1
|
Terreiro JFPR, Marquês JT, Antunes I, de Faria CF, Santos S, Martins F, de Almeida RFM. Membrane interaction studies of isoniazid derivatives active against drug-resistant tuberculosis. Eur J Pharm Sci 2025; 205:106986. [PMID: 39674553 DOI: 10.1016/j.ejps.2024.106986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/22/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Tuberculosis is one of the leading causes of mortality worldwide due to the growth of multi-drug resistant strains unsusceptible to currently available therapies. Four compounds, isoniazid (INH) and three derivatives, N'-decanoylisonicotinohydrazide (INHC10), N'-(E)-(4-phenoxybenzylidene)isonicotinohydrazide (N34) and N'-(4-phenoxybenzyl)isonicotinohydrazide (N34red), were studied. Owing to their advantageous in vitro selectivity index against the primary mutation responsible for drug resistance in Mycobacterium tuberculosis (Mtb), as well as their suitable lipophilicity and interaction with human serum albumin, INHC10 and N34 were deemed promising antitubercular compounds. N34red, despite differing from N34 only in the saturation of the N' = C bond, presents a poor selectivity index. To delve deeper into the therapeutic potential of these compounds, their interaction with biomembrane models, mimicking biological barriers on the way to the target inside Mtb cells, was herein evaluated. All compounds, except N34red, weakened the packing of the acyl chains in the rigid lipid gel phase, especially INHC10, which was the only compound disturbing liquid disordered membranes. Notably, all compounds except INH decreased membrane dipole potential, across all types of bilayers studied, but only N34red had a drastic effect. The insertion in gel phase bilayers suggests that the compounds may be able to penetrate the rigid cell wall of Mtb. Förster's resonance energy transfer (FRET) assays in ternary bilayers with liquid ordered/liquid disordered lateral heterogeneity mimicking human cell membranes, showed that the compounds affected neither the size nor the organization of lipid domains. These results provide molecular insights into the low toxicity against human cell lines and improved activity against drug-resistant Mtb of INHC10 and N34.
Collapse
Affiliation(s)
- Joana F P R Terreiro
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal
| | - Joaquim T Marquês
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal.
| | - Inês Antunes
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal
| | - Catarina Frazão de Faria
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal
| | - Susana Santos
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal
| | - Filomena Martins
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal.
| | - Rodrigo F M de Almeida
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal.
| |
Collapse
|
2
|
Sosa Ponce ML, Cobb JA, Zaremberg V. Lipids and chromatin: a tale of intriguing connections shaping genomic landscapes. Trends Cell Biol 2025; 35:141-152. [PMID: 39060139 DOI: 10.1016/j.tcb.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024]
Abstract
Recent studies in yeast reveal an intricate interplay between nuclear envelope (NE) architecture and lipid metabolism, and between lipid signaling and both epigenome and genome integrity. In this review, we highlight the reciprocal connection between lipids and histone modifications, which enable metabolic reprogramming in response to nutrients. The endoplasmic reticulum (ER)-NE regulates the compartmentalization and temporal availability of epigenetic metabolites and its lipid composition also impacts nuclear processes, such as transcriptional silencing and the DNA damage response (DDR). We also discuss recent work providing mechanistic insight into lipid droplet (LD) formation and sterols in the nucleus, and the collective data showing Opi1 as a central factor in both membrane sensing and transcriptional regulation of lipid-chromatin interrelated processes.
Collapse
Affiliation(s)
- Maria Laura Sosa Ponce
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Jennifer A Cobb
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Vanina Zaremberg
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
3
|
Nel L, Thaysen K, Jamecna D, Olesen E, Szomek M, Langer J, Frain KM, Höglinger D, Wüstner D, Pedersen BP. Structural and biochemical analysis of ligand binding in yeast Niemann-Pick type C1-related protein. Life Sci Alliance 2025; 8:e202402990. [PMID: 39455279 PMCID: PMC11512107 DOI: 10.26508/lsa.202402990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
In eukaryotes, integration of sterols into the vacuolar/lysosomal membrane is critically dependent on the Niemann-Pick type C (NPC) system. The system consists of an integral membrane protein, called NCR1 in yeast, and NPC2, a luminal soluble protein that transfers sterols to the N-terminal domain (NTD) of NCR1 before membrane integration. Both proteins have been implicated in sterol homeostasis of yeast and humans. Here, we investigate sterol and lipid binding of the NCR1/NPC2 transport system and determine crystal structures of the sterol binding NTD. The NTD binds both ergosterol and cholesterol, with nearly identical conformations of the binding pocket. Apart from sterols, the NTD can also bind fluorescent analogs of phosphatidylinositol, phosphatidylcholine, and phosphatidylserine, as well as sphingosine and ceramide. We confirm the multi-lipid scope of the NCR1/NPC2 system using photo-crosslinkable and clickable lipid analogs, namely, pac-cholesterol, pac-sphingosine, and pac-ceramide. Finally, we reconstitute the transfer of pac-sphingosine from NPC2 to the NTD in vitro. Collectively, our results support that the yeast NPC system can work as versatile machinery for vacuolar homeostasis of structurally diverse lipids, besides ergosterol.
Collapse
Affiliation(s)
- Lynette Nel
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Katja Thaysen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Denisa Jamecna
- Heidelberg University, Biochemistry Center, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - Esben Olesen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Maria Szomek
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Julia Langer
- Heidelberg University, Biochemistry Center, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - Kelly M Frain
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Doris Höglinger
- Heidelberg University, Biochemistry Center, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Bjørn P Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
4
|
Alonso-Pérez V, Hernández V, Calzado MA, Vicente-Blázquez A, Gajate C, Soler-Torronteras R, DeCicco-Skinner K, Sierra A, Mollinedo F. Suppression of metastatic organ colonization and antiangiogenic activity of the orally bioavailable lipid raft-targeted alkylphospholipid edelfosine. Biomed Pharmacother 2024; 171:116149. [PMID: 38266621 DOI: 10.1016/j.biopha.2024.116149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/26/2024] Open
Abstract
Metastasis is the leading cause of cancer mortality. Metastatic cancer is notoriously difficult to treat, and it accounts for the majority of cancer-related deaths. The ether lipid edelfosine is the prototype of a family of synthetic antitumor compounds collectively known as alkylphospholipid analogs, and its antitumor activity involves lipid raft reorganization. In this study, we examined the effect of edelfosine on metastatic colonization and angiogenesis. Using non-invasive bioluminescence imaging and histological examination, we found that oral administration of edelfosine in nude mice significantly inhibited the lung and brain colonization of luciferase-expressing 435-Lung-eGFP-CMV/Luc metastatic cells, resulting in prolonged survival. In metastatic 435-Lung and MDA-MB-231 breast cancer cells, we found that edelfosine also inhibited cell adhesion to collagen-I and laminin-I substrates, cell migration in chemotaxis and wound-healing assays, as well as cancer cell invasion. In 435-Lung and other MDA-MB-435-derived sublines with different organotropism, edelfosine induced G2/M cell cycle accumulation and apoptosis in a concentration- and time-dependent manner. Edelfosine also inhibited in vitro angiogenesis in human and mouse endothelial cell tube formation assays. The antimetastatic properties were specific to cancer cells, as edelfosine had no effects on viability in non-cancerous cells. Edelfosine accumulated in membrane rafts and endoplasmic reticulum of cancer cells, and membrane raft-located CD44 was downregulated upon drug treatment. Taken together, this study highlights the potential of edelfosine as an attractive drug to prevent metastatic growth and organ colonization in cancer therapy. The raft-targeted drug edelfosine displays a potent activity against metastatic organ colonization and angiogenesis, two major hallmarks of tumor malignancy.
