1
|
Çetinel ZÖ, Bilge D. Investigation of miltefosine-model membranes interactions at the molecular level for two different PS levels modeling cancer cells. J Bioenerg Biomembr 2024; 56:461-473. [PMID: 38833041 PMCID: PMC11217121 DOI: 10.1007/s10863-024-10025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/26/2024] [Indexed: 06/06/2024]
Abstract
Miltefosine (MLT) is a broad-spectrum drug included in the alkylphospholipids (APL) used against leishmania and various types of cancer. The most crucial feature of APLs is that they are thought to only kill cancerous cells without harming normal cells. However, the molecular mechanism of action of APLs is not completely understood. The increase in the phosphatidylserine (PS) ratio is a marker showing the stage of cancer and even metastasis. The goal of this research was to investigate the molecular effects of miltefosine at the molecular level in different PS ratios. The effects of MLT on membrane phase transition, membrane orders, and dynamics were studied using DPPC/DPPS (3:1) and DPPC/DPPS (1:1) multilayer (MLV) vesicles mimicking DPPS ratio variation, Differential Scanning Calorimetry (DSC), and Fourier Transform Infrared spectroscopy (FTIR). Our findings indicate that miltefosine is evidence at the molecular level that it is directed towards the tumor cell and that the drug's effect increases with the increase of anionic lipids in the membrane depending on the stage of cancer.
Collapse
Affiliation(s)
| | - Duygu Bilge
- Department of Physics, Faculty of Science, Ege University, Bornova, Izmir, 35100, Turkey.
| |
Collapse
|
2
|
Torrens-Mas M, Collado-Solé A, Sola-Leyva A, Carrasco-Jiménez MP, Oliver J, Pons DG, Roca P, Sastre-Serra J. Mitochondrial Functionality Is Regulated by Alkylphospholipids in Human Colon Cancer Cells. BIOLOGY 2023; 12:1457. [PMID: 38132283 PMCID: PMC10740929 DOI: 10.3390/biology12121457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023]
Abstract
Alkylphospholipids (APLs) have been studied as anticancer drugs that interfere with biological membranes without targeting DNA. Although their mechanism of action is not fully elucidated yet, it is known that they disrupt the intracellular trafficking of cholesterol and its metabolism. Here, we analyzed whether APLs could also interfere with mitochondrial function. For this purpose, we used HT29 colorectal cancer cells, derived from a primary tumor, and SW620 colorectal cancer cells, derived from a metastasis site. After treatment with the APLs miltefosine and perifosine, we analyzed various mitochondrial parameters, including mitochondrial mass, cardiolipin content, mitochondrial membrane potential, H2O2 production, the levels of oxidative phosphorylation (OXPHOS) complexes, metabolic enzymes activity, the oxygen consumption rate, and the levels of apoptosis and autophagy markers. APLs, especially perifosine, increased mitochondrial mass while OXPHOS complexes levels were decreased without affecting the total oxygen consumption rate. Additionally, we observed an increase in pyruvate dehydrogenase (PDH) and isocitrate dehydrogenase (IDH) levels and a decrease in lactate dehydrogenase (LDH) activity, suggesting a metabolic rewiring induced by perifosine. These alterations led to higher mitochondrial membrane potential, which was potentiated by decreased uncoupling protein 2 (UCP2) levels and increased reactive oxygen species (ROS) production. Consequently, perifosine induced an imbalance in mitochondrial function, resulting in higher ROS production that ultimately impacted cellular viability.
Collapse
Affiliation(s)
- Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
| | - Alejandro Collado-Solé
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
| | - Alberto Sola-Leyva
- Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, University of Granada, Av. Fuentenueva s/n, 18001 Granada, Spain; (A.S.-L.); (M.P.C.-J.)
| | - María Paz Carrasco-Jiménez
- Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, University of Granada, Av. Fuentenueva s/n, 18001 Granada, Spain; (A.S.-L.); (M.P.C.-J.)
| | - Jordi Oliver
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Gabriel Pons
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Jorge Sastre-Serra
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
3
|
Spadari CDC, Borba-Santos LP, Rozental S, Ishida K. Miltefosine repositioning: A review of potential alternative antifungal therapy. J Mycol Med 2023; 33:101436. [PMID: 37774486 DOI: 10.1016/j.mycmed.2023.101436] [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: 05/24/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Fungal infections are a global health problem with high mortality and morbidity rates. Available antifungal agents have high toxicity and pharmacodynamic and pharmacokinetic limitations. Moreover, the increased incidence of antifungal-resistant isolates and the emergence of intrinsically resistant species raise concerns about seeking alternatives for efficient antifungal therapy. In this context, we review literature data addressing the potential action of miltefosine (MFS), an anti-Leishmania and anticancer agent, as a repositioning drug for antifungal treatment. Here, we highlight the in vitro and in vivo data, MFS possible mechanisms of action, case reports, and nanocarrier-mediated MFS delivery, focusing on fungal infection therapy. Finally, many studies have demonstrated the promising antifungal action of MFS in vitro, but there is little or no data on antifungal activity in vertebrate animal models and clinical trials, so have a need to develop more research for the repositioning of MFS as an antifungal therapy.
Collapse
Affiliation(s)
| | - Luana Pereira Borba-Santos
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sonia Rozental
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kelly Ishida
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
4
|
Dakir EH, Gajate C, Mollinedo F. Antitumor activity of alkylphospholipid edelfosine in prostate cancer models and endoplasmic reticulum targeting. Biomed Pharmacother 2023; 167:115436. [PMID: 37683591 DOI: 10.1016/j.biopha.2023.115436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
Prostate cancer is the second most frequent cancer and the fifth leading cause of cancer death among men worldwide. While the five-year survival in local and regional prostate cancer is higher than 99%, it falls to about 28% in advanced metastatic prostate cancer. The ether lipid edelfosine is considered the prototype of a family of promising antitumor drugs collectively named as alkylphospholipid analogs. Here, we found that edelfosine was the most potent alkylphospholipid analog in inducing apoptosis in three different human prostate cancer cell lines (LNCaP, PC3, and DU145) with distinct androgen dependency, and differing in tumor suppressor phosphatase and tensin homolog (PTEN) and p53 status. Edelfosine accumulated in the endoplasmic reticulum of prostate cancer cells, leading to endoplasmic reticulum stress and cell death in the three prostate cancer cells. Inhibition of autophagy potentiated the pro-apoptotic activity of edelfosine in LNCaP and PC3 cells, where autophagy was induced as a survival response. Edelfosine induced a slight and transient inhibition of AKT in PTEN-negative LNCaP and PC3 cells, but not in PTEN-positive DU145 cells. Daily oral administration of edelfosine in murine prostate restricted AKT kinase transgenic mice, expressing active AKT in a prostate-specific manner, and in a DU145 xenograft mouse model resulted in significant tumor regression and apoptosis in tumor cells. Taken together, these results show a significant in vitro and in vivo antitumor activity of edelfosine against prostate cancer, and highlight the endoplasmic reticulum as a novel and promising therapeutic target in prostate cancer.
Collapse
Affiliation(s)
- El-Habib Dakir
- 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; Faculty of Biology, University of Latvia, Riga, Latvia.
| | - 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; 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.
| | - 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; 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.
| |
Collapse
|
5
|
Leroux M, Luquain-Costaz C, Lawton P, Azzouz-Maache S, Delton I. Fatty Acid Composition and Metabolism in Leishmania Parasite Species: Potential Biomarkers or Drug Targets for Leishmaniasis? Int J Mol Sci 2023; 24:ijms24054702. [PMID: 36902138 PMCID: PMC10003364 DOI: 10.3390/ijms24054702] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Fatty acids have received growing interest in Leishmania biology with the characterization of the enzymes allowing the complete fatty acid synthesis of this trypanosomatid parasite. This review presents a comparative analysis of the fatty acid profiles of the major classes of lipids and phospholipids in different species of Leishmania with cutaneous or visceral tropism. Specificities relating to the parasite forms, resistance to antileishmanial drugs, and host/parasite interactions are described as well as comparisons with other trypanosomatids. Emphasis is placed on polyunsaturated fatty acids and their metabolic and functional specificities, in particular, their conversion into oxygenated metabolites that are inflammatory mediators able to modulate metacyclogenesis and parasite infectivity. The impact of lipid status on the development of leishmaniasis and the potential of fatty acids as therapeutic targets or candidates for nutritional interventions are discussed.
