. For most of the past half century, medical approaches for treating cancer have relied primarily on cytotoxic chemotherapeutics that interfere with DNA replication and cell division, susceptibilities of rapidly dividing cancer cells. As a consequence, these therapies exert considerable cell stress, promoting the generation of ceramide through de novo synthesis and recycling of complex glycosphingolipids and sphingomyelin into apoptotic ceramide. Radiotherapy of cancer exerts similar geno- and cytotoxic cell stresses, and generation of ceramide following ionizing radiation therapy is a well-described feature of radiation-induced cell death. Emerging evidence now describes sphingolipids as mediators of death in response to newer targeted therapies, cementing ceramide generation as a common mechanism of cell death in response to cancer therapy. Many studies have now shown that dysregulation of ceramide accumulation-whether by reduced generation or accelerated metabolism-is a common mechanism of resistance to standard cancer therapies. The aims of this chapter will be to discuss described mechanisms of cancer resistance to therapy related to dysregulation of sphingolipid metabolism and to explore clinical and preclinical approaches to interdict sphingolipid metabolism to improve outcomes of standard cancer therapies.
Collapse
|
24
|
Acid ceramidase induces sphingosine kinase 1/S1P receptor 2-mediated activation of oncogenic Akt signaling. Oncogenesis 2013; 2:e49. [PMID: 23732709 PMCID: PMC3740300 DOI: 10.1038/oncsis.2013.14] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Acid ceramidase (AC) is overexpressed in most prostate tumors and confers oncogenic phenotypes to prostate cancer cells. AC modulates the cellular balance between ceramide, sphingosine and sphingosine 1-phosphate (S1P). These bioactive sphingolipids have diverse, powerful and often oppositional impacts on cell signaling, including the activation status of the oncogenic kinase Akt. Our studies show that AC expression correlates with phosphorylation of Akt in human prostate tumors, and elevation of phosphorylated Akt in tumor versus patient-matched benign tissue is contingent upon AC elevation. Investigation of the mechanism for AC-induced Akt activation revealed that AC activates Akt through sphingosine kinase 1 (SphK1)-derived generation of S1P. This signaling pathway proceeds through S1P receptor 2 (S1PR2)-dependent stimulation of PI3K. Functionally, AC-overexpressing cells are insensitive to cytotoxic chemotherapy, however, these cells are more susceptible to targeted inhibition of Akt. AC-overexpressing cells proliferate more rapidly than control cells and form more colonies in soft agar; however, these effects are profoundly sensitive to Akt inhibition, demonstrating increased dependence on Akt signaling for the oncogenic phenotypes of AC-overexpressing cells. These observations may have clinical implications for targeted therapy as PI3K and Akt inhibitors emerge from clinical trials.
Collapse
|
25
|
Pizzirani D, Pagliuca C, Realini N, Branduardi D, Bottegoni G, Mor M, Bertozzi F, Scarpelli R, Piomelli D, Bandiera T. Discovery of a New Class of Highly Potent Inhibitors of Acid Ceramidase: Synthesis and Structure–Activity Relationship (SAR). J Med Chem 2013; 56:3518-30. [DOI: 10.1021/jm301879g] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Daniela Pizzirani
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego
30, I-16163 Genova, Italy
| | - Chiara Pagliuca
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego
30, I-16163 Genova, Italy
| | - Natalia Realini
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego
30, I-16163 Genova, Italy
| | - Davide Branduardi
- Theoretical Molecular Biophysics
Group, Max Planck Institute for Biophysics, Max-von-Laue Strasse 3, 60438, Frankfurt am Main, Germany
| | - Giovanni Bottegoni
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego
30, I-16163 Genova, Italy
| | - Marco Mor
- Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle
Scienze 27/A, I-43124 Parma, Italy
| | - Fabio Bertozzi
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego
30, I-16163 Genova, Italy
| | - Rita Scarpelli
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego
30, I-16163 Genova, Italy
| | - Daniele Piomelli
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego
30, I-16163 Genova, Italy
- Department of Anatomy and Neurobiology, University of California—Irvine, Irvine, California
92697-4625, United States
| | - Tiziano Bandiera
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego
30, I-16163 Genova, Italy
| |
Collapse
|
26
|
Camacho L, Meca-Cortés O, Abad JL, García S, Rubio N, Díaz A, Celià-Terrassa T, Cingolani F, Bermudo R, Fernández PL, Blanco J, Delgado A, Casas J, Fabriàs G, Thomson TM. Acid ceramidase as a therapeutic target in metastatic prostate cancer. J Lipid Res 2013; 54:1207-20. [PMID: 23423838 DOI: 10.1194/jlr.m032375] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Acid ceramidase (AC) catalyzes the hydrolysis of ceramide into sphingosine, in turn a substrate of sphingosine kinases that catalyze its conversion into the mitogenic sphingosine-1-phosphate. AC is expressed at high levels in several tumor types and has been proposed as a cancer therapeutic target. Using a model derived from PC-3 prostate cancer cells, the highly tumorigenic, metastatic, and chemoresistant clone PC-3/Mc expressed higher levels of the AC ASAH1 than the nonmetastatic clone PC-3/S. Stable knockdown of ASAH1 in PC-3/Mc cells caused an accumulation of ceramides, inhibition of clonogenic potential, increased requirement for growth factors, and inhibition of tumorigenesis and lung metastases. We developed de novo ASAH1 inhibitors, which also caused a dose-dependent accumulation of ceramides in PC-3/Mc cells and inhibited their growth and clonogenicity. Finally, immunohistochemical analysis of primary prostate cancer samples showed that higher levels of ASAH1 were associated with more advanced stages of this neoplasia. These observations confirm ASAH1 as a therapeutic target in advanced and chemoresistant forms of prostate cancer and suggest that our new potent and specific AC inhibitors could act by counteracting critical growth properties of these highly aggressive tumor cells.