Collapse
Affiliation(s)
- Verónica Alonso-Pérez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Centro de Investigación del Cáncer (CIC), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Vanessa Hernández
- Biological Clues of the Invasive and Metastatic Phenotype Group, Molecular Oncology Department, Bellvitge Biomedical Research Institute (IDIBELL), E-08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Marco A Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), E-14004 Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, E-14004 Córdoba, Spain; Hospital Universitario Reina Sofía, E-14004 Córdoba, Spain
| | - Alba Vicente-Blázquez
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, C/ Ramiro de Maeztu 9, E-28040 Madrid, Spain; Department of Biology, American University, Washington, DC 20016, USA
| | - Consuelo Gajate
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Centro de Investigación del Cáncer (CIC), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, C/ Ramiro de Maeztu 9, E-28040 Madrid, Spain
| | - Rafael Soler-Torronteras
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), E-14004 Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, E-14004 Córdoba, Spain; Hospital Universitario Reina Sofía, E-14004 Córdoba, Spain
| | | | - Angels Sierra
- Biological Clues of the Invasive and Metastatic Phenotype Group, Molecular Oncology Department, Bellvitge Biomedical Research Institute (IDIBELL), E-08907 L'Hospitalet de Llobregat, Barcelona, Spain; Laboratory of Experimental Oncological Neurosurgery, Neurosurgery Service, Hospital Clinic de Barcelona-FCRB, E-08036 Barcelona, Spain; Department of Medicine and Life Sciences (MELIS), Faculty of Health and Live Sciences, Universitat Pompeu Fabra, E-08036 Barcelona, Spain
| | - Faustino Mollinedo
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Centro de Investigación del Cáncer (CIC), Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, C/ Ramiro de Maeztu 9, E-28040 Madrid, Spain.
| |
Collapse
|
5
|
Sosa Ponce ML, Remedios MH, Moradi-Fard S, Cobb JA, Zaremberg V. SIR telomere silencing depends on nuclear envelope lipids and modulates sensitivity to a lysolipid. J Cell Biol 2023; 222:e202206061. [PMID: 37042812 PMCID: PMC10103788 DOI: 10.1083/jcb.202206061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/29/2022] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
The nuclear envelope (NE) is important in maintaining genome organization. The role of lipids in communication between the NE and telomere regulation was investigated, including how changes in lipid composition impact gene expression and overall nuclear architecture. Yeast was treated with the non-metabolizable lysophosphatidylcholine analog edelfosine, known to accumulate at the perinuclear ER. Edelfosine induced NE deformation and disrupted telomere clustering but not anchoring. Additionally, the association of Sir4 at telomeres decreased. RNA-seq analysis showed altered expression of Sir-dependent genes located at sub-telomeric (0-10 kb) regions, consistent with Sir4 dispersion. Transcriptomic analysis revealed that two lipid metabolic circuits were activated in response to edelfosine, one mediated by the membrane sensing transcription factors, Spt23/Mga2, and the other by a transcriptional repressor, Opi1. Activation of these transcriptional programs resulted in higher levels of unsaturated fatty acids and the formation of nuclear lipid droplets. Interestingly, cells lacking Sir proteins displayed resistance to unsaturated-fatty acids and edelfosine, and this phenotype was connected to Rap1.
Collapse
Affiliation(s)
| | | | - Sarah Moradi-Fard
- Departments of Biochemistry and Molecular Biology and Oncology, Cumming School of Medicine, Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Calgary, Canada
| | - Jennifer A. Cobb
- Departments of Biochemistry and Molecular Biology and Oncology, Cumming School of Medicine, Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, Calgary, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
| | - Vanina Zaremberg
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| |
Collapse
|
6
|
Gelova SP, Chan K. Mutagenesis induced by protonation of single-stranded DNA is linked to glycolytic sugar metabolism. Mutat Res 2023; 826:111814. [PMID: 36634476 DOI: 10.1016/j.mrfmmm.2023.111814] [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: 10/09/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
Mutagenesis can be thought of as random, in the sense that the occurrence of each mutational event cannot be predicted with precision in space or time. However, when sufficiently large numbers of mutations are analyzed, recurrent patterns of base changes called mutational signatures can be identified. To date, some 60 single base substitution or SBS signatures have been derived from analysis of cancer genomics data. We recently reported that the ubiquitous signature SBS5 matches the pattern of single nucleotide polymorphisms (SNPs) in humans and has analogs in many species. Using a temperature-sensitive single-stranded DNA (ssDNA) mutation reporter system, we also showed that a similar mutational pattern in yeast is dependent on error-prone translesion DNA synthesis (TLS) and glycolytic sugar metabolism. Here, we further investigated mechanisms that are responsible for this form of mutagenesis in yeast. We first confirmed that excess sugar metabolism leads to increased mutation rate, which was detectable by fluctuation assay. Since glycolysis is known to produce excess protons, we then investigated the effects of experimental manipulations on pH and mutagenesis. We hypothesized that yeast metabolizing 8% glucose would produce more excess protons than cells metabolizing 2% glucose. Consistent with this, cells metabolizing 8% glucose had lower intracellular and extracellular pH values. Similarly, deletion of vma3 (encoding a vacuolar H+-ATPase subunit) increased mutagenesis. We also found that treating cells with edelfosine (which renders membranes more permeable, including to protons) or culturing in low pH media increased mutagenesis. Analysis of the mutational pattern attributable to 20 µM edelfosine treatment revealed similarity to the SBS5-like TLS- and glycolysis-dependant mutational patterns previously observed in ssDNA. Altogether, our results agree with multiple biochemical studies showing that protonation of nitrogenous bases can alter base pairing so as to stabilize some mispairs, and shed new light on a common form of intrinsic mutagenesis.
Collapse
Affiliation(s)
- Suzana P Gelova
- University of Ottawa Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada; Agriculture and Agri-Food Canada, 2585 County Road 20, Harrow, Ontario N0R 1G0, Canada
| | - Kin Chan
- University of Ottawa Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|
7
|
Starosta R, Santos TC, Dinis de Sousa AF, Santos MS, Corvo ML, Tomaz AI, de Almeida RFM. Assessing the role of membrane lipids in the action of ruthenium(III) anticancer compounds. Front Mol Biosci 2023; 9:1059116. [PMID: 36660430 PMCID: PMC9845782 DOI: 10.3389/fmolb.2022.1059116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
This work addresses the possible role of the cell membrane in the molecular mechanism of action of two salan-type ruthenium complexes that were previously shown to be active against human tumor cells, namely [Ru(III)(L1)(PPh3)Cl] and [Ru(III)(L2)(PPh3)Cl] (where L1 is 6,6'-(1R,2R)-cyclohexane-1,2-diylbis(azanediyl)bis(methylene)bis(3-methoxyphenol); and L2 is 2,2'-(1R,2R)-cyclohexane-1,2-diylbis(azanediyl)bis(methylene)bis(4-methoxyphenol)). One-component membrane models were first used, a disordered fluid bilayer of dioleoylphosphatodylcholine (DOPC), and an ordered rigid gel bilayer of dipalmitoylphosphatidylcholine. In addition, two quaternary mixtures of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin and cholesterol were used to mimic the lipid composition either of mammalian plasma membrane (1:1:1:1 mol ratio) or of a cancer cell line membrane (36.2:23.6:6.8:33.4 mol ratio). The results show that both salan ligands L1 and L2 bind relatively strongly to DOPC bilayers, but without significantly affecting their structure. The ruthenium complexes have moderate affinity for DOPC. However, their impact on the membranes was notable, leading to a significant increase in the permeability of the lipid vesicles. None of the compounds compromised liposome integrity, as revealed by dynamic light scattering. Fluorescence spectroscopy studies revealed changes in the biophysical properties of all membrane models analyzed in the presence of the two complexes, which promoted an increased fluidity and water penetration into the lipid bilayer in the one-component systems. In the quaternary mixtures, one of the complexes had an analogous effect (increasing water penetration), whereas the other complex reorganized the liquid ordered and liquid disordered domains. Thus, small structural differences in the metal ligands may lead to different outcomes. To better understand the effect of these complexes in cancer cells, the membrane dipole potential was also measured. For both Ru complexes, an increase in the dipole potential was observed for the cancer cell membrane model, while no alteration was detected on the non-cancer plasma membrane model. Our results show that the action of the Ru(III) complexes tested involves changes in the biophysical properties of the plasma membrane, and that it also depends on membrane lipid composition, which is frequently altered in cancer cells when compared to their normal counterparts.