Collapse
Affiliation(s)
- Marine Leroux
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Céline Luquain-Costaz
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
- Department of Biosciences, INSA Lyon, 69100 Villeurbanne, France
| | - Philippe Lawton
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Samira Azzouz-Maache
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Isabelle Delton
- CNRS 5007, LAGEPP, Université of Lyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
- Department of Biosciences, INSA Lyon, 69100 Villeurbanne, France
- Correspondence:
| |
Collapse
|
6
|
In Vitro Study of Cytotoxic Mechanisms of Alkylphospholipids and Alkyltriazoles in Acute Lymphoblastic Leukemia Models. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238633. [PMID: 36500726 PMCID: PMC9737184 DOI: 10.3390/molecules27238633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 12/12/2022]
Abstract
This study investigates the efficacy of miltefosine, alkylphospholipid, and alkyltriazolederivative compounds against leukemia lineages. The cytotoxic effects and cellular and molecular mechanisms of the compounds were investigated. The inhibitory potential and mechanism of inhibition of cathepsins B and L, molecular docking simulation, molecular dynamics and binding free energy evaluation were performed to determine the interaction of cathepsins and compounds. Among the 21 compounds tested, C9 and C21 mainly showed cytotoxic effects in Jurkat and CCRF-CEM cells, two human acute lymphoblastic leukemia (ALL) lineages. Activation of induced cell death by C9 and C21 with apoptotic and necrosis-like characteristics was observed, including an increase in annexin-V+propidium iodide-, annexin-V+propidium iodide+, cleaved caspase 3 and PARP, cytochrome c release, and nuclear alterations. Bax inhibitor, Z-VAD-FMK, pepstatin, and necrostatin partially reduced cell death, suggesting that involvement of the caspase-dependent and -independent mechanisms is related to cell type. Compounds C9 and C21 inhibited cathepsin L by a noncompetitive mechanism, and cathepsin B by a competitive and noncompetitive mechanism, respectively. Complexes cathepsin-C9 and cathepsin-C21 exhibited significant hydrophobic interactions, water bridges, and hydrogen bonds. In conclusion, alkyltriazoles present cytotoxic activity against acute lymphoblastic lineages and represent a promising scaffold for the development of molecules for this application.
Collapse
|
7
|
Homocysteine Concentration in the Serum of Dogs Naturally Infected with Leishmania Spp. – Association with the Stage of the Disease, Therapy, and Clinical Pathology Data. ACTA VET-BEOGRAD 2022. [DOI: 10.2478/acve-2022-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
Progressive tissue injury in canine leishmaniosis (CL) is related to the deposition of immune complexes, which induces vasculitis and leads to endothelial dysfunction. Homocysteine (Hcy) increase may worsen endothelial dysfunction, but data concerning its concentration in different CL stages and links to the acute phase response and oxidative stress are missing. We compared Hcy levels between dogs with mild (N=24) and moderate CL without treatment (N=17) and treated with anti-Leishmania drugs and vitamin B supplements (N=9). Dogs with moderate CL, regardless of therapy administration, had more distinct clinical signs, lower erythron values, and a higher level of acute-phase proteins (APPs), IgG against Leishmania spp., urea and creatinine, than dogs with mild CL. Hcy values did not differ between stages, but treated dogs had the lowest levels of Hcy. An inverse relationship existed between Hcy and the CL stage, therapy, levels of IgG, and clinical pathology data. The only positive relationship existed between Hcy and the erythron state. The disease stage and therapeutic intervention were not related to the oxidative stress level, except in the case of paraoxonase-1/Hcy ratio, indicating favorable conditions for antioxidative defense in treated dogs. In conclusion, changes in Hcy levels indicated its possible involvement with endothelial dysfunction and inverse relationship to tissue injury evaluated by APPs. Finally, Hcy might be an early marker of favorable conditions for endothelium recovery in CL.
Collapse
|
8
|
Çetinel ZÖ, Bilge D. The effects of miltefosine on the structure and dynamics of DPPC and DPPS liposomes mimicking normal and cancer cell membranes: FTIR and DSC studies. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
9
|
Dadhich R, Kapoor S. Lipidomic and Membrane Mechanical Signatures in Triple-Negative Breast Cancer: Scope for Membrane-Based Theranostics. Mol Cell Biochem 2022; 477:2507-2528. [PMID: 35595957 DOI: 10.1007/s11010-022-04459-4] [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: 09/28/2021] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer associated with poor prognosis, higher grade, and a high rate of metastatic occurrence. Limited therapeutic interventions and the compounding issue of drug resistance in triple-negative breast cancer warrants the discovery of novel therapeutic targets and diagnostic modules. To this view, in addition to proteins, lipids also regulate cellular functions via the formation of membranes that modulate membrane protein function, diffusion, and their localization; thus, orchestrating signaling hot spots enriched in specific lipids/proteins on cell membranes. Lipid deregulation in cancer leads to reprogramming of the membrane dynamics and functions impacting cell proliferation, metabolism, and metastasis, providing exciting starting points for developing lipid-based approaches for treating TNBC. In this review, we provide a detailed account of specific lipidic changes in breast cancer, link the altered lipidome with membrane structure and mechanical properties, and describe how these are linked to subsequent downstream functions implicit in cancer progression, metastasis, and chemoresistance. At the fundamental level, we discuss how the lipid-centric findings in TNBC are providing cues for developing lipid-inspired theranostic strategies while bridging existing gaps in our understanding of the functional involvement of lipid membranes in cancer.
Collapse
Affiliation(s)
- Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, 400076, India. .,Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8528, Japan.
| |
Collapse
|
10
|
Qin H, Zhang J, Dong K, Chen D, Yuan D, Chen J. Metabolic characterization and biomarkers screening for visceral leishmaniasis in golden hamsters. Acta Trop 2022; 225:106222. [PMID: 34757045 DOI: 10.1016/j.actatropica.2021.106222] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/26/2022]
Abstract
A better understanding of the changes in metabolic molecules during visceral leishmaniasis (VL) is essential to develop new strategies for diagnosis and treatment. Previous metabolomics studies on Leishmania have increased our knowledge of leishmaniasis and its causative pathogen. As these studies were mainly carried out in vitro, to go further, we conducted this global metabolomics analysis on the serum of golden hamsters. Serum samples were detected over a time course of 2, 4, 8 and 12 weeks post infection. Our results revealed that under extensively disturbed metabolomes between the infection group and controls, glycerophospholipid (GPL) metabolism was most affected over the infection time, followed by α-linoleic acid metabolism and arachidonic acid metabolism. Within GPLs, phosphatidylcholine (PC) and phosphatidylethanolamine (PE) were found to be significantly increased, while their enzyme-catalysed metabolites lysophosphatidylcholine (LPC) and lysophosphatidylethanolamine (LPE) showed no significant changes. Moreover, eight differential metabolites were selected. The ability of these metabolites to be used as a diagnostic biomarker panel was supported by receiver operating characteristic (ROC) analysis. Our findings revealed that GPL metabolism might play an important role in the response of the host to Leishmania infection. The metabolism of PC and PE might be crucial in the in vivo progression of VL. The panel of eight potential biomarkers might contribute to the diagnosis of VL.
Collapse
Affiliation(s)
- Hanxiao Qin
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jianhui Zhang
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Kai Dong
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dali Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dongmei Yuan
- Department of Human Anatomy, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Jianping Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
11
|
Wang T, Ma F, Qian HL. Defueling the cancer: ATP synthase as an emerging target in cancer therapy. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:82-95. [PMID: 34703878 PMCID: PMC8517097 DOI: 10.1016/j.omto.2021.08.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Reprogramming of cellular metabolism is a hallmark of cancer. Mitochondrial ATP synthase (MAS) produces most of the ATP that drives the cell. High expression of the MAS-composing proteins is found during cancer and is linked to a poor prognosis in glioblastoma, ovarian cancer, prostate cancer, breast cancer, and clear cell renal cell carcinoma. Cell surface-expressed ATP synthase, translocated from mitochondrion to cell membrane, involves the angiogenesis, tumorigenesis, and metastasis of cancer. ATP synthase has therefore been considered a therapeutic target. We review recent various ATP synthase inhibitors that suppress tumor growth and are being tested for the clinic.
Collapse
Affiliation(s)
- Ting Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing 100021, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hai-Li Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
12
|
Parreira de Aquino G, Mendes Gomes MA, Köpke Salinas R, Laranjeira-Silva MF. Lipid and fatty acid metabolism in trypanosomatids. MICROBIAL CELL 2021; 8:262-275. [PMID: 34782859 PMCID: PMC8561143 DOI: 10.15698/mic2021.11.764] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/18/2022]
Abstract
Trypanosomiases and leishmaniases are neglected tropical diseases that have been spreading to previously non-affected areas in recent years. Identification of new chemotherapeutics is needed as there are no vaccines and the currently available treatment options are highly toxic and often ineffective. The causative agents for these diseases are the protozoan parasites of the Trypanosomatidae family, and they alternate between invertebrate and vertebrate hosts during their life cycles. Hence, these parasites must be able to adapt to different environments and compete with their hosts for several essential compounds, such as amino acids, vitamins, ions, carbohydrates, and lipids. Among these nutrients, lipids and fatty acids (FAs) are essential for parasite survival. Trypanosomatids require massive amounts of FAs, and they can either synthesize FAs de novo or scavenge them from the host. Moreover, FAs are the major energy source during specific life cycle stages of T. brucei, T. cruzi, and Leishmania. Therefore, considering the distinctive features of FAs metabolism in trypanosomatids, these pathways could be exploited for the development of novel antiparasitic drugs. In this review, we highlight specific aspects of lipid and FA metabolism in the protozoan parasites T. brucei, T. cruzi, and Leishmania spp., as well as the pathways that have been explored for the development of new chemotherapies.