Collapse
Affiliation(s)
- Luz Camacho
- Department of Biomedicinal Chemistry, Research Unit on Bioactive Molecules (RUBAM), Institute for Advanced Chemistry of Catalonia, National Research Council (IQAC-CSIC), Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
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
|
28
|
Scheffer L, Raghavendra PR, Ma J, Acharya JK. Ceramide transfer protein and cancer. Anticancer Agents Med Chem 2012; 11:904-10. [PMID: 21707482 DOI: 10.2174/187152011797655087] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 05/13/2011] [Accepted: 05/26/2011] [Indexed: 12/31/2022]
Abstract
Sphingolipids are important structural components of membranes, and play an equally important role in basic cellular processes as second messengers. Recently, sphingolipids are receiving increasing attention in cancer research. Ceramide is the central molecule that regulates sphingolipid metabolism forming the basic structural backbone of sphingolipids and the precursor of all complex sphingolipids. It is been proposed to be an important regulator of tumor cell death following exposure to stress stimuli. The increase or decrease of ceramide levels leading to change in sensitivity of cancer cells to stress stimuli provides support for a central role of ceramide signaling in cell death. In this review, we have focused on ceramide transfer protein (CERT) as a major regulator of ceramide flux in the cell.
Collapse
Affiliation(s)
- Luana Scheffer
- Laboratory of Cell and Developmental Signaling, National Cancer Institute Frederick, MD 21702, USA
| | | | | | | |
Collapse
|
29
|
Abstract
The bioactive sphingolipids including, ceramide, sphingosine, and sphingosine-1-phosphate (S1P) have important roles in several types of signaling and regulation of many cellular processes including cell proliferation, apoptosis, senescence, angiogenesis, and transformation. Recent accumulating evidence suggests that ceramide- and S1P-mediated pathways have been implicated in cancer development, progression, and chemotherapy. Ceramide mediates numerous cell-stress responses, such as induction of apoptosis and cell senescence, whereas S1P plays pivotal roles in cell survival, migration, and inflammation. These sphingolipids with opposing roles can be interconverted within cells, suggesting that the balance between them is related to cell fate. Importantly, these sphingolipids are metabolically related through actions of enzymes including ceramidases, ceramide synthases, sphingosine kinases, and S1P phosphatases thereby forming a network of metabolically interrelated bioactive lipid mediators whose importance in normal cellular function and diseases is gaining appreciation. In this review, we summarize involvement of sphingolipids and their related enzymes in pathogenesis and therapy of cancer and discuss future directions of sphingolipid field in cancer research.
Collapse
Affiliation(s)
- Hideki Furuya
- University of Hawaii Cancer Center, 651 Ilalo Street, BSB #222H, Honolulu, HI 96813, USA
| | | | | |
Collapse
|
30
|
Beckham TH, Lu P, Cheng JC, Zhao D, Turner LS, Zhang X, Hoffman S, Armeson KE, Liu A, Marrison T, Hannun YA, Liu X. Acid ceramidase-mediated production of sphingosine 1-phosphate promotes prostate cancer invasion through upregulation of cathepsin B. Int J Cancer 2012; 131:2034-43. [PMID: 22322590 DOI: 10.1002/ijc.27480] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 01/27/2012] [Indexed: 12/16/2022]
Abstract
Invasiveness is one of the key features of aggressive prostate cancer; however, our understanding of the precise mechanisms effecting invasion remains limited. The ceramide hydrolyzing enzyme acid ceramidase (AC), overexpressed in most prostate tumors, causes an aggressive and invasive phenotype through downstream effectors that have not yet been well characterized. Here, we demonstrate that AC, through generation of sphingosine-1-phosphate (S1P), promotes Ets1 nuclear expression and binding to the promoter region of matrix-degrading protease cathepsin B. Through confocal microscopy and flow cytometry, we found that AC overexpression promotes pericellular localization of cathepsin B and its translocation to the outer leaflet of the cell membrane. AC overexpressing cells have an increased abundance of cathepsin B-enriched invasive structures and enhanced ability to invade through a collagen matrix, but not in the presence of an inhibitor of cathepsin B. In human prostate tissues, AC and cathepsin B overexpression were strongly associated and may relate to poor outcome. These results demonstrate a novel pathway by which AC, through S1P, promotes an invasive phenotype in prostate cancer by causing overexpression and secretion of cathepsin B through activation and nuclear expression of Ets1. As prostate cancer prognosis is dramatically worse when invasion has occurred, this study provides critical insight into the progression toward lethal prostate cancer.