Collapse
Affiliation(s)
- Radoslaw Starosta
- Faculty of Chemistry, University of Wroclaw, Wroclaw, Poland,Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Telma C. Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia F. Dinis de Sousa
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Soledade Santos
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - M. Luisa Corvo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Isabel Tomaz
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal,*Correspondence: Rodrigo F. M. de Almeida, ; Ana Isabel Tomaz,
| | - Rodrigo F. M. de Almeida
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal,*Correspondence: Rodrigo F. M. de Almeida, ; Ana Isabel Tomaz,
| |
Collapse
|
8
|
Clusters of apoptotic signaling molecule-enriched rafts, CASMERs: membrane platforms for protein assembly in Fas/CD95 signaling and targets in cancer therapy. Biochem Soc Trans 2022; 50:1105-1118. [PMID: 35587168 PMCID: PMC9246327 DOI: 10.1042/bst20211115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/13/2022]
Abstract
Mammalian cells show the ability to commit suicide through the activation of death receptors at the cell surface. Death receptors, among which Fas/CD95 is one of their most representative members, lack enzymatic activity, and depend on protein-protein interactions to signal apoptosis. Fas/CD95 death receptor-mediated apoptosis requires the formation of the so-called death-inducing signaling complex (DISC), bringing together Fas/CD95, Fas-associated death domain-containing protein and procaspase-8. In the last two decades, cholesterol-rich lipid raft platforms have emerged as scaffolds where Fas/CD95 can be recruited and clustered. The co-clustering of Fas/CD95 and rafts facilitates DISC formation, bringing procaspase-8 molecules to be bunched together in a limited membrane region, and leading to their autoproteolytic activation by oligomerization. Lipid raft platforms serve as a specific region for the clustering of Fas/CD95 and DISC, as well as for the recruitment of additional downstream signaling molecules, thus forming the so-called cluster of apoptotic signaling molecule-enriched rafts, or CASMER. These raft/CASMER structures float in the membrane like icebergs, in which the larger portion lies inside the cell and communicates with other subcellular structures to facilitate apoptotic signal transmission. This allows an efficient spatiotemporal compartmentalization of apoptosis signaling machinery during the triggering of cell death. This concept of proapoptotic raft platforms as a basic chemical-biological structure in the regulation of cell death has wide-ranging implications in human biology and disease, as well as in cancer therapy. Here, we discuss how these raft-centered proapoptotic hubs operate as a major linchpin for apoptosis signaling and as a promising target in cancer therapy.
Collapse
|
9
|
Gajate C, Gayet O, Fraunhoffer NA, Iovanna J, Dusetti N, Mollinedo F. Induction of Apoptosis in Human Pancreatic Cancer Stem Cells by the Endoplasmic Reticulum-Targeted Alkylphospholipid Analog Edelfosine and Potentiation by Autophagy Inhibition. Cancers (Basel) 2021; 13:cancers13236124. [PMID: 34885233 PMCID: PMC8656492 DOI: 10.3390/cancers13236124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies with a poor and gloomy prognosis and the highest mortality-to-incidence ratio. Pancreatic cancer remains an incurable malignancy, and current therapies are ineffective. We isolated cancer stem cells (CSCs) from the human PANC-1 pancreatic cancer cell line as CD44+CD24+EpCAM+ cells. These CSCs form pancreatic cancer spheres or spheroids and develop tumors in SCID mice after subcutaneous injection of as few as 100 cells per mouse. Here, we found that the alkylphospholipid analog edelfosine inhibited CSC pancreatic cancer spheroid formation and induced cell death, as assessed by an increase in the percentage of cells in the sub-G0/G1 region by means of flow cytometry, indicative of DNA breakdown and apoptosis. This correlated with an increase in caspase-3 activity and PARP breakdown, as a major substrate of caspase-3, following PANC-1 CSC treatment with edelfosine. The antitumor ether lipid edelfosine colocalized with the endoplasmic reticulum in both PANC-1 cells as well as PANC-1 CSCs by using a fluorescent edelfosine analog, and induced an endoplasmic reticulum stress response in both PANC-1 cells and PANC-1 CSCs, with a potent CHOP/GADD153 upregulation. Edelfosine elicited a strong autophagy response in both PANC-1 cells and PANC-1 CSCs, and preincubation of CSCs with autophagy inhibitors, chloroquine or bafilomycin A1, enhanced edelfosine-induced apoptosis. Primary cultures from pancreatic cancer patients were sensitive to edelfosine, as well as their respective isolated CSCs. Nontumorigenic pancreatic human cell line HPNE and normal human fibroblasts were largely spared. These data suggest that pancreatic CSCs isolated from established cell lines and pancreatic cancer patients are sensitive to edelfosine through its accumulation in the endoplasmic reticulum and induction of endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Consuelo Gajate
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, E-28040 Madrid, Spain;
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Campus Miguel de Unamuno, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, E-37007 Salamanca, Spain
| | - Odile Gayet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Parc Scientifique et Technologique de Luminy, CEDEX 09, 13288 Marseille, France; (O.G.); (N.A.F.); (J.I.); (N.D.)
| | - Nicolas A. Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Parc Scientifique et Technologique de Luminy, CEDEX 09, 13288 Marseille, France; (O.G.); (N.A.F.); (J.I.); (N.D.)
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Parc Scientifique et Technologique de Luminy, CEDEX 09, 13288 Marseille, France; (O.G.); (N.A.F.); (J.I.); (N.D.)
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille Université, Parc Scientifique et Technologique de Luminy, CEDEX 09, 13288 Marseille, France; (O.G.); (N.A.F.); (J.I.); (N.D.)
| | - Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, E-28040 Madrid, Spain;
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Campus Miguel de Unamuno, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, E-37007 Salamanca, Spain
- Correspondence:
| |
Collapse
|
10
|
Garcia JM, Schwabe MJ, Voelker DR, Riekhof WR. A functional genomic screen in Saccharomyces cerevisiae reveals divergent mechanisms of resistance to different alkylphosphocholine chemotherapeutic agents. G3-GENES GENOMES GENETICS 2021; 11:6347582. [PMID: 34568930 PMCID: PMC8496327 DOI: 10.1093/g3journal/jkab233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 05/03/2021] [Indexed: 11/12/2022]
Abstract
The alkylphosphocholine (APC) class of antineoplastic and antiprotozoal drugs, such as edelfosine and miltefosine, are structural mimics of lyso-phosphatidylcholine (lyso-PC), and are inhibitory to the yeast Saccharomyces cerevisiae at low micromolar concentrations. Cytotoxic effects related to inhibition of phospholipid synthesis, induction of an unfolded protein response, inhibition of oxidative phosphorylation, and disruption of lipid rafts have been attributed to members of this drug class, however, the molecular mechanisms of action of these drugs remain incompletely understood. Cytostatic and cytotoxic effects of the APCs exhibit variability with regard to chemical structure, leading to differences in effectiveness against different organisms or cell types. We now report the comprehensive identification of S. cerevisiae titratable-essential gene and haploid nonessential gene deletion mutants that are resistant to the APC drug miltefosine (hexadecyl-O-phosphocholine). Fifty-eight strains out of ∼5600 tested displayed robust and reproducible resistance to miltefosine. This gene set was heavily enriched in functions associated with vesicular transport steps, especially those involving endocytosis and retrograde transport of endosome derived vesicles to the Golgi or vacuole, suggesting a role for these trafficking pathways in transport of miltefosine to potential sites of action in the endoplasmic reticulum and mitochondrion. In addition, we identified mutants with defects in phosphatidylinositol-4-phosphate synthesis (TetO::STT4) and hydrolysis (sac1Δ), an oxysterol binding protein homolog (osh2Δ), a number of ER-resident proteins, and multiple components of the eisosome. These findings suggest that ER-plasma membrane contact sites and retrograde vesicle transport are involved in the interorganelle transport of lyso-PtdCho and related lyso-phospholipid-like analogs to their intracellular sites of cytotoxic activity.