Collapse
Affiliation(s)
| | | | - Roberto Köpke Salinas
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
13
|
Park S, Kim J, Choi J, Lee C, Lee W, Park S, Park Z, Baek J, Nam J. Lipid raft-disrupting miltefosine preferentially induces the death of colorectal cancer stem-like cells. Clin Transl Med 2021; 11:e552. [PMID: 34841679 PMCID: PMC8567043 DOI: 10.1002/ctm2.552] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/22/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Lipid rafts (LRs), cholesterol-enriched microdomains on cell membranes, are increasingly viewed as signalling platforms governing critical facets of cancer progression. The phenotype of cancer stem-like cells (CSCs) presents significant hurdles for successful cancer treatment, and the expression of several CSC markers is associated with LR integrity. However, LR implications in CSCs remain unclear. METHODS This study evaluated the biological and molecular functions of LRs in colorectal cancer (CRC) by using an LR-disrupting alkylphospholipid (APL) drug, miltefosine. The mechanistic role of miltefosine in CSC inhibition was examined through normal or tumour intestinal mouse organoid, human CRC cell, CRC xenograft and miltefosine treatment gene expression profile analyses. RESULTS Miltefosine suppresses CSC populations and their self-renewal activities in CRC cells, a CSC-targeting effect leading to irreversible disruption of tumour-initiating potential in vivo. Mechanistically, miltefosine reduced the expression of a set of genes, leading to stem cell death. Among them, miltefosine transcriptionally inhibited checkpoint kinase 1 (CHEK1), indicating that LR integrity is essential for CHEK1 expression regulation. In isolated CD44high CSCs, we found that CSCs exhibited stronger therapy resistance than non-CSC counterparts by preventing cell death through CHEK1-mediated cell cycle checkpoints. However, inhibition of the LR/CHEK1 axis by miltefosine released cell cycle checkpoints, forcing CSCs to enter inappropriate mitosis with accumulated DNA damage and resulting in catastrophic cell death. CONCLUSION Our findings underscore the therapeutic potential of LR-targeting APLs for CRC treatment that overcomes the therapy-resistant phenotype of CSCs, highlighting the importance of the LR/CHEK1 axis as a novel mechanism of APLs.
Collapse
Affiliation(s)
- So‐Yeon Park
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
- Cell Logistics Research CenterGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Jee‐Heun Kim
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Jang‐Hyun Choi
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Choong‐Jae Lee
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Won‐Jae Lee
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Sehoon Park
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Zee‐Yong Park
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Jeong‐Heum Baek
- Division of Colon and Rectal SurgeryDepartment of SurgeryGil Medical CenterGachon University College of MedicineIncheonRepublic of Korea
| | - Jeong‐Seok Nam
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
- Cell Logistics Research CenterGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| |
Collapse
|
14
|
Kaushik D, Granato JT, Macedo GC, Dib PRB, Piplani S, Fung J, da Silva AD, Coimbra ES, Petrovsky N, Salunke DB. Toll-like receptor-7/8 agonist kill Leishmania amazonensis by acting as pro-oxidant and pro-inflammatory agent. J Pharm Pharmacol 2021; 73:1180-1190. [PMID: 33940589 PMCID: PMC8359742 DOI: 10.1093/jpp/rgab063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 03/29/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Evaluation of the anti-Leishmanial activity of imidazoquinoline-based TLR7/8 agonists. METHODS TLR7/8-active imidazoquinolines (2 and 3) were synthesized and assessed for activity against Leishmania amazonensis-intracellular amastigotes using mouse peritoneal macrophages. The production of reactive oxygen species (ROS), nitric oxide (NO) and cytokines was determined in infected and non-infected macrophages. KEY FINDINGS The imidazoquinolines, 2 and 3, were primarily agonists of TLR7 with compound 3 also showing modest TLR8 activity. Docking studies showed them to occupy the same binding pocket on TLR7 and 8 as the known agonists, imiquimod and resiquimod. Compounds 2 and 3 inhibited the growth of L. amazonensis-intracellular amastigotes with the most potent compound (3, IC50 = 5.93 µM) having an IC50 value close to miltefosine (IC50 = 4.05 µM), a known anti-Leishmanial drug. Compound 3 induced macrophages to produce ROS, NO and inflammatory cytokines that likely explain the anti-Leishmanial effects. CONCLUSIONS This study shows that activating TLR7 using compounds 2 or 3 induces anti-Leishmanial activity associated with induction of free radicals and inflammatory cytokines able to kill the parasites. While 2 and 3 had a very narrow cytotoxicity window for macrophages, this identifies the possibility to further develop this chemical scaffold to less cytotoxic TLR7/8 agonist for potential use as anti-Leishmanial drug.
Collapse
Affiliation(s)
- Deepender Kaushik
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, India
| | - Juliana T Granato
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Gilson C Macedo
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Paula R B Dib
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Sakshi Piplani
- Vaxine Pty Ltd., Warradale, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Johnson Fung
- Vaxine Pty Ltd., Warradale, South Australia, Australia
| | - Adilson D da Silva
- Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Warradale, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, India
- National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials (NICOVIA), Panjab University, Chandigarh, India
| |
Collapse
|
15
|
Toshkova RA, Todorova KS, Tzoneva RD. Antineoplastic effects of erufosine on Graffi myeloid tumour in hamsters. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2021. [DOI: 10.15547/bjvm.2278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cancer has become one of the most significant health challenges for both human and veterinary medicine. The present study examined the antineoplastic and antimetastatic activity of the novel membrane-targeting anticancer agent erufosine. The antitumour effects of erufosine on Graffi virus-induced experimental myeloid tumour in hamsters was assessed by histopathological methods and evaluation of some biometric parameters of tumour growth. Two schemes of experimental antitumour therapy were applied - one that started simultaneously with the tumour transplantation and a second one that started after the appearance of palpable tumours. The results demonstrated protective antitumour effect of erufosine, expressed by decrease of transplantability, tumour growth inhibition, suppression of metastatic activity and extension of mean survival time. The effectivity of the experimental therapy was more pronounced when it was started simultaneously with the transplantation of the tumour cells. Presented results suggest that erufosine is a promising drug candidate for treatment of haematological malignances.
Collapse
Affiliation(s)
- R. A. Toshkova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - K. S. Todorova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - R. D. Tzoneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
16
|
Gaillard B, Remy JS, Pons F, Lebeau L. Dual Gene Delivery Reagents From Antiproliferative Alkylphospholipids for Combined Antitumor Therapy. Front Chem 2020; 8:581260. [PMID: 33134279 PMCID: PMC7566913 DOI: 10.3389/fchem.2020.581260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/31/2020] [Indexed: 11/13/2022] Open
Abstract
Alkylphospholipids (APLs) have elicited great interest as antitumor agents due to their unique mode of action on cell membranes. However, their clinical applications have been limited so far by high hemolytic activity. Recently, cationic prodrugs of erufosine, a most promising APL, have been shown to mediate efficient intracellular gene delivery, while preserving the antiproliferative properties of the parent APL. Here, cationic prodrugs of the two APLs that are currently used in the clinic, miltefosine, and perifosine, are investigated and compared to the erufosine prodrugs. Their synthesis, stability, gene delivery and self-assembly properties, and hemolytic activity are discussed in detail. Finally, the potential of the pro-miltefosine and pro-perifosine compounds ME12 and PE12 in combined antitumor therapy is demonstrated using pUNO1-hTRAIL, a plasmid DNA encoding TRAIL, a member of the TNF superfamily. With these pro-APL compounds, we provide a proof of concept for a new promising strategy for cancer therapy combining gene therapy and APL-based chemotherapy.