Collapse
Affiliation(s)
- Thomas H Beckham
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Villa-Morales M, Fernández-Piqueras J. Targeting the Fas/FasL signaling pathway in cancer therapy. Expert Opin Ther Targets 2012; 16:85-101. [PMID: 22239437 DOI: 10.1517/14728222.2011.628937] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The Fas/FasL system plays a significant role in tumorigenesis. Research has shown that its impairment in cancer cells may lead to apoptosis resistance and contribute to tumor progression. Thus, the development of effective therapies targeting the Fas/FasL system may play an important role in the fight against cancer. AREAS COVERED In this review the recent literature on targeting the Fas/FasL system for therapeutic exploitation at different levels is reviewed. Promising pre-clinical approaches and various exceptions are highlighted. The potential of combined therapies is also explored, whereby tumor sensitivity to Fas-mediated apoptosis is restored, before an effective targeted therapy is employed. EXPERT OPINION The success of the Fas/FasL system targeting for therapeutics will require a better understanding of the alterations conferring resistance, in order to use the most appropriate sensitizing chemotherapeutic or radiotherapeutic agents in combination with effective targeted therapies.
Collapse
Affiliation(s)
- María Villa-Morales
- Department of Biology, Universidad Autónoma de Madrid, CIBER de Enfermedades Raras, Madrid, Spain
| | | |
Collapse
|
32
|
Draper JM, Xia Z, Smith RA, Zhuang Y, Wang W, Smith CD. Discovery and evaluation of inhibitors of human ceramidase. Mol Cancer Ther 2011; 10:2052-61. [PMID: 21885864 DOI: 10.1158/1535-7163.mct-11-0365] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ceramide/sphingosine-1-phosphate (S1P) rheostat has been hypothesized to play a critical role in regulating tumor cell fate, with elevated levels of ceramide inducing death and elevated levels of S1P leading to survival and proliferation. Ceramidases are key enzymes that control this rheostat by hydrolyzing ceramide to produce sphingosine and may also confer resistance to drugs and radiation. Therefore, ceramidase inhibitors have excellent potential for development as new anticancer drugs. In this study, we identify a novel ceramidase inhibitor (Ceranib-1) by screening a small molecule library and describe the synthesis of a more potent analogue (Ceranib-2). In a cell-based assay, both compounds were found to inhibit cellular ceramidase activity toward an exogenous ceramide analogue, induce the accumulation of multiple ceramide species, decrease levels of sphingosine and S1P, inhibit the proliferation of cells alone and in combination with paclitaxel, and induce cell-cycle arrest and cell death. In vivo, Ceranib-2 was found to delay tumor growth in a syngeneic tumor model without hematologic suppression or overt signs of toxicity. These data support the selection of ceramidases as suitable targets for anticancer drug development and provide the first nonlipid inhibitors of human ceramidase activity.
Collapse
Affiliation(s)
- Jeremiah M Draper
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|
33
|
Ji X, Jiang C, Liu Y, Bu D, Xiao S. Fas ligand gene transfer effectively induces apoptosis in head and neck cancer cells. Acta Otolaryngol 2011; 131:876-81. [PMID: 21504271 DOI: 10.3109/00016489.2011.562539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONCLUSION Fas ligand (FasL) gene therapy may provide a new efficient therapeutic model for head and neck squamous cell cancer (HNSCC). Acid ceramidase (AC) may not play an important role in the sensitivity of HNSCC cell lines to Fas-induced apoptosis. OBJECTIVES The aims of this study were to investigate the efficacy of FasL gene therapy for HNSCC in vitro and to determine whether the expression of AC in different kinds of HNSCC cell lines is related to the sensitivity of HNSCC cell lines to Fas-mediated apoptotic induction. METHODS Three HNSCC cell lines (Hep-2, MMSI, and SCCVII) were transfected with pEGFP-FasL, a plasmid containing a modified human FasL gene fused to enhanced green fluorescent protein (GFP). pEGFP-C1, a plasmid containing the GFP gene alone, was used as a control. Cell death was observed by fluorescence imaging and quantified using a tetrazolium-based (MTS) assay. SCCVII cells were analyzed by flow cytometry to determine the presence of apoptotic induction. Hep-2 and MMSI cells were evaluated by quantitative real-time PCR to evaluate the expression of AC. RESULTS Transfection of pEGFP-FasL plasmid was shown to be able to induce cell death, the sensitivity of Fas-mediated apoptosis in HNSCC was different, and the level of AC did not correlate with the sensitivity of HNSCC cells to Fas-induced apoptosis.