Collapse
Affiliation(s)
- Jaquelin M Garcia
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Michael J Schwabe
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Dennis R Voelker
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Wayne R Riekhof
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
11
|
Mollinedo F, Gajate C. Direct Endoplasmic Reticulum Targeting by the Selective Alkylphospholipid Analog and Antitumor Ether Lipid Edelfosine as a Therapeutic Approach in Pancreatic Cancer. Cancers (Basel) 2021; 13:4173. [PMID: 34439330 PMCID: PMC8394177 DOI: 10.3390/cancers13164173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/11/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common malignancy of the pancreas, shows a dismal and grim overall prognosis and survival rate, which have remained virtually unchanged for over half a century. PDAC is the most lethal of all cancers, with the highest mortality-to-incidence ratio. PDAC responds poorly to current therapies and remains an incurable malignancy. Therefore, novel therapeutic targets and drugs are urgently needed for pancreatic cancer treatment. Selective induction of apoptosis in cancer cells is an appealing approach in cancer therapy. Apoptotic cell death is highly regulated by different signaling routes that involve a variety of subcellular organelles. Endoplasmic reticulum (ER) stress acts as a double-edged sword at the interface of cell survival and death. Pancreatic cells exhibit high hormone and enzyme secretory functions, and thereby show a highly developed ER. Thus, pancreatic cancer cells display a prominent ER. Solid tumors have to cope with adverse situations in which hypoxia, lack of certain nutrients, and the action of certain antitumor agents lead to a complex interplay and crosstalk between ER stress and autophagy-the latter acting as an adaptive survival response. ER stress also mediates cell death induced by a number of anticancer drugs and experimental conditions, highlighting the pivotal role of ER stress in modulating cell fate. The alkylphospholipid analog prototype edelfosine is selectively taken up by tumor cells, accumulates in the ER of a number of human solid tumor cells-including pancreatic cancer cells-and promotes apoptosis through a persistent ER-stress-mediated mechanism both in vitro and in vivo. Here, we discuss and propose that direct ER targeting may be a promising approach in the therapy of pancreatic cancer, opening up a new avenue for the treatment of this currently incurable and deadly cancer. Furthermore, because autophagy acts as a cytoprotective response to ER stress, potentiation of the triggering of a persistent ER response by combination therapy, together with the use of autophagy blockers, could improve the current gloomy expectations for finding a cure for this type of cancer.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, C/Ramiro de Maeztu 9, E-28040 Madrid, Spain;
| | | |
Collapse
|
12
|
Mollinedo F, Gajate C. Mitochondrial Targeting Involving Cholesterol-Rich Lipid Rafts in the Mechanism of Action of the Antitumor Ether Lipid and Alkylphospholipid Analog Edelfosine. Pharmaceutics 2021; 13:763. [PMID: 34065546 PMCID: PMC8161315 DOI: 10.3390/pharmaceutics13050763] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
The ether lipid edelfosine induces apoptosis selectively in tumor cells and is the prototypic molecule of a family of synthetic antitumor compounds collectively known as alkylphospholipid analogs. Cumulative evidence shows that edelfosine interacts with cholesterol-rich lipid rafts, endoplasmic reticulum (ER) and mitochondria. Edelfosine induces apoptosis in a number of hematological cancer cells by recruiting death receptors and downstream apoptotic signaling into lipid rafts, whereas it promotes apoptosis in solid tumor cells through an ER stress response. Edelfosine-induced apoptosis, mediated by lipid rafts and/or ER, requires the involvement of a mitochondrial-dependent step to eventually elicit cell death, leading to the loss of mitochondrial membrane potential, cytochrome c release and the triggering of cell death. The overexpression of Bcl-2 or Bcl-xL blocks edelfosine-induced apoptosis. Edelfosine induces the redistribution of lipid rafts from the plasma membrane to the mitochondria. The pro-apoptotic action of edelfosine on cancer cells is associated with the recruitment of F1FO-ATP synthase into cholesterol-rich lipid rafts. Specific inhibition of the FO sector of the F1FO-ATP synthase, which contains the membrane-embedded c-subunit ring that constitutes the mitochondrial permeability transcription pore, hinders edelfosine-induced cell death. Taking together, the evidence shown here suggests that the ether lipid edelfosine could modulate cell death in cancer cells by direct interaction with mitochondria, and the reorganization of raft-located mitochondrial proteins that critically modulate cell death or survival. Here, we summarize and discuss the involvement of mitochondria in the antitumor action of the ether lipid edelfosine, pointing out the mitochondrial targeting of this drug as a major therapeutic approach, which can be extrapolated to other alkylphospholipid analogs. We also discuss the involvement of cholesterol transport and cholesterol-rich lipid rafts in the interactions between the organelles as well as in the role of mitochondria in the regulation of apoptosis in cancer cells and cancer therapy.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, C/Ramiro de Maeztu 9, E-28040 Madrid, Spain;
| | | |
Collapse
|
13
|
Athanasopoulos A, André B, Sophianopoulou V, Gournas C. Fungal plasma membrane domains. FEMS Microbiol Rev 2020; 43:642-673. [PMID: 31504467 DOI: 10.1093/femsre/fuz022] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/25/2019] [Indexed: 12/11/2022] Open
Abstract
The plasma membrane (PM) performs a plethora of physiological processes, the coordination of which requires spatial and temporal organization into specialized domains of different sizes, stability, protein/lipid composition and overall architecture. Compartmentalization of the PM has been particularly well studied in the yeast Saccharomyces cerevisiae, where five non-overlapping domains have been described: The Membrane Compartments containing the arginine permease Can1 (MCC), the H+-ATPase Pma1 (MCP), the TORC2 kinase (MCT), the sterol transporters Ltc3/4 (MCL), and the cell wall stress mechanosensor Wsc1 (MCW). Additional cortical foci at the fungal PM are the sites where clathrin-dependent endocytosis occurs, the sites where the external pH sensing complex PAL/Rim localizes, and sterol-rich domains found in apically grown regions of fungal membranes. In this review, we summarize knowledge from several fungal species regarding the organization of the lateral PM segregation. We discuss the mechanisms of formation of these domains, and the mechanisms of partitioning of proteins there. Finally, we discuss the physiological roles of the best-known membrane compartments, including the regulation of membrane and cell wall homeostasis, apical growth of fungal cells and the newly emerging role of MCCs as starvation-protective membrane domains.
Collapse
Affiliation(s)
- Alexandros Athanasopoulos
- Microbial Molecular Genetics Laboratory, Institute of Biosciences and Applications, National Centre for Scientific Research 'Demokritos,' Patr. Grigoriou E & 27 Neapoleos St. 15341, Agia Paraskevi, Greece
| | - Bruno André
- Molecular Physiology of the Cell laboratory, Université Libre de Bruxelles (ULB), Institut de Biologie et de Médecine Moléculaires, rue des Pr Jeener et Brachet 12, 6041, Gosselies, Belgium
| | - Vicky Sophianopoulou
- Microbial Molecular Genetics Laboratory, Institute of Biosciences and Applications, National Centre for Scientific Research 'Demokritos,' Patr. Grigoriou E & 27 Neapoleos St. 15341, Agia Paraskevi, Greece
| | - Christos Gournas
- Microbial Molecular Genetics Laboratory, Institute of Biosciences and Applications, National Centre for Scientific Research 'Demokritos,' Patr. Grigoriou E & 27 Neapoleos St. 15341, Agia Paraskevi, Greece
| |
Collapse
|
14
|
Zaremberg V, Ganesan S, Mahadeo M. Lipids and Membrane Microdomains: The Glycerolipid and Alkylphosphocholine Class of Cancer Chemotherapeutic Drugs. Handb Exp Pharmacol 2020; 259:261-288. [PMID: 31302758 DOI: 10.1007/164_2019_222] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Synthetic antitumor lipids are metabolically stable lysophosphatidylcholine derivatives, encompassing a class of non-mutagenic drugs that selectively target cancerous cells. In this chapter we review the literature as relates to the clinical efficacy of these antitumor lipid drugs and how our understanding of their mode of action has evolved alongside key advances in our knowledge of membrane structure, organization, and function. First, the history of the development of this class of drugs is described, providing a summary of clinical outcomes of key members including edelfosine, miltefosine, perifosine, erufosine, and erucylphosphocholine. A detailed description of the biophysical properties of these drugs and specific drug-lipid interactions which may contribute to the selectivity of the antitumor lipids for cancer cells follows. An updated model on the mode of action of these lipid drugs as membrane disorganizing agents is presented. Membrane domain organization as opposed to targeting specific proteins on membranes is discussed. By altering membranes, these antitumor lipids inhibit many survival pathways while activating pro-apoptotic signals leading to cell demise.
Collapse
|
15
|
How Surrogate and Chemical Genetics in Model Organisms Can Suggest Therapies for Human Genetic Diseases. Genetics 2018; 208:833-851. [PMID: 29487144 PMCID: PMC5844338 DOI: 10.1534/genetics.117.300124] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/26/2017] [Indexed: 12/12/2022] Open
Abstract
Genetic diseases are both inherited and acquired. Many genetic diseases fall under the paradigm of orphan diseases, a disease found in < 1 in 2000 persons. With rapid and cost-effective genome sequencing becoming the norm, many causal mutations for genetic diseases are being rapidly determined. In this regard, model organisms are playing an important role in validating if specific mutations identified in patients drive the observed phenotype. An emerging challenge for model organism researchers is the application of genetic and chemical genetic platforms to discover drug targets and drugs/drug-like molecules for potential treatment options for patients with genetic disease. This review provides an overview of how model organisms have contributed to our understanding of genetic disease, with a focus on the roles of yeast and zebrafish in gene discovery and the identification of compounds that could potentially treat human genetic diseases.