Collapse
Affiliation(s)
- Boris Gaillard
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Jean-Serge Remy
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Françoise Pons
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Luc Lebeau
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| |
Collapse
|
17
|
Sarangi NK, Prabhakaran A, Keyes TE. Interaction of Miltefosine with Microcavity Supported Lipid Membrane: Biophysical Insights from Electrochemical Impedance Spectroscopy. ELECTROANAL 2020. [DOI: 10.1002/elan.202060424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Nirod Kumar Sarangi
- School of Chemical Sciences and National Centre for Sensor Research Dublin City University DCU Glasnevin Campus Dublin 9 D09 W6Y4 Ireland
| | - Amrutha Prabhakaran
- School of Chemical Sciences and National Centre for Sensor Research Dublin City University DCU Glasnevin Campus Dublin 9 D09 W6Y4 Ireland
| | - Tia E. Keyes
- School of Chemical Sciences and National Centre for Sensor Research Dublin City University DCU Glasnevin Campus Dublin 9 D09 W6Y4 Ireland
| |
Collapse
|
18
|
Lange Y, Steck TL. Active cholesterol 20 years on. Traffic 2020; 21:662-674. [PMID: 32930466 DOI: 10.1111/tra.12762] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
This review considers the following hypotheses, some well-supported and some speculative. Almost all of the sterol molecules in plasma membranes are associated with bilayer phospholipids in complexes of varied strength and stoichiometry. These complexes underlie many of the material properties of the bilayer. The small fraction of cholesterol molecules exceeding the binding capacity of the phospholipids is thermodynamically active and serves diverse functions. It circulates briskly among the cell membranes, particularly through contact sites linking the organelles. Active cholesterol provides the upstream feedback signal to multiple mechanisms governing plasma membrane homeostasis, pegging the sterol level to a threshold set by its phospholipids. Active cholesterol could also be the cargo for various inter-organelle transporters and the form excreted from cells by reverse transport. Furthermore, it is integral to the function of caveolae; a mediator of Hedgehog regulation; and a ligand for the binding of cytolytic toxins to membranes. Active cholesterol modulates a variety of plasma membrane proteins-receptors, channels and transporters-at least in vitro.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
19
|
Zulueta Díaz YDLM, Ambroggio EE, Fanani ML. Miltefosine inhibits the membrane remodeling caused by phospholipase action by changing membrane physical properties. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183407. [DOI: 10.1016/j.bbamem.2020.183407] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 01/04/2023]
|
20
|
Maffucci T, Falasca M. Inositol Polyphosphate-Based Compounds as Inhibitors of Phosphoinositide 3-Kinase-Dependent Signaling. Int J Mol Sci 2020; 21:E7198. [PMID: 33003448 PMCID: PMC7582811 DOI: 10.3390/ijms21197198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Signaling pathways regulated by the phosphoinositide 3-kinase (PI3K) enzymes have a well-established role in cancer development and progression. Over the past 30 years, the therapeutic potential of targeting this pathway has been well recognized, and this has led to the development of a multitude of drugs, some of which have progressed into clinical trials, with few of them currently approved for use in specific cancer settings. While many inhibitors compete with ATP, hence preventing the catalytic activity of the kinases directly, a deep understanding of the mechanisms of PI3K-dependent activation of its downstream effectors led to the development of additional strategies to prevent the initiation of this signaling pathway. This review summarizes previously published studies that led to the identification of inositol polyphosphates as promising parent molecules to design novel inhibitors of PI3K-dependent signals. We focus our attention on the inhibition of protein-membrane interactions mediated by binding of pleckstrin homology domains and phosphoinositides that we proposed 20 years ago as a novel therapeutic strategy.
Collapse
Affiliation(s)
- Tania Maffucci
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| |
Collapse
|
21
|
Preta G. New Insights Into Targeting Membrane Lipids for Cancer Therapy. Front Cell Dev Biol 2020; 8:571237. [PMID: 32984352 PMCID: PMC7492565 DOI: 10.3389/fcell.2020.571237] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Modulation of membrane lipid composition and organization is currently developing as an effective therapeutic strategy against a wide range of diseases, including cancer. This field, known as membrane-lipid therapy, has risen from new discoveries on the complex organization of lipids and between lipids and proteins in the plasma membranes. Membrane microdomains present in the membrane of all eukaryotic cells, known as lipid rafts, have been recognized as an important concentrating platform for protein receptors involved in the regulation of intracellular signaling, apoptosis, redox balance and immune response. The difference in lipid composition between the cellular membranes of healthy cells and tumor cells allows for the development of novel therapies based on targeting membrane lipids in cancer cells to increase sensitivity to chemotherapeutic agents and consequently defeat multidrug resistance. In the current manuscript strategies based on influencing cholesterol/sphingolipids content will be presented together with innovative ones, more focused in changing biophysical properties of the membrane bilayer without affecting the composition of its constituents.
Collapse
Affiliation(s)
- Giulio Preta
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
22
|
Tzoneva R, Stoyanova T, Petrich A, Popova D, Uzunova V, Momchilova A, Chiantia S. Effect of Erufosine on Membrane Lipid Order in Breast Cancer Cell Models. Biomolecules 2020; 10:E802. [PMID: 32455962 PMCID: PMC7277205 DOI: 10.3390/biom10050802] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Alkylphospholipids are a novel class of antineoplastic drugs showing remarkable therapeutic potential. Among them, erufosine (EPC3) is a promising drug for the treatment of several types of tumors. While EPC3 is supposed to exert its function by interacting with lipid membranes, the exact molecular mechanisms involved are not known yet. In this work, we applied a combination of several fluorescence microscopy and analytical chemistry approaches (i.e., scanning fluorescence correlation spectroscopy, line-scan fluorescence correlation spectroscopy, generalized polarization imaging, as well as thin layer and gas chromatography) to quantify the effect of EPC3 in biophysical models of the plasma membrane, as well as in cancer cell lines. Our results indicate that EPC3 affects lipid-lipid interactions in cellular membranes by decreasing lipid packing and increasing membrane disorder and fluidity. As a consequence of these alterations in the lateral organization of lipid bilayers, the diffusive dynamics of membrane proteins are also significantly increased. Taken together, these findings suggest that the mechanism of action of EPC3 could be linked to its effects on fundamental biophysical properties of lipid membranes, as well as on lipid metabolism in cancer cells.
Collapse
Affiliation(s)
- Rumiana Tzoneva
- Bulgarian Academy of Sciences, Institute of Biophysics and Biomedical Engineering, 1113 Sofia, Bulgaria; (R.T.); (T.S.); (D.P.); (V.U.); (A.M.)
| | - Tihomira Stoyanova
- Bulgarian Academy of Sciences, Institute of Biophysics and Biomedical Engineering, 1113 Sofia, Bulgaria; (R.T.); (T.S.); (D.P.); (V.U.); (A.M.)
| | - Annett Petrich
- Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Street 24-25, 14476 Potsdam, Germany;
| | - Desislava Popova
- Bulgarian Academy of Sciences, Institute of Biophysics and Biomedical Engineering, 1113 Sofia, Bulgaria; (R.T.); (T.S.); (D.P.); (V.U.); (A.M.)
| | - Veselina Uzunova
- Bulgarian Academy of Sciences, Institute of Biophysics and Biomedical Engineering, 1113 Sofia, Bulgaria; (R.T.); (T.S.); (D.P.); (V.U.); (A.M.)
| | - Albena Momchilova
- Bulgarian Academy of Sciences, Institute of Biophysics and Biomedical Engineering, 1113 Sofia, Bulgaria; (R.T.); (T.S.); (D.P.); (V.U.); (A.M.)
| | - Salvatore Chiantia
- Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Street 24-25, 14476 Potsdam, Germany;
| |
Collapse
|
23
|
Rodríguez-Nogales C, Mura S, Couvreur P, Blanco-Prieto MJ. Squalenoyl-gemcitabine/edelfosine nanoassemblies: Anticancer activity in pediatric cancer cells and pharmacokinetic profile in mice. Int J Pharm 2020; 582:119345. [PMID: 32311470 DOI: 10.1016/j.ijpharm.2020.119345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022]
Abstract
Despite the great advances accomplished in the treatment of pediatric cancers, recurrences and metastases still exacerbate prognosis in some aggressive solid tumors such as neuroblastoma and osteosarcoma. In view of the poor efficacy and toxicity of current chemotherapeutic treatments, we propose a single multitherapeutic nanotechnology-based strategy by co-assembling in the same nanodevice two amphiphilic antitumor agents: squalenoyl-gemcitabine and edelfosine. Homogeneous batches of nanoassemblies were easily formulated by the nanoprecipitation method. Their anticancer activity was tested in pediatric cancer cell lines and pharmacokinetic studies were performed in mice. In vitro assays revealed a synergistic effect when gemcitabine was co-administered with edelfosine. Squalenoyl-gemcitabine/edelfosine nanoassemblies were found to be capable of intracellular translocation in patient-derived metastatic pediatric osteosarcoma cells and showed a better antitumor profile than squalenoyl-gemcitabine nanoassemblies alone. The intravenous administration of this combinatorial nanomedicine in mice exhibited a controlled release behavior of gemcitabine and diminished edelfosine plasma peak concentrations. These findings make it a suitable pre-clinical candidate for childhood cancer therapy.