Collapse
Affiliation(s)
- Xiaojun Ji
- Department of Otolaryngology-Head & Neck Surgery, Peking University First Hospital, Beijing, China
| | | | | | | | | |
Collapse
|
34
|
Gouazé-Andersson V, Flowers M, Karimi R, Fabriás G, Delgado A, Casas J, Cabot MC. Inhibition of acid ceramidase by a 2-substituted aminoethanol amide synergistically sensitizes prostate cancer cells to N-(4-hydroxyphenyl) retinamide. Prostate 2011; 71:1064-73. [PMID: 21557271 DOI: 10.1002/pros.21321] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Accepted: 11/18/2010] [Indexed: 11/12/2022]
Abstract
BACKGROUND The purpose of this study was to determine whether the therapeutic efficacy of fenretinide (4-HPR), a ceramide-generating anticancer agent, could be enhanced in prostate cancer cells by inclusion of a novel synthetic acid ceramidase (AC) inhibitor, DM102, a pivaloylamide of a 2-substituted aminoethanol. In prostate cancer, AC plays a role in progression and resistance to chemotherapy. METHODS PC-3 and DU 145 hormone-refractory human prostate cancer cell lines were used. Cells were exposed to 4-HPR, DM102, and combinations; viability, apoptosis, cell migration, ceramide metabolism, and levels of reactive oxygen species (ROS) were assessed. RESULTS Single agent 4-HPR and DM102 (2.5-10 µM) were weakly cytotoxic; however, combinations synergistically decreased cell viably to as low as 1.5% of control. N-oleoylethanolamine (NOE), a frequently employed AC inhibitor, was not effective in producing synergy. The 4-HPR/DM102 regimen enhanced caspase activity and increased [(3) H](dihydro)ceramide and ROS levels 6- and 30-fold over control, respectively. The antioxidant vitamin E, but not the de novo ceramide synthesis inhibitor myriocin, partially rescued cells from 4-HPR/DM102 cytotoxicity. The 4-HPR/DM102 combination also elicited synergistic cytotoxicity in DU 145 cells, another human hormone-refractory prostate cancer cell line. CONCLUSION This study shows that 4-HPR cytotoxicity is enhanced in a synergistic fashion by inclusion of the AC inhibitor DM102, by a mechanism that enlists generation of ROS, and thus provides a system to raise 4-HPR therapeutic potential. The role of ceramide however in the cytotoxic response is not clear, as blocking ceramide generation failed to rescue PC-3 cells from 4-HPR/DM102 cytotoxicity.
Collapse
|
35
|
Apoptotic sphingolipid ceramide in cancer therapy. J Lipids 2011; 2011:565316. [PMID: 21490804 PMCID: PMC3066853 DOI: 10.1155/2011/565316] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 10/26/2010] [Indexed: 11/18/2022] Open
Abstract
Apoptosis, also called programmed cell death, is physiologically and pathologically involved in cellular homeostasis. Escape of apoptotic signaling is a critical strategy commonly used for cancer tumorigenesis. Ceramide, a derivative of sphingolipid breakdown products, acts as second messenger for multiple extracellular stimuli including growth factors, chemical agents, and environmental stresses, such as hypoxia, and heat stress as well as irradiation. Also, ceramide acts as tumor-suppressor lipid because a variety of stress stimuli cause apoptosis by increasing intracellular ceramide to initiate apoptotic signaling. Defects on ceramide generation and sphingolipid metabolism are developed for cancer cell survival and cancer therapy resistance. Alternatively, targeting ceramide metabolism to correct these defects might provide opportunities to overcome cancer therapy resistance.
Collapse
|
36
|
Kim YJ, Kim EA, Sohn UD, Yim CB, Im C. Cytotoxic Activity and Structure Activity Relationship of Ceramide Analogues in Caki-2 and HL-60 Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2010; 14:441-7. [PMID: 21311687 DOI: 10.4196/kjpp.2010.14.6.441] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 12/03/2010] [Accepted: 12/10/2010] [Indexed: 12/23/2022]
Abstract
B13, a ceramide analogue, is a ceramidase inhibitor and induces apoptosis to give potent anticancer activity. A series of thiourea B13 analogues was evaluated for their in vitro cytotoxic activities against human renal cancer Caki-2 and leukemic cancer HL-60 in the MTT assay. Some compounds (12, 15, and 16) showed stronger cytotoxicity than B13 and C6-ceramide against both tumor cell lines, and compound (12) gave the most potent activity with IC(50) values of 36 and 9 µM, respectively. Molecular modeling of thiourea B13 analogues was carried out by comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA). We obtained highly reliable and predictive CoMSIA models with cross-validated q(2) values of 0.707 and 0.753 and CoMSIA contour maps to show the structural requirements for potent activity. These data suggest that the amide group of B13 could be replaced by thiourea, that the stereochemistry of 1,3-propandiol may not be essential for activity and that long alkyl chains increase cytotoxicity.
Collapse
Affiliation(s)
- Yong Jin Kim
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | | | | | | | | |
Collapse
|
37
|
Turner LS, Cheng JC, Beckham TH, Keane TE, Norris JS, Liu X. Autophagy is increased in prostate cancer cells overexpressing acid ceramidase and enhances resistance to C6 ceramide. Prostate Cancer Prostatic Dis 2010; 14:30-7. [PMID: 21116286 PMCID: PMC4203650 DOI: 10.1038/pcan.2010.47] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Acid ceramidase (AC) overexpression has been observed in prostate cancer cell lines and primary tumors, and contributes to resistance to chemotherapy and radiation. The consequence of AC overexpression is the ability to convert ceramide, which is often produced as a proapoptotic response to stress, to sphingosine, which can then be converted to the prosurvival molecule sphingosine-1-phosphate. In addition to their ability to metabolize ceramide produced in response to stress, we show here that prostate cancer cell lines overexpressing AC also have increased lysosomal density and increased levels of autophagy. Furthermore, pretreatment with 3-methyladenine restores sensitivity of these cells to treatment with C(6) ceramide. We also observed increased expression of the lysosomal stabilizing protein KIF5B and increased sensitivity to the lysosomotropic agent LCL385. Thus, we conclude that AC overexpression increases autophagy in prostate cancer cells, and that increased autophagy enhances resistance to ceramide.