Collapse
|
16
|
Bello C, Bai J, Zambron BK, Elías-Rodríguez P, Gajate C, Robina I, Caffa I, Cea M, Montecucco F, Nencioni A, Nahimana A, Aubry D, Breton C, Duchosal MA, Mollinedo F, Vogel P. Induction of cell killing and autophagy by amphiphilic pyrrolidine derivatives on human pancreatic cancer cells. Eur J Med Chem 2018; 150:457-478. [PMID: 29547833 DOI: 10.1016/j.ejmech.2018.02.086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/19/2018] [Accepted: 02/27/2018] [Indexed: 12/15/2022]
|
17
|
Yeast Cells Exposed to Exogenous Palmitoleic Acid Either Adapt to Stress and Survive or Commit to Regulated Liponecrosis and Die. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3074769. [PMID: 29636840 PMCID: PMC5831759 DOI: 10.1155/2018/3074769] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 11/27/2017] [Accepted: 12/20/2017] [Indexed: 12/11/2022]
Abstract
A disturbed homeostasis of cellular lipids and the resulting lipotoxicity are considered to be key contributors to many human pathologies, including obesity, metabolic syndrome, type 2 diabetes, cardiovascular diseases, and cancer. The yeast Saccharomyces cerevisiae has been successfully used for uncovering molecular mechanisms through which impaired lipid metabolism causes lipotoxicity and elicits different forms of regulated cell death. Here, we discuss mechanisms of the “liponecrotic” mode of regulated cell death in S. cerevisiae. This mode of regulated cell death can be initiated in response to a brief treatment of yeast with exogenous palmitoleic acid. Such treatment prompts the incorporation of exogenously added palmitoleic acid into phospholipids and neutral lipids. This orchestrates a global remodeling of lipid metabolism and transfer in the endoplasmic reticulum, mitochondria, lipid droplets, and the plasma membrane. Certain features of such remodeling play essential roles either in committing yeast to liponecrosis or in executing this mode of regulated cell death. We also outline four processes through which yeast cells actively resist liponecrosis by adapting to the cellular stress imposed by palmitoleic acid and maintaining viability. These prosurvival cellular processes are confined in the endoplasmic reticulum, lipid droplets, peroxisomes, autophagosomes, vacuoles, and the cytosol.
Collapse
|
18
|
Ansari SS, Sharma AK, Zepp M, Ivanova E, Bergmann F, König R, Berger MR. Upregulation of cell cycle genes in head and neck cancer patients may be antagonized by erufosine's down regulation of cell cycle processes in OSCC cells. Oncotarget 2017; 9:5797-5810. [PMID: 29464035 PMCID: PMC5814175 DOI: 10.18632/oncotarget.23537] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023] Open
Abstract
The TCGA database was analyzed to identify deregulation of cell cycle genes across 24 cancer types and ensuing effects on patient survival. Pan-cancer analysis showed that head and neck squamous cell carcinoma (HNSCC) ranks amongst the top four cancers showing deregulated cell cycle genes. Also, the median gene expression of all CDKs and cyclins in HNSCC patient samples was higher than that of the global gene expression. This was verified by IHC staining of CCND1 from HNSCC patients. When evaluating the quartiles with highest and lowest expression, increased CCND1/CDK6 levels had negative implication on patient survival. In search for a drug, which may antagonize this tumor profile, the potential of the alkylphosphocholine erufosine was evaluated against cell lines of the HNSCC subtype, oral squamous cell carcinoma (OSCC) using in-vitro and in-vivo assays. Erufosine inhibited growth of OSCC cell lines concentration dependently. Initial microarray findings revealed that cyclins and CDKs were down-regulated concentration dependently upon exposure to erufosine and participated in negative enrichment of cell cycle processes. These findings, indicating a pan-cdk/cyclin inhibition by erufosine, were verified at both, mRNA and protein levels. Erufosine caused a G2/M block and inhibition of colony formation. Significant tumor growth retardation was seen upon treatment with erufosine in a xenograft model. For the decreased cyclin D1 and CDK 4/6 levels found in tumor tissue, these proteins can serve as biomarker for erufosine intervention. The findings demonstrate the potential of erufosine as cell cycle inhibitor in HNSCC treatment, alone or in combination with current therapeutic agents.
Collapse
Affiliation(s)
- Shariq S Ansari
- Toxicology and Chemotherapy Unit, German Cancer Research Center, Heidelberg, Germany
| | - Ashwini K Sharma
- Institute for Pharmacy and Molecular Biotechnology (IPMB) and BioQuant, Heidelberg University, Heidelberg, Germany.,Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Zepp
- Toxicology and Chemotherapy Unit, German Cancer Research Center, Heidelberg, Germany
| | - Elizabet Ivanova
- Laboratory for Experimental Chemotherapy, Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Bulgaria
| | - Frank Bergmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Rainer König
- Integrated Research and Treatment Center Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Network Modeling, Leibniz Institute for Natural Products Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Martin R Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
19
|
Tambellini NP, Zaremberg V, Krishnaiah S, Turner RJ, Weljie AM. Primary Metabolism and Medium-Chain Fatty Acid Alterations Precede Long-Chain Fatty Acid Changes Impacting Neutral Lipid Metabolism in Response to an Anticancer Lysophosphatidylcholine Analogue in Yeast. J Proteome Res 2017; 16:3741-3752. [PMID: 28849941 DOI: 10.1021/acs.jproteome.7b00430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The nonmetabolizable lysophosphatidylcholine (LysoPC) analogue edelfosine is the prototype of a class of compounds being investigated for their potential as selective chemotherapeutic agents. Edelfosine targets membranes, disturbing cellular homeostasis. Is not clear at this point how membrane alterations are communicated between intracellular compartments leading to growth inhibition and eventual cell death. In the present study, a combined metabolomics/lipidomics approach for the unbiased identification of metabolic pathways altered in yeast treated with sublethal concentrations of the LysoPC analogue was employed. Mass spectrometry of polar metabolites, fatty acids, and lipidomic profiling was used to study the effects of edelfosine on yeast metabolism. Amino acid and sugar metabolism, the Krebs cycle, and fatty acid profiles were most disrupted, with polar metabolites and short-medium chain fatty acid changes preceding long and very long-chain fatty acid variations. Initial increases in metabolites such as trehalose, proline, and γ-amino butyric acid with a concomitant decrease in metabolites of the Krebs cycle, citrate and fumarate, are interpreted as a cellular attempt to offset oxidative stress in response to mitochondrial dysfunction induced by the treatment. Notably, alanine, inositol, and myristoleic acid showed a steady increase during the period analyzed (2, 4, and 6 h after treatment). Of importance was the finding that edelfosine induced significant alterations in neutral glycerolipid metabolism resulting in a significant increase in the signaling lipid diacylglycerol.
Collapse
Affiliation(s)
- Nicolas P Tambellini
- Department of Biological Sciences, University of Calgary , Calgary, Alberta T2N 1N4, Canada.,Metabolomics Research Centre, University of Calgary , Calgary, Alberta T2N 1N4, Canada
| | - Vanina Zaremberg
- Department of Biological Sciences, University of Calgary , Calgary, Alberta T2N 1N4, Canada
| | - Saikumari Krishnaiah
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania 19104-5158, United States of America
| | - Raymond J Turner
- Department of Biological Sciences, University of Calgary , Calgary, Alberta T2N 1N4, Canada
| | - Aalim M Weljie
- Department of Biological Sciences, University of Calgary , Calgary, Alberta T2N 1N4, Canada.,Metabolomics Research Centre, University of Calgary , Calgary, Alberta T2N 1N4, Canada.,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania 19104-5158, United States of America
| |
Collapse
|
20
|
Cheng B, Morales LD, Zhang Y, Mito S, Tsin A. Niclosamide induces protein ubiquitination and inhibits multiple pro-survival signaling pathways in the human glioblastoma U-87 MG cell line. PLoS One 2017; 12:e0184324. [PMID: 28877265 PMCID: PMC5587337 DOI: 10.1371/journal.pone.0184324] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/22/2017] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma is the most common and lethal malignant primary brain tumor for which the development of efficacious chemotherapeutic agents remains an urgent need. The anti-helminthic drug niclosamide, which has long been in use to treat tapeworm infections, has recently attracted renewed interest due to its apparent anticancer effects in a variety of in vitro and in vivo cancer models. However, the mechanism(s) of action remains to be elucidated. In the present study, we found that niclosamide induced cell toxicity in human glioblastoma cells corresponding with increased protein ubiquitination, ER stress and autophagy. In addition, niclosamide treatment led to down-regulation of Wnt/β-catenin, PI3K/AKT, MAPK/ERK, and STAT3 pro-survival signal transduction pathways to further reduce U-87 MG cell viability. Taken together, these results provide new insights into the glioblastoma suppressive capabilities of niclosamide, showing that niclosamide can target multiple major cell signaling pathways simultaneously to effectively promote cell death in U-87 MG cells. Niclosamide constitutes a new prospect for a therapeutic treatment against human glioblastoma.