Collapse
Affiliation(s)
- C Rodríguez-Nogales
- Chemistry and Pharmaceutical Technology Department, Universidad de Navarra, Pamplona 31008, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - S Mura
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 92296 Châtenay-Malabry, France
| | - P Couvreur
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 92296 Châtenay-Malabry, France.
| | - M J Blanco-Prieto
- Chemistry and Pharmaceutical Technology Department, Universidad de Navarra, Pamplona 31008, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain.
| |
Collapse
|
24
|
Potent Anticancer Effect of the Natural Steroidal Saponin Gracillin Is Produced by Inhibiting Glycolysis and Oxidative Phosphorylation-Mediated Bioenergetics. Cancers (Basel) 2020; 12:cancers12040913. [PMID: 32276500 PMCID: PMC7226187 DOI: 10.3390/cancers12040913] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 01/19/2023] Open
Abstract
Metabolic rewiring to utilize aerobic glycolysis is a hallmark of cancer. However, recent findings suggest the role of mitochondria in energy generation in cancer cells and the metabolic switch to oxidative phosphorylation (OXPHOS) in response to the blockade of glycolysis. We previously demonstrated that the antitumor effect of gracillin occurs through the inhibition of mitochondrial complex II-mediated energy production. Here, we investigated the potential of gracillin as an anticancer agent targeting both glycolysis and OXPHOS in breast and lung cancer cells. Along with the reduction in adenosine triphosphate (ATP) production, gracillin markedly suppresses the production of several glycolysis-associated metabolites. A docking analysis and enzyme assay suggested phosphoglycerate kinase 1 (PGK1) is a potential target for the antiglycolytic effect of gracillin. Gracillin reduced the viability and colony formation ability of breast cancer cells by inducing apoptosis. Gracillin displayed efficacious antitumor effects in mice bearing breast cancer cell line or breast cancer patient-derived tumor xenografts with no overt changes in body weight. An analysis of publicly available datasets further suggested that PGK1 expression is associated with metastasis status and poor prognosis in patients with breast cancer. These results suggest that gracillin is a natural anticancer agent that inhibits both glycolysis and mitochondria-mediated bioenergetics.
Collapse
|
25
|
Reimão JQ, Pita Pedro DP, Coelho AC. The preclinical discovery and development of oral miltefosine for the treatment of visceral leishmaniasis: a case history. Expert Opin Drug Discov 2020; 15:647-658. [PMID: 32202449 DOI: 10.1080/17460441.2020.1743674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Visceral leishmaniasis (VL) is a vector-borne disease caused by Leishmania donovani or Leishmania infantum. Closely related to poverty, VL is fatal and represents one of the main burdens on public health in developing countries. Treatment of VL relies exclusively on chemotherapy, a strategy still experiencing numerous limitations. Miltefosine (MF) has been used in the chemotherapy of VL in some endemic areas, and has been expanded to other regions, being considered crucial in eradication programs. AREAS COVERED This article reviews the most relevant preclinical and clinical aspects of MF, its mechanism of action and resistance to Leishmania parasites, as well as its limitations. The authors also give their perspectives on the treatment of VL. EXPERT OPINION The discovery of MF represented an enormous advance in the chemotherapy of VL, since it was the first oral drug for this neglected disease. Beyond selection of resistant parasites due to drug pressure, several other factors can lead to treatment failure such as, for example, factors intrinsic to the host, parasite and the drug itself. Although its efficacy as a monotherapy has reduced over recent years, MF is still an important alternative in VL chemotherapy, especially when used in combination with other drugs.
Collapse
Affiliation(s)
- Juliana Q Reimão
- Departamento de Morfologia e Patologia Básica, Faculdade de Medicina de Jundiaí , Jundiaí, Brazil
| | - Débora P Pita Pedro
- Departamento de Morfologia e Patologia Básica, Faculdade de Medicina de Jundiaí , Jundiaí, Brazil
| | - Adriano C Coelho
- Departamento de Biologia Animal, Instituto de Biologia, Universidade Estadual de Campinas , Campinas, Brazil
| |
Collapse
|
26
|
Eleftheriou K, Kaminari A, Panagiotaki KN, Sideratou Z, Zachariadis M, Anastassopoulou J, Tsiourvas D. A combination drug delivery system employing thermosensitive liposomes for enhanced cell penetration and improved in vitro efficacy. Int J Pharm 2020; 574:118912. [PMID: 31809858 DOI: 10.1016/j.ijpharm.2019.118912] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/15/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023]
Abstract
Drug-loaded thermosensitive liposomes are investigated as drug delivery systems in combination with local mild hyperthermia therapy due to their capacity to release their cargo at a specific temperature range (40-42 °C). Additional benefit can be achieved by the development of such systems that combine two different anticancer drugs, have cell penetration properties and, when heated, release their drug payload in a controlled fashion. To this end, liposomes were developed incorporating at low concentration (5 mol%) a number of monoalkylether phosphatidylcholine lipids, encompassing the platelet activating factor, PAF, and its analogues that induce thermoresponsiveness and have anticancer biological activity. These thermoresponsive liposomes were efficiently (>90%) loaded with doxorubicin (DOX), and their thermal properties, stability and drug release were investigated both at 37 ◦C and at elevated temperatures. In vitro studies of the most advantageous liposomal formulation containing the methylated PAF derivative (methyl-PAF, edelfosine), an established antitumor agent, were performed on human prostate cancer cell lines. This system exhibits controlled release of DOX at 40-42 °C, enhanced cell uptake due to the presence of methyl-PAF, and improved cell viability inhibition due to the combined action of both medications.
Collapse
Affiliation(s)
- Kleopatra Eleftheriou
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Archontia Kaminari
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Katerina N Panagiotaki
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Zili Sideratou
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Michael Zachariadis
- Institute of Biosciences and Applications, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Jane Anastassopoulou
- Radiation Chemistry and Biospectroscopy, School of Chemical Engineering, National Technical University of Athens, Athens, Greece
| | - Dimitris Tsiourvas
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece.
| |
Collapse
|
27
|
Gaillard B, Seguin C, Remy JS, Pons F, Lebeau L. Erufosine (ErPC3) Cationic Prodrugs as Dual Gene Delivery Reagents for Combined Antitumor Therapy. Chemistry 2019; 25:15662-15679. [PMID: 31549752 DOI: 10.1002/chem.201903976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/23/2019] [Indexed: 12/14/2022]
Abstract
Sixteen cationic prodrugs of the antitumor alkylphospholipid (APL) erufosine were rationally synthesized to provide original gene delivery reagents with improved cytotoxicity profile. The DNA complexation properties of these cationic lipids were determined and associated transfection rates were measured. Furthermore, the self-assembly properties of the pro-erufosine compounds were investigated and their critical aggregation concentration was determined. Their hydrolytic stability under pH conditions mimicking the extracellular environment and the late endosome milieu was measured. Hemolytic activity and cytotoxicity of the compounds were investigated. The results obtained in various cell lines demonstrate that the prodrugs of erufosine display antineoplastic activity similar to that of the parent antitumor drug but are not associated with hemolytic toxicity, which is a dose-limiting side effect of APLs and a major obstacle to their use in anticancer therapeutic regimen. Furthermore, by using lipoplexes prepared from a prodrug of erufosine and a plasmid DNA encoding a pro-apoptotic protein (TRAIL), evidence was provided for selective cytotoxicity towards tumor cells while nontumor cells were resistant. This study demonstrates that the combination approach involving well tolerated erufosine cationic prodrugs and cancer gene therapy holds significant promise in tumor therapy.
Collapse
Affiliation(s)
- Boris Gaillard
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| | - Cendrine Seguin
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| | - Jean-Serge Remy
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| | - Françoise Pons
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| | - Luc Lebeau
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| |
Collapse
|
28
|
Aron M, Vince O, Gray M, Mannaris C, Stride E. Investigating the Role of Lipid Transfer in Microbubble-Mediated Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:13205-13215. [PMID: 31517490 DOI: 10.1021/acs.langmuir.9b02404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Sonoporation, the permeabilization of cell membranes following exposure to microbubbles and ultrasound, has considerable potential for therapeutic delivery. To date, engineering of microbubbles for these applications has focused primarily upon optimizing microbubble size and stability, or attachment of targeting species and/or drug molecules. In this work, it is demonstrated that the microbubble coating can also be tailored to directly influence cell permeabilization. Specifically, lipid exchange mechanisms between phospholipid microbubbles and cells can be exploited to significantly increase sonoporation efficiency in vitro. A theoretical analysis of the energy required for pore formation was carried out. From this, it was hypothesized that sonoporation could be promoted by the transfer of lipid molecules with appropriate carbon chain length and/or shape (cylindrical or conical). Spectral imaging with a hydration-sensitive membrane probe (C-Laurdan) was used to measure changes in the membrane lipid order of A-549 cancer cells following exposure to suspensions of different phospholipids. Two candidate lipids were identified, a short-chain-length phospholipid (1,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC)) and a medium-chain-length lysolipid (1-palmitoyl-2-hydroxy-sn-glycero-3-phosphocholine (16:0 lyso-PC)). Microbubbles were prepared with matched concentrations, size distributions, and acoustic responses. Confocal microscopy was used to measure cell uptake of a model drug (propidium iodide) with and without ultrasound exposure (1 MHz, 250 kPa peak negative pressure, 1 kHz pulse repetition frequency, 10% duty cycle, 15 s exposure). Despite significantly decreasing the cell membrane lipid order, DLPC did not increase sonoporation. Microbubbles containing 16:0 lyso-PC, however, produced a ∼5-fold increase in sonoporation compared to control microbubbles. Importantly, the lyso-PC molecules were incorporated into the microbubble coating and did not affect cell permeability prior to ultrasound exposure. These findings indicate that microbubbles can be engineered to exploit lipid exchange between microbubble shells and cell membranes to enhance drug delivery, a new optimization route that may lead to enhanced therapeutic efficacy of ultrasound-mediated treatments.