Collapse
Affiliation(s)
- L S Turner
- Department of Biology, Francis Marion University, Florence, SC, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Beckham TH, Elojeimy S, Cheng JC, Turner LS, Hoffman SR, Norris JS, Liu X. Targeting sphingolipid metabolism in head and neck cancer: rational therapeutic potentials. Expert Opin Ther Targets 2010; 14:529-39. [PMID: 20334489 DOI: 10.1517/14728221003752768] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
IMPORTANCE OF THE FIELD Ceramide accumulation has been shown to be a conserved mechanism of apoptosis initiation in normal physiological processes as well as in response to cancer treatments. Therefore, it is unsurprising that many cancers develop aberrations of sphingolipid metabolism that prevent the accumulation of ceramide, whether by reduction of ceramide generation or by enhanced ceramide catabolism, particularly dangerous when catabolism leads to generation of pro-tumor sphingosine-1-phosphate and ceramide-1-phosphate. Numerous studies have now implicated dysregulation of sphingolipid metabolism in head and neck cancers. AREAS COVERED IN THIS REVIEW This review highlights the importance of sphingolipid metabolism and brings sphingolipid metabolism to the forefront in the investigation of novel therapies for head and neck cancer. It reviews sphingolipid-centric therapies under investigation in preclinical and clinical trials of cancers of the head and neck. WHAT THE READER WILL GAIN The roles of sphingolipids and sphingolipid metabolism in cancer are reviewed and the reader will be brought up to date with discoveries in the field of sphingolipid metabolism in head and neck cancer. TAKE HOME MESSAGE As treatments for head and neck cancers are currently limited, the potential of targeting sphingolipid metabolism should be taken into consideration as we seek novel ways to combat this group of tumors.
Collapse
Affiliation(s)
- Thomas H Beckham
- Medical University of South Carolina, Department of Biochemistry & Molecular Biology, 173 Ashley Avenue, MSC 504, Charleston, South Carolina 29425-5040, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Xia Z, Draper JM, Smith CD. Improved synthesis of a fluorogenic ceramidase substrate. Bioorg Med Chem 2010; 18:1003-9. [PMID: 20085856 PMCID: PMC2841511 DOI: 10.1016/j.bmc.2009.12.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2009] [Revised: 12/24/2009] [Accepted: 12/31/2009] [Indexed: 12/11/2022]
Abstract
Substantial interest has focused on the roles of sphingolipid metabolizing enzymes in a variety of hyperproliferative and inflammatory diseases. A key family of enzymes involved in these pathologies is the ceramidases. Ceramidases cleave the pro-apoptotic lipid ceramide into a long-chain fatty acid and sphingosine, which can then be further metabolized to the mitogenic and inflammatory lipid sphingosine 1-phosphate. Consequently, development of ceramidase inhibitors would provide useful pharmacologic probes for further studies of sphingolipid metabolism, as well as lead compounds for drug development. This effort has been hampered by the lack of in vitro and cellular ceramidase assays that are amenable to high-throughput screening. Recently, a fluorogenic ceramide analog has been described as a substrate for use in ceramidase assays. The synthesis of this compound has now been substantially improved in terms of both the required effort and the overall yield of the process. Key improvements include: reduction in number of required steps, use of a hydroboration reaction; incorporation of a Mitsunobu reaction; improved acylation by the addition of triethylamine; together providing a fourfold increase in the overall yield. In addition, it has been demonstrated that the ceramide analog can be used in high-throughput assays to identify ceramidase inhibitors. Overall, the improved efficiency in the preparation of this ceramidase substrate should accelerate discovery efforts relating to sphingolipid metabolism.