Collapse
Affiliation(s)
- Benxu Cheng
- Department of Biomedical Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
- * E-mail:
| | - Liza Doreen Morales
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
| | - Yonghong Zhang
- Department of Chemistry, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
| | - Shizue Mito
- Department of Chemistry, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
| | - Andrew Tsin
- Department of Biomedical Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas, United States of America
| |
Collapse
|
21
|
Alkyl ether lipids, ion channels and lipid raft reorganization in cancer therapy. Pharmacol Ther 2016; 165:114-31. [DOI: 10.1016/j.pharmthera.2016.06.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 05/26/2016] [Indexed: 12/21/2022]
|
22
|
Yeast ABC transporters in lipid trafficking. Fungal Genet Biol 2016; 93:25-34. [DOI: 10.1016/j.fgb.2016.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/28/2016] [Accepted: 05/31/2016] [Indexed: 12/31/2022]
|
23
|
Goldgof GM, Durrant JD, Ottilie S, Vigil E, Allen KE, Gunawan F, Kostylev M, Henderson KA, Yang J, Schenken J, LaMonte GM, Manary MJ, Murao A, Nachon M, Murray R, Prescott M, McNamara CW, Slayman CW, Amaro RE, Suzuki Y, Winzeler EA. Comparative chemical genomics reveal that the spiroindolone antimalarial KAE609 (Cipargamin) is a P-type ATPase inhibitor. Sci Rep 2016; 6:27806. [PMID: 27291296 PMCID: PMC4904242 DOI: 10.1038/srep27806] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/20/2016] [Indexed: 11/30/2022] Open
Abstract
The spiroindolones, a new class of antimalarial medicines discovered in a cellular screen, are rendered less active by mutations in a parasite P-type ATPase, PfATP4. We show here that S. cerevisiae also acquires mutations in a gene encoding a P-type ATPase (ScPMA1) after exposure to spiroindolones and that these mutations are sufficient for resistance. KAE609 resistance mutations in ScPMA1 do not confer resistance to unrelated antimicrobials, but do confer cross sensitivity to the alkyl-lysophospholipid edelfosine, which is known to displace ScPma1p from the plasma membrane. Using an in vitro cell-free assay, we demonstrate that KAE609 directly inhibits ScPma1p ATPase activity. KAE609 also increases cytoplasmic hydrogen ion concentrations in yeast cells. Computer docking into a ScPma1p homology model identifies a binding mode that supports genetic resistance determinants and in vitro experimental structure-activity relationships in both P. falciparum and S. cerevisiae. This model also suggests a shared binding site with the dihydroisoquinolones antimalarials. Our data support a model in which KAE609 exerts its antimalarial activity by directly interfering with P-type ATPase activity.
Collapse
Affiliation(s)
- Gregory M. Goldgof
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
- Department of Synthetic Biology and Bioenergy, J. Craig Venter
Institute, La Jolla, California, USA
| | - Jacob D. Durrant
- Department of Chemistry & Biochemistry and the National
Biomedical Computation Resource, University of California, San
Diego, La Jolla, California, USA
| | - Sabine Ottilie
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Edgar Vigil
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Kenneth E. Allen
- Department of Genetics, Yale University School of
Medicine, New Haven, Connecticut, USA
| | - Felicia Gunawan
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Maxim Kostylev
- Department of Synthetic Biology and Bioenergy, J. Craig Venter
Institute, La Jolla, California, USA
| | | | - Jennifer Yang
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Jake Schenken
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Gregory M. LaMonte
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Micah J. Manary
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Ayako Murao
- Department of Synthetic Biology and Bioenergy, J. Craig Venter
Institute, La Jolla, California, USA
| | - Marie Nachon
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Rebecca Murray
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Maximo Prescott
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| | - Case W. McNamara
- Genomics Institute of the Novartis Research Foundation,
San Diego, California, USA
| | - Carolyn W. Slayman
- Department of Genetics, Yale University School of
Medicine, New Haven, Connecticut, USA
| | - Rommie E. Amaro
- Department of Chemistry & Biochemistry and the National
Biomedical Computation Resource, University of California, San
Diego, La Jolla, California, USA
| | - Yo Suzuki
- Department of Synthetic Biology and Bioenergy, J. Craig Venter
Institute, La Jolla, California, USA
| | - Elizabeth A. Winzeler
- Division of Pharmacology and Drug Discovery, Department of
Pediatrics, University of California, San Diego, School of Medicine,
La Jolla, California, USA
| |
Collapse
|
24
|
Udayakumar TS, Stoyanova R, Shareef MM, Mu Z, Philip S, Burnstein KL, Pollack A. Edelfosine Promotes Apoptosis in Androgen-Deprived Prostate Tumors by Increasing ATF3 and Inhibiting Androgen Receptor Activity. Mol Cancer Ther 2016; 15:1353-63. [PMID: 26944919 DOI: 10.1158/1535-7163.mct-15-0332] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 02/15/2016] [Indexed: 12/31/2022]
Abstract
Edelfosine is a synthetic alkyl-lysophospholipid that possesses significant antitumor activity in several human tumor models. Here, we investigated the effects of edelfosine combined with androgen deprivation (AD) in LNCaP and VCaP human prostate cancer cells. This treatment regimen greatly decreased cell proliferation compared with single agent or AD alone, resulting in higher levels of apoptosis in LNCaP compared with VCaP cells. Edelfosine caused a dose-dependent decrease in AKT activity, but did not affect the expression of total AKT in either cell line. Furthermore, edelfosine treatment inhibited the expression of androgen receptor (AR) and was associated with an increase in activating transcription factor 3 (ATF3) expression levels, a stress response gene and a negative regulator of AR transactivation. ATF3 binds to AR after edelfosine + AD and represses the transcriptional activation of AR as demonstrated by PSA promoter studies. Knockdown of ATF3 using siRNA-ATF3 reversed the inhibition of PSA promoter activity, suggesting that the growth inhibition effect of edelfosine was ATF3 dependent. Moreover, expression of AR variant 7 (ARv7) and TMPRSS2-ERG fusion gene were greatly inhibited after combined treatment with AD and edelfosine in VCaP cells. In vivo experiments using an orthotopic LNCaP model confirmed the antitumor effects of edelfosine + AD over the individual treatments. A significant decrease in tumor volume and PSA levels was observed when edelfosine and AD were combined, compared with edelfosine alone. Edelfosine shows promise in combination with AD for the treatment of prostate cancer patients. Mol Cancer Ther; 15(6); 1353-63. ©2016 AACR.