Collapse
Affiliation(s)
- Miles Aron
- Institute of Biomedical Engineering , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , U.K
| | - Oliver Vince
- Institute of Biomedical Engineering , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , U.K
| | - Michael Gray
- Institute of Biomedical Engineering , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , U.K
| | - Christophoros Mannaris
- Institute of Biomedical Engineering , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , U.K
| | - Eleanor Stride
- Institute of Biomedical Engineering , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , U.K
| |
Collapse
|
29
|
Sarmento-Ribeiro AB, Scorilas A, Gonçalves AC, Efferth T, Trougakos IP. The emergence of drug resistance to targeted cancer therapies: Clinical evidence. Drug Resist Updat 2019; 47:100646. [PMID: 31733611 DOI: 10.1016/j.drup.2019.100646] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022]
Abstract
For many decades classical anti-tumor therapies included chemotherapy, radiation and surgery; however, in the last two decades, following the identification of the genomic drivers and main hallmarks of cancer, the introduction of therapies that target specific tumor-promoting oncogenic or non-oncogenic pathways, has revolutionized cancer therapeutics. Despite the significant progress in cancer therapy, clinical oncologists are often facing the primary impediment of anticancer drug resistance, as many cancer patients display either intrinsic chemoresistance from the very beginning of the therapy or after initial responses and upon repeated drug treatment cycles, acquired drug resistance develops and thus relapse emerges, resulting in increased mortality. Our attempts to understand the molecular basis underlying these drug resistance phenotypes in pre-clinical models and patient specimens revealed the extreme plasticity and adaptive pathways employed by tumor cells, being under sustained stress and extensive genomic/proteomic instability due to the applied therapeutic regimens. Subsequent efforts have yielded more effective inhibitors and combinatorial approaches (e.g. the use of specific pharmacologic inhibitors with immunotherapy) that exhibit synergistic effects against tumor cells, hence enhancing therapeutic indices. Furthermore, new advanced methodologies that allow for the early detection of genetic/epigenetic alterations that lead to drug chemoresistance and prospective validation of biomarkers which identify patients that will benefit from certain drug classes, have started to improve the clinical outcome. This review discusses emerging principles of drug resistance to cancer therapies targeting a wide array of oncogenic kinases, along with hedgehog pathway and the proteasome and apoptotic inducers, as well as epigenetic and metabolic modulators. We further discuss mechanisms of resistance to monoclonal antibodies, immunomodulators and immune checkpoint inhibitors, potential biomarkers of drug response/drug resistance, along with possible new therapeutic avenues for the clinicians to combat devastating drug resistant malignancies. It is foreseen that these topics will be major areas of focused multidisciplinary translational research in the years to come.
Collapse
Affiliation(s)
- Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Hematology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal.
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
30
|
Rodríguez-Nogales C, Sebastián V, Irusta S, Desmaële D, Couvreur P, Blanco-Prieto MJ. A unique multidrug nanomedicine made of squalenoyl-gemcitabine and alkyl-lysophospholipid edelfosine. Eur J Pharm Biopharm 2019; 144:165-173. [PMID: 31546021 DOI: 10.1016/j.ejpb.2019.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 02/04/2023]
Abstract
Among anticancer nanomedicines, squalenoyl nanocomposites have obtained encouraging outcomes in a great variety of tumors. The prodrug squalenoyl-gemcitabine has been chosen in this study to construct a novel multidrug nanosystem in combination with edelfosine, an alkyl-lysophopholipid with proven anticancer activity. Given their amphiphilic nature, it was hypothesized that both anticancer compounds, with complementary molecular targets, could lead to the formation of a new multitherapy nanomedicine. Nanoassemblies were formulated by the nanoprecipitation method and characterized by dynamic light scattering, transmission electron microscopy and X-ray photoelectron spectroscopy. Because free edelfosine is highly hemolytic, hemolysis experiments were performed using human blood erythrocytes and nanoassemblies efficacy was evaluated in a patient-derived metastatic pediatric osteosarcoma cell line. It was observed that these molecules spontaneously self-assembled as stable and monodisperse nanoassemblies of 51 ± 1 nm in a surfactant/polymer free-aqueous suspension. Compared to squalenoyl-gemcitabine nanoassemblies, the combination of squalenoyl-gemcitabine with edelfosine resulted in smaller particle size and a new supramolecular conformation, with higher stability and drug content, and ameliorated antitumor profile.
Collapse
Affiliation(s)
- C Rodríguez-Nogales
- Chemistry and Pharmaceutical Technology Department, Universidad de Navarra, Pamplona 31008, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - V Sebastián
- Department of Chemical and Environmental Engineering & Institute of Nanoscience of Aragon (INA), University of Zaragoza, Zaragoza 50018, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid 28029, Spain
| | - S Irusta
- Department of Chemical and Environmental Engineering & Institute of Nanoscience of Aragon (INA), University of Zaragoza, Zaragoza 50018, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid 28029, Spain
| | - D Desmaële
- Institut Galien Paris-Sud, UMR CNRS 8612, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry Cedex 92290, France
| | - P Couvreur
- Institut Galien Paris-Sud, UMR CNRS 8612, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry Cedex 92290, France.
| | - M J Blanco-Prieto
- Chemistry and Pharmaceutical Technology Department, Universidad de Navarra, Pamplona 31008, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain.
| |
Collapse
|
31
|
Sánchez-Fernández EM, García-Moreno MI, García-Hernández R, Padrón JM, García Fernández JM, Gamarro F, Ortiz Mellet C. Thiol-ene "Click" Synthesis and Pharmacological Evaluation of C-Glycoside sp 2-Iminosugar Glycolipids. Molecules 2019; 24:E2882. [PMID: 31398901 PMCID: PMC6720825 DOI: 10.3390/molecules24162882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/30/2022] Open
Abstract
The unique stereoelectronic properties of sp2-iminosugars enable their participation in glycosylation reactions, thereby behaving as true carbohydrate chemical mimics. Among sp2-iminosugar conjugates, the sp2-iminosugar glycolipids (sp2-IGLs) have shown a variety of interesting pharmacological properties ranging from glycosidase inhibition to antiproliferative, antiparasitic, and anti-inflammatory activities. Developing strategies compatible with molecular diversity-oriented strategies for structure-activity relationship studies was therefore highly wanted. Here we show that a reaction sequence consisting in stereoselective C-allylation followed by thiol-ene "click" coupling provides a very convenient access to α-C-glycoside sp2-IGLs. Both the glycone moiety and the aglycone tail can be modified by using sp2-iminosugar precursors with different configurational profiles (d-gluco or d-galacto in this work) and varied thiols, as well as by oxidation of the sulfide adducts (to the corresponding sulfones in this work). A series of derivatives was prepared in this manner and their glycosidase inhibitory, antiproliferative and antileishmanial activities were evaluated in different settings. The results confirm that the inhibition of glycosidases, particularly α-glucosidase, and the antitumor/leishmanicidal activities are unrelated. The data are also consistent with the two later activities arising from the ability of the sp2-IGLs to interfere in the immune system response in a cell line and cell context dependent manner.
Collapse
Affiliation(s)
- Elena M Sánchez-Fernández
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/Profesor García González 1, 41012 Seville, Spain.
| | - M Isabel García-Moreno
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/Profesor García González 1, 41012 Seville, Spain
| | - Raquel García-Hernández
- Instituto de Parasitología y Biomedicina "López-Neyra", IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, 18016 Granada, Spain
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Centro de Investigaciones Biomédicas de Canarias (CIBCAN), Universidad de La Laguna, 38206 La Laguna, Spain
| | - José M García Fernández
- Instituto de Investigaciones Químicas (IIQ), CSIC - University of Sevilla, Avda. Américo Vespucio 49, 41092 Sevilla, Spain
| | - Francisco Gamarro
- Instituto de Parasitología y Biomedicina "López-Neyra", IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, 18016 Granada, Spain
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/Profesor García González 1, 41012 Seville, Spain.
| |
Collapse
|
32
|
Bouraoui A, Ghanem R, Berchel M, Vié V, Le Guen Y, Paboeuf G, Deschamps L, Le Gall T, Montier T, Jaffrès PA. Bis-Thioether-Containing Lipid Chains in Cationic Amphiphiles: Physicochemical Properties and Applications in Gene Delivery. Chemphyschem 2019; 20:2187-2194. [PMID: 31393059 DOI: 10.1002/cphc.201900626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/18/2019] [Indexed: 11/08/2022]
Abstract
Cationic amphiphiles featuring two thioether functions in each lipid chain of bicatenar cationic amphiphiles are reported here for the first time. The physicochemical properties and transfection abilities of these new amphiphiles were compared with those of already reported analogues featuring either (i) saturated, (ii) unsaturated or (iii) mono-thioether containing lipid chains. The homogeneity of the series of new compounds allowed to clearly underscore the effect of bis-thioether containing lipid chains. This study shows that besides previous strategies based on unsaturation or ramification, the incorporation of two thioether functions per lipid chain constitutes an original complementary alternative to tune the supramolecular properties of amphiphilic compounds. The potential of this strategy was evaluated in the context of gene delivery and report that two cationic amphiphiles (i. e. 4 a and 4 b) can be proposed as new efficient transfection reagents.