Collapse
Affiliation(s)
| | | | - Charles D. Smith
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, MSC 140, Charleston, SC 29425, United States
| |
Collapse
|
40
|
Guinn B, Casey G, Möller MG, Kasahara N, O'Sullivan GC, Peng KW, Tangney M. International Society for Cell and Gene Therapy of Cancer 2009 Annual Meeting Held in Cork, Ireland. Hum Gene Ther 2010; 21:9-26. [DOI: 10.1089/hum.2009.205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Barbara Guinn
- Cancer Sciences Division (MP824), University of Southampton School of Medicine, Somers Cancer Research Building, Southampton General Hospital, Southampton SO16 6YD, UK
- Department of Haematological Medicine, King's College London School of Medicine, London SE5 9NU, UK
| | - Garrett Casey
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
| | - Mecker G. Möller
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL 33136
| | - Noriyuki Kasahara
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Gerald C. O'Sullivan
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905
| | - Mark Tangney
- Cork Cancer Research Centre, Mercy University Hospital and Leslie C. Quick Jr. Laboratory, University College Cork, Cork, Ireland
| |
Collapse
|
41
|
Liu X, Cheng JC, Turner LS, Elojeimy S, Beckham TH, Bielawska A, Keane TE, Hannun YA, Norris JS. Acid ceramidase upregulation in prostate cancer: role in tumor development and implications for therapy. Expert Opin Ther Targets 2009; 13:1449-58. [PMID: 19874262 PMCID: PMC2796572 DOI: 10.1517/14728220903357512] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bioactive sphingolipids, such as ceramide, sphingosine and sphingosine-1-phosphate are known bio-effector molecules which play important roles in various aspects of cancer biology including cell proliferation, growth arrest, apoptosis, metastasis, senescence and inflammation. Therefore, enzymes involved in ceramide metabolism are gaining recognition as being critical regulators of cancer cell growth and/or survival. We previously observed that the ceramide metabolizing enzyme, acid ceramidase (AC) is upregulated in tumor tissues. Studies have now concluded that this creates a dysfunctional ceramide pathway, which is responsible for tumor progression and resistance to chemotherapy and radiation. This suggests that development of small-molecule drugs that inhibit AC enzyme activity is a promising approach for improving standard cancer therapy and patient's clinical outcomes.
Collapse
Affiliation(s)
- Xiang Liu
- Assistant Professor, Division of Basic Sciences, Departments of: Biochemistry & Molecular Biology, Cell and Molecular Pharmacology & Experimental Therapeutics, Microbiology & Immunology, MUSC, 173 Ashley Avenue, MSC 504, Charleston, South Carolina 29425-5040, FAX: 843.792.4882, Phone: 843.792.7412
| | - Joseph C. Cheng
- MD/PhD Student, Division of Basic Sciences, Departments of: Biochemistry & Molecular Biology, Cell and Molecular Pharmacology & Experimental Therapeutics, Microbiology & Immunology, MUSC, 173 Ashley Avenue, MSC 504, Charleston, South Carolina 29425-5040, FAX: 843.792.4882, Phone: 843.792.8499
| | - Lorianne S. Turner
- Postdoctoral Fellow, Division of Basic Sciences, Departments of: Biochemistry & Molecular Biology, Cell and Molecular Pharmacology & Experimental Therapeutics, Microbiology & Immunology, MUSC, 173 Ashley Avenue, MSC 504, Charleston, South Carolina 29425-5040, FAX: 843.792.4882, Phone: 843.792.8499
| | - Saeed Elojeimy
- Division of Basic Sciences, Departments of: Biochemistry & Molecular Biology, Cell and Molecular Pharmacology & Experimental Therapeutics, Microbiology & Immunology, MUSC, 173 Ashley Avenue, MSC 504, Charleston, South Carolina 29425-5040, FAX: 843.792.4882, Phone: 843.814.7010
| | - Thomas H. Beckham
- MD/PhD Student, Departments of: Biochemistry & Molecular Biology, Cell and Molecular Pharmacology & Experimental Therapeutics, Microbiology & Immunology, MUSC, 173 Ashley Avenue, MSC 504, Charleston, South Carolina 29425-5040, FAX: 843.792.4882, Phone: 843.792.8499
| | - Alicja Bielawska
- Professor, Departments of: Biochemistry & Molecular Biology, Cell and Molecular Pharmacology & Experimental Therapeutics, Microbiology & Immunology, MUSC, 173 Ashley Avenue, MSC 504, Charleston, South Carolina 29425-5040, FAX: 843.792.1627, Phone: 843.792.0273
| | - Thomas E. Keane
- Professor and Chair, Department of Urology, MUSC, 96 Jonathan Lucas Street, Room 644, Clinical Science Building, Phone: 843.792.1666
| | - Yusuf A. Hannun
- Senior Associate Dean for Basic Sciences, Director, Division of Basic Sciences, Distinguished University Professor, Chair, Department of Biochemistry & Molecular Biology, Cell and Molecular Pharmacology & Experimental Therapeutics, Microbiology & Immunology, MUSC, 173 Ashley Avenue, MSC 509, Charleston, South Carolina 29425-5090, FAX: 843.792.4322, Phone: 843.792.9318
| | - James S. Norris
- Professor and Chair, Department of Microbiology & Immunology, MUSC, 173 Ashley Avenue, MSC 504, Charleston, South Carolina 29425-5040, FAX: 843.792.4882, Phone: 843.792.7915
| |
Collapse
|
42
|
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: 94] [Impact Index Per Article: 5.5] [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
|
43
|
Papenfuss K, Cordier SM, Walczak H. Death receptors as targets for anti-cancer therapy. J Cell Mol Med 2008; 12:2566-85. [PMID: 19210756 PMCID: PMC3828874 DOI: 10.1111/j.1582-4934.2008.00514.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Accepted: 09/25/2008] [Indexed: 01/06/2023] Open
Abstract
Human tumour cells are characterized by their ability to avoid the normal regulatory mechanisms of cell growth, division and death. The classical chemotherapy aims to kill tumour cells by causing DNA damage-induced apoptosis. However, as many tumour cells possess mutations in intracellular apoptosis-sensing molecules like p53, they are not capable of inducing apoptosis on their own and are therefore resistant to chemotherapy. With the discovery of the death receptors the opportunity arose to directly trigger apoptosis from the outside of tumour cells, thereby circumventing chemotherapeutic resistance. Death receptors belong to the tumour necrosis factor receptor superfamily, with tumour necrosis factor (TNF) receptor-1, CD95 and TNF-related apoptosis-inducing ligand-R1 and -R2 being the most prominent members. This review covers the current knowledge about these four death receptors, summarizes pre-clinical approaches engaging these death receptors in anti-cancer therapy and also gives an overview about their application in clinical trials conducted to date.