Collapse
Affiliation(s)
- Thirupandiyur S Udayakumar
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Radka Stoyanova
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Mohammed M Shareef
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Zhaomei Mu
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sakhi Philip
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Kerry L Burnstein
- Department of Molecular and Cellular Pharmacology, University of Miami, Miami, Florida
| | - Alan Pollack
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.
| |
Collapse
|
25
|
Mahadeo M, Nathoo S, Ganesan S, Driedger M, Zaremberg V, Prenner EJ. Disruption of lipid domain organization in monolayers of complex yeast lipid extracts induced by the lysophosphatidylcholine analogue edelfosine in vivo. Chem Phys Lipids 2015; 191:153-62. [PMID: 26386399 DOI: 10.1016/j.chemphyslip.2015.09.004] [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: 06/16/2015] [Revised: 09/08/2015] [Accepted: 09/15/2015] [Indexed: 10/23/2022]
Abstract
The lysophosphatidylcholine analogue edelfosine is a potent antitumor and antiparasitic drug that targets cell membranes. Previous studies have shown that edelfosine alters membrane domain organization inducing internalization of sterols and endocytosis of plasma membrane transporters. These early events affect signaling pathways that result in cell death. It has been shown that edelfosine preferentially partitions into more rigid lipid domains in mammalian as well as in yeast cells. In this work we aimed at investigating the effect of edelfosine on membrane domain organization using monolayers prepared from whole cell lipid extracts of cells treated with edelfosine compared to control conditions. In Langmuir monolayers we were able to detect important differences to the lipid packing of the membrane monofilm. Domain formation visualized by means of Brewster angle microscopy also showed major morphological changes between edelfosine treated versus control samples. Importantly, edelfosine resistant cells defective in drug uptake did not display the same differences. In addition, co-spread samples of control lipid extracts with edelfosine added post extraction did not fully mimic the results obtained with lipid extracts from treated cells. Altogether these results indicate that edelfosine induces changes in membrane domain organization and that these changes depend on drug uptake. Our work also validates the use of monolayers derived from complex cell lipid extracts combined with Brewster angle microscopy, as a sensitive approach to distinguish between conditions associated with susceptibility or resistance to lysophosphatidylcholine analogues.
Collapse
Affiliation(s)
- Mark Mahadeo
- Department of Biological Sciences, Faculty of Science, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Safia Nathoo
- Department of Biological Sciences, Faculty of Science, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Suriakarthiga Ganesan
- Department of Biological Sciences, Faculty of Science, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Michael Driedger
- Department of Biological Sciences, Faculty of Science, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Vanina Zaremberg
- Department of Biological Sciences, Faculty of Science, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada.
| | - Elmar J Prenner
- Department of Biological Sciences, Faculty of Science, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
26
|
Sánchez-Blanco A, Rodríguez-Matellán AG, Reis-Sobreiro M, Sáenz-Narciso B, Cabello J, Mohler WA, Mollinedo F. Caenorhabditis elegans as a platform to study the mechanism of action of synthetic antitumor lipids. Cell Cycle 2015; 13:3375-89. [PMID: 25485582 DOI: 10.4161/15384101.2014.952183] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Drugs capable of specifically recognizing and killing cancer cells while sparing healthy cells are of great interest in anti-cancer therapy. An example of such a drug is edelfosine, the prototype molecule of a family of synthetic lipids collectively known as antitumor lipids (ATLs). A better understanding of the selectivity and the mechanism of action of these compounds would lead to better anticancer treatments. Using Caenorhabditis elegans, we modeled key features of the ATL selectivity against cancer cells. Edelfosine induced a selective and direct killing action on C. elegans embryos, which was dependent on cholesterol, without affecting adult worms and larvae. Distinct ATLs ranked differently in their embryonic lethal effect with edelfosine > perifosine > erucylphosphocholine >> miltefosine. Following a biased screening of 57 C. elegans mutants we found that inactivation of components of the insulin/IGF-1 signaling pathway led to resistance against the ATL edelfosine in both C. elegans and human tumor cells. This paper shows that C. elegans can be used as a rapid platform to facilitate ATL research and to further understand the mechanism of action of edelfosine and other synthetic ATLs.
Collapse
Affiliation(s)
- Adolfo Sánchez-Blanco
- a Instituto de Biología Molecular y Celular del Cáncer ; Centro de Investigación del Cáncer ; CSIC-Universidad de Salamanca ; Campus Miguel de Unamuno ; Salamanca , Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
Mollinedo F, Gajate C. Lipid rafts as major platforms for signaling regulation in cancer. Adv Biol Regul 2015; 57:130-146. [PMID: 25465296 DOI: 10.1016/j.jbior.2014.10.003] [Citation(s) in RCA: 232] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 10/10/2014] [Accepted: 10/11/2014] [Indexed: 06/04/2023]
Abstract
Cell signaling does not apparently occur randomly over the cell surface, but it seems to be integrated very often into cholesterol-rich membrane domains, termed lipid rafts. Membrane lipid rafts are highly ordered membrane domains that are enriched in cholesterol, sphingolipids and gangliosides, and behave as major modulators of membrane geometry, lateral movement of molecules, traffic and signal transduction. Because the lipid and protein composition of membrane rafts differs from that of the surrounding membrane, they provide an additional level of compartmentalization, serving as sorting platforms and hubs for signal transduction proteins. A wide number of signal transduction processes related to cell adhesion, migration, as well as to cell survival and proliferation, which play major roles in cancer development and progression, are dependent on lipid rafts. Despite lipid rafts harbor mainly critical survival signaling pathways, including insulin-like growth factor I (IGF-I)/phosphatidylinositol 3-kinase (PI3K)/Akt signaling, recent evidence suggests that these membrane domains can also house death receptor-mediated apoptotic signaling. Recruitment of this death receptor signaling pathway in membrane rafts can be pharmacologically modulated, thus opening up the possibility to regulate cell demise with a therapeutic use. The synthetic ether phospholipid edelfosine shows a high affinity for cholesterol and accumulates in lipid rafts in a number of malignant hematological cells, leading to an efficient in vitro and in vivo antitumor activity by inducing translocation of death receptors and downstream signaling molecules to these membrane domains. Additional antitumor drugs have also been shown to act, at least in part, by recruiting death receptors in lipid rafts. The partition of death receptors together with downstream apoptotic signaling molecules in membrane rafts has led us to postulate the concept of a special liquid-ordered membrane platform coined as "cluster of apoptotic signaling molecule-enriched rafts" (CASMER), referring to raft platforms enriched in apoptotic molecules. CASMERs act as scaffolds for apoptosis signaling compartmentalization, facilitating and stabilizing protein-protein interactions by local assembly of cross-interacting molecules, which leads to apoptosis amplification and a decrease in apoptotic signal threshold. Edelfosine also displaced survival PI3K/Akt signaling from lipid rafts, leading to Akt inhibition, in mantle cell lymphoma cells. Thus, membrane rafts could act as scaffold structures where segregation of pro- from anti-apoptotic molecules could take place. In this review, we summarize our view of how reorganization of the protein composition of lipid raft membrane domains regulates cell death and therefore it might be envisaged as a novel target in the treatment of cancer.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, E-37007 Salamanca, Spain.
| | - Consuelo Gajate
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, E-37007 Salamanca, Spain.