Collapse
Affiliation(s)
- Amal Bouraoui
- Univ Brest, CNRS, CEMCA, UMR CNRS 6521, 6 Avenue Victor Le Gorgeu, F-29238, Brest, France
| | - Rosy Ghanem
- Univ Brest, INSERM UMR 1078, IBSAM, UFR Médecine et Sciences de la Santé, CHRU Brest, 22 avenue Camille Desmoulins, F-29238, Brest, France
| | - Mathieu Berchel
- Univ Brest, CNRS, CEMCA, UMR CNRS 6521, 6 Avenue Victor Le Gorgeu, F-29238, Brest, France
| | - Véronique Vié
- Univ Rennes, CNRS, IPR - UMR 6251, ScanMAT - UMS 2001, F-35000, Rennes, France
| | - Yann Le Guen
- Univ Brest, INSERM UMR 1078, IBSAM, UFR Médecine et Sciences de la Santé, CHRU Brest, 22 avenue Camille Desmoulins, F-29238, Brest, France
| | - Gilles Paboeuf
- Univ Rennes, CNRS, IPR - UMR 6251, ScanMAT - UMS 2001, F-35000, Rennes, France
| | - Laure Deschamps
- Univ Brest, CNRS, CEMCA, UMR CNRS 6521, 6 Avenue Victor Le Gorgeu, F-29238, Brest, France
| | - Tony Le Gall
- Univ Brest, INSERM UMR 1078, IBSAM, UFR Médecine et Sciences de la Santé, CHRU Brest, 22 avenue Camille Desmoulins, F-29238, Brest, France
| | - Tristan Montier
- Univ Brest, INSERM UMR 1078, IBSAM, UFR Médecine et Sciences de la Santé, CHRU Brest, 22 avenue Camille Desmoulins, F-29238, Brest, France
| | - Paul-Alain Jaffrès
- Univ Brest, CNRS, CEMCA, UMR CNRS 6521, 6 Avenue Victor Le Gorgeu, F-29238, Brest, France
| |
Collapse
|
33
|
Zhou Z, Luo B, Liu X, Chen M, Lan W, Iovanna JL, Peng L, Xia Y. Flavonoid-alkylphospholipid conjugates elicit dual inhibition of cancer cell growth and lipid accumulation. Chem Commun (Camb) 2019; 55:8919-8922. [PMID: 31270526 DOI: 10.1039/c9cc04084f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer development is often associated with lipid metabolic reprogramming, including aberrant lipid accumulation. We create novel paradigms endowed with dual functions of anticancer activity and inhibition of lipid accumulation by conjugating the natural product quercetin and synthetic alkylphospholipid drugs, and harnessing the biomedical effects of both. These conjugates offer fresh perspectives in the search for anticancer candidates.
Collapse
Affiliation(s)
- Zhengwei Zhou
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, China.
| | - Biyao Luo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, China.
| | - Xi Liu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, China.
| | - Mimi Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, China.
| | - Wenjun Lan
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, Equipe Labellisé par La Ligue, France. and Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille 13288, France
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille 13288, France
| | - Ling Peng
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, Equipe Labellisé par La Ligue, France.
| | - Yi Xia
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, China.
| |
Collapse
|
34
|
Uzunova V, Tzoneva R, Stoyanova T, Pankov R, Skrobanska R, Georgiev G, Maslenkova L, Tsonchev Z, Momchilova A. Dimethylsphingosine and miltefosine induce apoptosis in lung adenocarcinoma A549 cells in a synergistic manner. Chem Biol Interact 2019; 310:108731. [PMID: 31265827 DOI: 10.1016/j.cbi.2019.108731] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/11/2019] [Accepted: 06/27/2019] [Indexed: 10/26/2022]
Abstract
Lung cancer is one of the most common and lethal types of oncological diseases. Despite the advanced therapeutic approaches, the prognosis for lung cancer still remains poor. Apparently, there is an imperative need for more efficient therapeutic strategies. In this work we report that concurrent treatment of human adenocarcinoma A549 cells with specific concentrations of two antitumor agents, the sphingosine kinase 1 inhibitor N, N dimethylsphingosine (DMS) and the alkylphosphocholine miltefosine, induced synergistic cytotoxic effect, which was confirmed by calculation of the combination index. The simultaneous action of these agents, induced significant decrease of A549 cell number, as well as pronounced morphological alterations. Combined drugs caused substantial apoptotic events, and significant reduction of the pro-survival marker sphingosine- 1-phosphate (S1P), when compared to the individual treatments with each of the anticancer drugs alone. Miltefosine is known to affect the synthesis of choline-containing phospholipids, including sphingomyelin, but we report for the first time that it also reduces S1P. Here we suggest a putative mechanism underlying the effect of miltefosine on sphingosine kinase 1, involving miltefosine-induced inhibition of protein kinase C. In conclusion, our findings provide a possibility for treatment of lung cancer cells with lower concentrations of the two antitumor drugs, DMS and miltefosine, which is favorable, regarding their potential cytotoxicity to normal cells.
Collapse
Affiliation(s)
- Veselina Uzunova
- Department of Lipid-Protein Interactions, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev str. bl 21, 1113, Sofia, Bulgaria
| | - Rumiana Tzoneva
- Department of Lipid-Protein Interactions, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev str. bl 21, 1113, Sofia, Bulgaria
| | - Tihomira Stoyanova
- Department of Lipid-Protein Interactions, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev str. bl 21, 1113, Sofia, Bulgaria
| | - Roumen Pankov
- Department of Cytology, Histology and Embryology, Biological Faculty, Sofia University, 8, Dragan Tzankov str, 1164, Sofia, Bulgaria
| | - Ralica Skrobanska
- Department of Cytology, Histology and Embryology, Biological Faculty, Sofia University, 8, Dragan Tzankov str, 1164, Sofia, Bulgaria
| | - Georgi Georgiev
- Department of Cytology, Histology and Embryology, Biological Faculty, Sofia University, 8, Dragan Tzankov str, 1164, Sofia, Bulgaria
| | - Liliana Maslenkova
- Department of Lipid-Protein Interactions, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev str. bl 21, 1113, Sofia, Bulgaria
| | - Zlatan Tsonchev
- Department of Neurology, ISUL Hospital Tsaritsa Yoanna, 8 Bialo more str, 1527, Sofia, Bulgaria
| | - Albena Momchilova
- Department of Lipid-Protein Interactions, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev str. bl 21, 1113, Sofia, Bulgaria.
| |
Collapse
|
35
|
Enkavi G, Javanainen M, Kulig W, Róg T, Vattulainen I. Multiscale Simulations of Biological Membranes: The Challenge To Understand Biological Phenomena in a Living Substance. Chem Rev 2019; 119:5607-5774. [PMID: 30859819 PMCID: PMC6727218 DOI: 10.1021/acs.chemrev.8b00538] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Indexed: 12/23/2022]
Abstract
Biological membranes are tricky to investigate. They are complex in terms of molecular composition and structure, functional over a wide range of time scales, and characterized by nonequilibrium conditions. Because of all of these features, simulations are a great technique to study biomembrane behavior. A significant part of the functional processes in biological membranes takes place at the molecular level; thus computer simulations are the method of choice to explore how their properties emerge from specific molecular features and how the interplay among the numerous molecules gives rise to function over spatial and time scales larger than the molecular ones. In this review, we focus on this broad theme. We discuss the current state-of-the-art of biomembrane simulations that, until now, have largely focused on a rather narrow picture of the complexity of the membranes. Given this, we also discuss the challenges that we should unravel in the foreseeable future. Numerous features such as the actin-cytoskeleton network, the glycocalyx network, and nonequilibrium transport under ATP-driven conditions have so far received very little attention; however, the potential of simulations to solve them would be exceptionally high. A major milestone for this research would be that one day we could say that computer simulations genuinely research biological membranes, not just lipid bilayers.