Collapse
Affiliation(s)
| | | | - Henning Walczak
- Tumour Immunology Unit, Division of Medicine, Imperial College LondonUnited Kingdom
| |
Collapse
|
44
|
Zeidan YH, Jenkins RW, Korman JB, Liu X, Obeid LM, Norris JS, Hannun YA. Molecular targeting of acid ceramidase: implications to cancer therapy. Curr Drug Targets 2008; 9:653-61. [PMID: 18691012 DOI: 10.2174/138945008785132358] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increasingly recognized as bioactive molecules, sphingolipids have been studied in a variety of disease models. The impact of sphingolipids on cancer research facilitated the entry of sphingolipid analogues and enzyme modulators into clinical trials. Owing to its ability to regulate two bioactive sphingolipids, ceramide and sphingosine-1-phosphate, acid ceramidase (AC) emerges as an attractive target for drug development within the sphingolipid metabolic pathway. Indeed, there is extensive evidence supporting a pivotal role for AC in lipid metabolism and cancer biology. In this article, we review the current knowledge of the biochemical properties of AC, its relevance to tumor promotion, and its molecular targeting approaches.
Collapse
Affiliation(s)
- Youssef H Zeidan
- Department of Biochemistry and Molecular Biology Medical University of South Carolina .175 Ashley Avenue, P.O. Box 250509. Charleston, South Carolina, 29425, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Szulc ZM, Mayroo N, Bai A, Bielawski J, Liu X, Norris JS, Hannun YA, Bielawska A. Novel analogs of D-e-MAPP and B13. Part 1: synthesis and evaluation as potential anticancer agents. Bioorg Med Chem 2008; 16:1015-31. [PMID: 17869115 PMCID: PMC2287182 DOI: 10.1016/j.bmc.2007.08.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 08/16/2007] [Accepted: 08/20/2007] [Indexed: 12/31/2022]
Abstract
A series of novel isosteric analogs of the ceramidase inhibitors, (1S,2R)-N-myristoylamino-phenylpropanol-1 (d-e-MAPP) and (1R,2R)-N-myristoylamino-4'-nitro-phenylpropandiol-1,3 (B13), with modified targeting and physicochemical properties were designed, synthesized, and evaluated as potential anticancer agents. When MCF7 cells were treated with the analogs, results indicated that the new analogs were of equal or greater potency compared to the parent compounds. Their activity was predominantly defined by the nature of the modification of the N-acyl hydrophobic interfaces: N-acyl analogs (class A), urea analogs (class B), N-alkyl analogs (class C, lysosomotropic agents), and omega-cationic-N-acyl analogs (class D, mitochondriotropic agents). The most potent compounds belonged to either class D, the aromatic ceramidoids, or to class C, the aromatic N-alkylaminoalcohols. Representative analogs selected from this study were also evaluated by the National Cancer Institute In Vitro Anticancer Drug Discovery Screen. Again, results showed a similar class-dependent activity. In general, the active analogs were non-selectively broad spectrum and had promising activity against all cancer cell lines. However, some active analogs of the d-e-MAPP family were selective against different types of cancer. Compounds LCL85, LCL120, LCL385, LCL284, and LCL204 were identified to be promising lead compounds for therapeutic development.