| |
Collapse
|
28
|
Walker ME, Nguyen TD, Liccioli T, Schmid F, Kalatzis N, Sundstrom JF, Gardner JM, Jiranek V. Genome-wide identification of the Fermentome; genes required for successful and timely completion of wine-like fermentation by Saccharomyces cerevisiae. BMC Genomics 2014. [DOI: 10.1186/1471-2164-15-552 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
29
|
Walker ME, Nguyen TD, Liccioli T, Schmid F, Kalatzis N, Sundstrom JF, Gardner JM, Jiranek V. Genome-wide identification of the Fermentome; genes required for successful and timely completion of wine-like fermentation by Saccharomyces cerevisiae. BMC Genomics 2014. [DOI: 10.1186/1471-2164-15-552 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
30
|
Walker ME, Nguyen TD, Liccioli T, Schmid F, Kalatzis N, Sundstrom JF, Gardner JM, Jiranek V. Genome-wide identification of the Fermentome; genes required for successful and timely completion of wine-like fermentation by Saccharomyces cerevisiae. BMC Genomics 2014. [DOI: 10.1186/1471-2164-15-552 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
31
|
Walker ME, Nguyen TD, Liccioli T, Schmid F, Kalatzis N, Sundstrom JF, Gardner JM, Jiranek V. Genome-wide identification of the Fermentome; genes required for successful and timely completion of wine-like fermentation by Saccharomyces cerevisiae. BMC Genomics 2014. [DOI: 10.1186/1471-2164-15-552 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
32
|
Walker ME, Nguyen TD, Liccioli T, Schmid F, Kalatzis N, Sundstrom JF, Gardner JM, Jiranek V. Genome-wide identification of the Fermentome; genes required for successful and timely completion of wine-like fermentation by Saccharomyces cerevisiae. BMC Genomics 2014. [DOI: 10.1186/1471-2164-15-552 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
33
|
Walker ME, Nguyen TD, Liccioli T, Schmid F, Kalatzis N, Sundstrom JF, Gardner JM, Jiranek V. Genome-wide identification of the Fermentome; genes required for successful and timely completion of wine-like fermentation by Saccharomyces cerevisiae. BMC Genomics 2014. [DOI: 10.1186/1471-2164-15-552 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
34
|
Walker ME, Nguyen TD, Liccioli T, Schmid F, Kalatzis N, Sundstrom JF, Gardner JM, Jiranek V. Genome-wide identification of the Fermentome; genes required for successful and timely completion of wine-like fermentation by Saccharomyces cerevisiae. BMC Genomics 2014. [DOI: 10.1186/1471-2164-15-552 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
35
|
Walker ME, Nguyen TD, Liccioli T, Schmid F, Kalatzis N, Sundstrom JF, Gardner JM, Jiranek V. Genome-wide identification of the Fermentome; genes required for successful and timely completion of wine-like fermentation by Saccharomyces cerevisiae. BMC Genomics 2014; 15:552. [PMID: 24993029 PMCID: PMC4099481 DOI: 10.1186/1471-2164-15-552] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 06/27/2014] [Indexed: 12/21/2022] Open
Abstract
Background Wine fermentation is a harsh ecological niche to which wine yeast are well adapted. The initial high osmotic pressure and acidity of grape juice is followed by nutrient depletion and increasing concentrations of ethanol as the fermentation progresses. Yeast’s adaptation to these and many other environmental stresses, enables successful completion of high-sugar fermentations. Earlier transcriptomic and growth studies have tentatively identified genes important for high-sugar fermentation. Whilst useful, such studies did not consider extended growth (>5 days) in a temporally dynamic multi-stressor environment such as that found in many industrial fermentation processes. Here, we identify genes whose deletion has minimal or no effect on growth, but results in failure to achieve timely completion of the fermentation of a chemically defined grape juice with 200 g L−1 total sugar. Results Micro- and laboratory-scale experimental fermentations were conducted to identify 72 clones from ~5,100 homozygous diploid single-gene yeast deletants, which exhibited protracted fermentation in a high-sugar medium. Another 21 clones (related by gene function, but initially eliminated from the screen because of possible growth defects) were also included. Clustering and numerical enrichment of genes annotated to specific Gene Ontology (GO) terms highlighted the vacuole’s role in ion homeostasis and pH regulation, through vacuole acidification. Conclusion We have identified 93 genes whose deletion resulted in the duration of fermentation being at least 20% longer than the wild type. An extreme phenotype, ‘stuck’ fermentation, was also observed when DOA4, NPT1, PLC1, PTK2, SIN3, SSQ1, TPS1, TPS2 or ZAP1 were deleted. These 93 Fermentation Essential Genes (FEG) are required to complete an extended high-sugar (wine-like) fermentation. Their importance is highlighted in our Fermentation Relevant Yeast Genes (FRYG) database, generated from literature and the fermentation-relevant phenotypic characteristics of null mutants described in the Saccharomyces Genome Database. The 93-gene set is collectively referred to as the ‘Fermentome’. The fact that 10 genes highlighted in this study have not previously been linked to fermentation-related stresses, supports our experimental rationale. These findings, together with investigations of the genetic diversity of industrial strains, are crucial for understanding the mechanisms behind yeast’s response and adaptation to stresses imposed during high-sugar fermentations. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-552) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Vladimir Jiranek
- School of Agriculture, Food and Wine, University of Adelaide, Urrbrae, SA 5064, Australia.
| |
Collapse
|
36
|
Yucel EB, Eraslan S, Ulgen KO. The impact of medium acidity on the chronological life span ofSaccharomyces cerevisiae - lipids, signaling cascades, mitochondrial and vacuolar functions. FEBS J 2014; 281:1281-303. [DOI: 10.1111/febs.12705] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 12/20/2013] [Accepted: 12/23/2013] [Indexed: 12/18/2022]
Affiliation(s)
- Esra B. Yucel
- Department of Chemical Engineering; Boğaziçi University; Istanbul Turkey
| | - Serpil Eraslan
- Department of Chemical Engineering; Boğaziçi University; Istanbul Turkey
| | - Kutlu O. Ulgen
- Department of Chemical Engineering; Boğaziçi University; Istanbul Turkey
| |
Collapse
|
37
|
Eisenberg T, Büttner S. Lipids and cell death in yeast. FEMS Yeast Res 2013; 14:179-97. [PMID: 24119111 PMCID: PMC4255311 DOI: 10.1111/1567-1364.12105] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 08/21/2013] [Accepted: 09/25/2013] [Indexed: 01/22/2023] Open
Abstract
Understanding lipid-induced malfunction represents a major challenge of today's biomedical research. The connection of lipids to cellular and organ dysfunction, cell death, and disease (often referred to as lipotoxicity) is more complex than the sole lipotoxic effects of excess free fatty acids and requires genetically tractable model systems for mechanistic investigation. We herein summarize recent advances in the field of lipid-induced toxicity that employ the established model system for cell death and aging research of budding yeast Saccharomyces cerevisiae. Studies in yeast have shed light on various aspects of lipotoxicity, including free fatty acid toxicity, sphingolipid-modulated cell death as well as the involvement of cardiolipin and lipid peroxidation in the mitochondrial pathways of apoptosis. Regimens used range from exogenously applied lipids, genetic modulation of lipolysis and triacylglyceride synthesis, variations in sphingolipid/ceramide metabolism as well as changes in peroxisome function by either genetic or pharmacological means. In future, the yeast model of programmed cell death will further contribute to the clarification of crucial questions of lipid-associated malfunction.
Collapse
Affiliation(s)
- Tobias Eisenberg
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | |
Collapse
|
38
|
Cuesta-Marbán Á, Botet J, Czyz O, Cacharro LM, Gajate C, Hornillos V, Delgado J, Zhang H, Amat-Guerri F, Acuña AU, McMaster CR, Revuelta JL, Zaremberg V, Mollinedo F. Drug uptake, lipid rafts, and vesicle trafficking modulate resistance to an anticancer lysophosphatidylcholine analogue in yeast. J Biol Chem 2013; 288:8405-8418. [PMID: 23335509 DOI: 10.1074/jbc.m112.425769] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The ether-phospholipid edelfosine, a prototype antitumor lipid (ATL), kills yeast cells and selectively kills several cancer cell types. To gain insight into its mechanism of action, we performed chemogenomic screens in the Saccharomyces cerevisiae gene-deletion strain collection, identifying edelfosine-resistant mutants. LEM3, AGP2, and DOC1 genes were required for drug uptake. Edelfosine displaced the essential proton pump Pma1p from rafts, inducing its internalization into the vacuole. Additional ATLs, including miltefosine and perifosine, also displaced Pma1p from rafts to the vacuole, suggesting that this process is a major hallmark of ATL cytotoxicity in yeast. Radioactive and synthetic fluorescent edelfosine analogues accumulated in yeast plasma membrane rafts and subsequently the endoplasmic reticulum. Although both edelfosine and Pma1p were initially located at membrane rafts, internalization of the drug toward endoplasmic reticulum and Pma1p to the vacuole followed different routes. Drug internalization was not dependent on endocytosis and was not critical for yeast cytotoxicity. However, mutants affecting endocytosis, vesicle sorting, or trafficking to the vacuole, including the retromer and ESCRT complexes, prevented Pma1p internalization and were edelfosine-resistant. Our data suggest that edelfosine-induced cytotoxicity involves raft reorganization and retromer- and ESCRT-mediated vesicular transport and degradation of essential raft proteins leading to cell death. Cytotoxicity of ATLs is mainly dependent on the changes they induce in plasma membrane raft-located proteins that lead to their internalization and subsequent degradation. Edelfosine toxicity can be circumvented by inactivating genes that then result in the recycling of internalized cell-surface proteins back to the plasma membrane.
Collapse
Affiliation(s)
- Álvaro Cuesta-Marbán
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Javier Botet
- Departamento de Microbiología y Genética, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Ola Czyz
- Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Luis M Cacharro
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Consuelo Gajate
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Valentín Hornillos
- Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas, Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Javier Delgado
- Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas, Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Hui Zhang
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Francisco Amat-Guerri
- Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas, Juan de la Cierva 3, E-28006 Madrid, Spain
| | - A Ulises Acuña
- Instituto de Química Física Rocasolano, Consejo Superior de Investigaciones Científicas, Serrano 119, E-28006 Madrid, Spain
| | - Christopher R McMaster
- Department of Pharmacology, Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - José Luis Revuelta
- Departamento de Microbiología y Genética, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Vanina Zaremberg
- Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada.
| | - Faustino Mollinedo
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain.
| |
Collapse
|