Collapse
Affiliation(s)
- Zdzislaw M. Szulc
- Departments of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - Nalini Mayroo
- Departments of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - AiPing Bai
- Departments of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - Jacek Bielawski
- Departments of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - Xiang Liu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - James S. Norris
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Yusuf A. Hannun
- Departments of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| | - Alicja Bielawska
- Departments of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
46
|
Bielawska A, Bielawski J, Szulc ZM, Mayroo N, Liu X, Bai A, Elojeimy S, Rembiesa B, Pierce J, Norris JS, Hannun YA. Novel analogs of D-e-MAPP and B13. Part 2: signature effects on bioactive sphingolipids. Bioorg Med Chem 2008; 16:1032-45. [PMID: 17881234 PMCID: PMC2268750 DOI: 10.1016/j.bmc.2007.08.032] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 08/16/2007] [Accepted: 08/20/2007] [Indexed: 11/30/2022]
Abstract
Novel isosteric analogs of the ceramidase inhibitors (1S,2R)-N-myristoylamino-phenylpropanol-1 (d-e-MAPP) and (1R,2R)-N-myristoylamino-4'-nitro-phenylpropandiol-1,3 (B13) with modified targeting and physicochemical properties were developed and evaluated for their effects on endogenous bioactive sphingolipids: ceramide, sphingosine, and sphingosine 1-phosphate (Cer, Sph, and S1P) in MCF7 cells as determined by high-performance liquid chromatography-mass spectrometry (HPLC-MS/MS). Time- and dose-response studies on the effects of these compounds on Cer species and Sph levels, combined with structure-activity relationship (SAR) data, revealed 4 distinct classes of analogs which were predominantly defined by modifications of the N-acyl-hydrophobic interfaces: N-acyl-analogs (class A), urea-analogs (class B), N-alkyl-analogs (class C), and omega-cationic-N-acyl analogs (class D). Signature patterns recognized for two of the classes correspond to the cellular compartment of action of the new analogs, with class D acting as mitochondriotropic agents and class C compounds acting as lysosomotropic agents. The neutral agents, classes A and B, do not have this compartmental preference. Moreover, we observed a close correlation between the selective increase of C(16)-, C(14)-, and C(18)-Cers and inhibitory effects on MCF7 cell growth. The results are discussed in the context of compartmentally targeted regulators of Sph, Cer species, and S1P in cancer cell death, emphasizing the role of C(16)-Cer. These novel analogs should be useful in cell-based studies as specific regulators of Cer-Sph-S1P inter-metabolism, in vitro enzymatic studies, and for therapeutic development.
Collapse
Affiliation(s)
- Alicja Bielawska
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, 173 Ashley Avenue, PO Box 250509, Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Holman DH, Turner LS, El-Zawahry A, Elojeimy S, Liu X, Bielawski J, Szulc ZM, Norris K, Zeidan YH, Hannun YA, Bielawska A, Norris JS. Lysosomotropic acid ceramidase inhibitor induces apoptosis in prostate cancer cells. Cancer Chemother Pharmacol 2007; 61:231-42. [PMID: 17429631 DOI: 10.1007/s00280-007-0465-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Accepted: 03/04/2007] [Indexed: 12/25/2022]
Abstract
PURPOSE Alterations in ceramide metabolism have been reported in prostate cancer (PCa), resulting in escape of cancer cells from ceramide-induced apoptosis. Specifically, increased expression of lysosomal acid ceramidase (AC) has been shown in some primary PCa tissues and in several PCa cell lines. To determine if this represents a novel therapeutic target, we designed and synthesized LCL204, a lysosomotropic analog of B13, a previously reported inhibitor of AC METHODS: Prostate cancer cell lines were treated with LCL204 for varying times and concentrations. Effects of treatment on cytotoxicity, sphingolipid content, and apoptotic markers were assessed. RESULTS Treatment of DU145 PCa cells resulted in increased ceramide and decreased sphingosine levels. Interestingly, LCL204 caused degradation of AC in a cathepsin-dependent manner. We also observed rapid destabilization of lysosomes and the release of lysosomal proteases into the cytosol following treatment with LCL204. Combined, these events resulted in mitochondria depolarization and executioner caspase activation, ultimately ending in apoptosis CONCLUSIONS These results provide evidence that treatment with molecules such as LCL204, which restore ceramide levels in PCa cells may serve as a new viable treatment option for PCa.
Collapse
Affiliation(s)
- David H Holman
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29403, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Elojeimy S, Liu X, McKillop JC, El-Zawahry AM, Holman DH, Cheng JY, Meacham WD, Mahdy AE, Saad AF, Turner LS, Cheng J, A Day T, Dong JY, Bielawska A, Hannun YA, Norris JS. Role of acid ceramidase in resistance to FasL: therapeutic approaches based on acid ceramidase inhibitors and FasL gene therapy. Mol Ther 2007; 15:1259-63. [PMID: 17426710 DOI: 10.1038/sj.mt.6300167] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Head and neck squamous cell cancers (HNSCC) are particularly aggressive and are resistant to many forms of treatment. Ceramide metabolism has been shown to play an important role in cancer progression and cancer resistance to therapy in many tumor models, including HNSCC. Here, we study the role of the ceramide-metabolizing enzyme acid ceramidase (AC) in therapeutic responses in HNSCC. First, we show that AC is over-expressed in 70% of head and neck squamous cell tumors compared with normal tissues, suggesting that this enzyme may play an important role in facilitating HNSCC growth. Next, comparison of three HNSCC cell lines with low, medium, and high levels of AC reveals an inverse correlation between the levels of AC and their response to exogenous C-6-ceramide. Furthermore, over-expression of AC in SCC-1 cells increased resistance to Fas-induced cell killing. Conversely, down-regulation of AC using specific AC small interfering RNA (siRNA) sensitized the SCC-1 cancer cell line to Fas-induced apoptosis. Finally, we show that the AC inhibitor LCL 204 can sensitize HNSCC cell lines to Fas-induced apoptosis both in vitro and in a xenograft model in vivo, suggesting that the combination of FasL gene therapy and LCL 204 may become a new treatment option for advanced-stage head and neck cancer.
Collapse
Affiliation(s)
- Saeed Elojeimy
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|