Collapse
Affiliation(s)
- Giray Enkavi
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Matti Javanainen
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy
of Sciences, Flemingovo naḿesti 542/2, 16610 Prague, Czech Republic
- Computational
Physics Laboratory, Tampere University, P.O. Box 692, FI-33014 Tampere, Finland
| | - Waldemar Kulig
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Tomasz Róg
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- Computational
Physics Laboratory, Tampere University, P.O. Box 692, FI-33014 Tampere, Finland
| | - Ilpo Vattulainen
- Department
of Physics, University of
Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- Computational
Physics Laboratory, Tampere University, P.O. Box 692, FI-33014 Tampere, Finland
- MEMPHYS-Center
for Biomembrane Physics
| |
Collapse
|
36
|
Nagaraja S, Ankri S. Target identification and intervention strategies against amebiasis. Drug Resist Updat 2019; 44:1-14. [PMID: 31112766 DOI: 10.1016/j.drup.2019.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 04/27/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022]
Abstract
Entamoeba histolytica is the etiological agent of amebiasis, which is an endemic parasitic disease in developing countries and is the cause of approximately 70,000 deaths annually. E. histolytica trophozoites usually reside in the colon as a non-pathogenic commensal in most infected individuals (90% of infected individuals are asymptomatic). For unknown reasons, these trophozoites can become virulent and invasive, cause amebic dysentery, and migrate to the liver where they cause hepatocellular damage. Amebiasis is usually treated either by amebicides which are classified as (a) luminal and are active against the luminal forms of the parasite, (b) tissue and are effective against those parasites that have invaded tissues, and (c) mixed and are effective against the luminal forms of the parasite and those forms which invaded the host's tissues. Of the amebicides, the luminal amebicide, metronidazole (MTZ), is the most widely used drug to treat amebiasis. Although well tolerated, concerns about its adverse effects and the possible emergence of MTZ-resistant strains of E. histolytica have led to the development of new therapeutic strategies against amebiasis. These strategies include improving the potency of existing amebicides, discovering new uses for approved drugs (repurposing of existing drugs), drug rediscovery, vaccination, drug targeting of essential E. histolytica components, and the use of probiotics and bioactive natural products. This review examines each of these strategies in the light of the current knowledge on the gut microbiota of patients with amebiasis.
Collapse
Affiliation(s)
- Shruti Nagaraja
- Department of Molecular Microbiology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Serge Ankri
- Department of Molecular Microbiology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
37
|
Choline kinase inhibitors EB-3D and EB-3P interferes with lipid homeostasis in HepG2 cells. Sci Rep 2019; 9:5109. [PMID: 30911014 PMCID: PMC6433853 DOI: 10.1038/s41598-019-40885-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/12/2019] [Indexed: 01/11/2023] Open
Abstract
A full understanding of the molecular mechanism of action of choline kinase α (ChoKα) inhibitors at the cell level is essential for developing therapeutic and preventive approaches for cancer. The aim of the present study was to evaluate the effects of the ChoKα inhibitors EB-3D and EB-3P on lipid metabolism in HepG2 cells. We used [methyl-14C]choline, [1,2-14C]acetic acid and [2-3H]glycerol as exogenous precursors of the corresponding phospholipids and neutral lipids. [Methyl-14C]choline was also used to determine choline uptake. Protein levels were determined by Western blot. Ultrastructural alterations were investigated by transmission electron microscopy. In this work, we demonstrate that EB-3D and EB-3P interfere with phosphatidylcholine biosynthesis via both CDP-choline pathway and choline uptake by the cell. Moreover, the synthesis of both diacylglycerols and triacylglycerols was affected by cell exposure to both inhibitors. These effects were accompanied by a substantial decrease in cholesterol biosynthesis, as well as alterations in the expression of proteins related to cholesterol homeostasis. We also found that EB-3D and EB-3P lowered ChoKα protein levels. All these effects could be explained by the modulation of the AMP-activated protein kinase signalling pathway. We show that both inhibitors cause mitochondrial alteration and an endoplasmic reticulum stress response. EB-3D and EB-3P exert effects on ChoKα expression, AMPK activation, apoptosis, endoplasmic reticulum stress and lipid metabolism. Taken together, results show that EB-3D and EB-3P have potential anti-cancer activity through the deregulation of lipid metabolism.
Collapse
|
38
|
Margaritova Zaharieva M, Dimitrov Kroumov A, Dimitrova L, Tsvetkova I, Trochopoulos A, Mihaylov Konstantinov S, Reinhold Berger M, Momchilova M, Yoncheva K, Miladinov Najdenski H. Micellar curcumin improves the antibacterial activity of the alkylphosphocholines erufosine and miltefosine against pathogenic Staphyloccocus aureus strains. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2018.1533792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Maya Margaritova Zaharieva
- Department of Infectious Microbiology, The Stephan Angeloff Institute of Microbiology Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Alexander Dimitrov Kroumov
- Department of Applied Microbiology, The Stephan Angeloff Institute of Microbiology Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Lyudmila Dimitrova
- Department of Infectious Microbiology, The Stephan Angeloff Institute of Microbiology Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Iva Tsvetkova
- Department of Infectious Microbiology, The Stephan Angeloff Institute of Microbiology Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Antonios Trochopoulos
- Department of Pharmacology Pharmacotherapy and Toxicology Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Spiro Mihaylov Konstantinov
- Department of Pharmacology Pharmacotherapy and Toxicology Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | | | - Milena Momchilova
- Department of Pharmaceutical Technology and Biopharmaceutics Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Krassimira Yoncheva
- Department of Pharmaceutical Technology and Biopharmaceutics Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Hristo Miladinov Najdenski
- Department of Infectious Microbiology, The Stephan Angeloff Institute of Microbiology Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
39
|
In vitro effects of the antitumor drug miltefosine on human erythrocytes and molecular models of its membrane. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:17-25. [DOI: 10.1016/j.bbamem.2018.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/31/2022]
|
40
|
Bernardes N, Fialho AM. Perturbing the Dynamics and Organization of Cell Membrane Components: A New Paradigm for Cancer-Targeted Therapies. Int J Mol Sci 2018; 19:E3871. [PMID: 30518103 PMCID: PMC6321595 DOI: 10.3390/ijms19123871] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 01/26/2023] Open
Abstract
Cancer is a multi-process disease where different mechanisms exist in parallel to ensure cell survival and constant adaptation to the extracellular environment. To adapt rapidly, cancer cells re-arrange their plasma membranes to sustain proliferation, avoid apoptosis and resist anticancer drugs. In this review, we discuss novel approaches based on the modifications and manipulations that new classes of molecules can exert in the plasma membrane lateral organization and order of cancer cells, affecting growth factor signaling, invasiveness, and drug resistance. Furthermore, we present azurin, an anticancer protein from bacterial origin, as a new approach in the development of therapeutic strategies that target the cell membrane to improve the existing standard therapies.
Collapse
Affiliation(s)
- Nuno Bernardes
- iBB-Institute for Bioengineering and Biosciences, Biological Sciences Research Group, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal.
| | - Arsenio M Fialho
- iBB-Institute for Bioengineering and Biosciences, Biological Sciences Research Group, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal.
- Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal.
| |
Collapse
|
41
|
Hassan AHE, Park HR, Yoon YM, Kim HI, Yoo SY, Lee KW, Lee YS. Antiproliferative 3-deoxysphingomyelin analogs: Design, synthesis, biological evaluation and molecular docking of pyrrolidine-based 3-deoxysphingomyelin analogs as anticancer agents. Bioorg Chem 2018; 84:444-455. [PMID: 30576908 DOI: 10.1016/j.bioorg.2018.11.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 11/21/2018] [Accepted: 11/24/2018] [Indexed: 12/13/2022]
Abstract
Sphingomyelins and glycerophospholipids are structurally related phospholipids. Nevertheless, glycerophospholipids analogs are known as antitumor agents while sphingomyelin analogs were reported as cytoprotective agents. Herein, we have addressed the development of 3-deoxysphingomyelin analogs as cytotoxic agents possessing modified sphingobases. Thus, pyrrolidine-based 3-deoxysphingomyelin analogs were synthesized and evaluated against a panel of cell lines representing four major types of cancers. Compounds 3d, 4d and 6d elicited better GI50 values than the FDA approved drug miltefosine. Investigation of their impact on Akt phosphorylation as a possible mechanism for the antiproliferative activity of this class of compounds revealed that these compounds might elicit a concentration-dependent mechanism via inhibition of Akt phosphorylation at the lower concentration. Molecular docking predicted their binding modes to Akt to involve polar head binding to the Pleckstrin homology domain and hydrophobic tail extension into a hydrophobic pocket connecting the Pleckstrin homology domain and the kinase domain. As a whole, the described work suggests compounds 3d, 4d and 6d as promising pyrrolidine-based 3-deoxysphingomyelin analogs for development of novel cancer therapies.
Collapse
Affiliation(s)
- Ahmed H E Hassan
- Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Hye Rim Park
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoon Mi Yoon
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hye In Kim
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Yeun Yoo
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kun Won Lee
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yong Sup Lee
- Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Life and Nanopharmaceutical Science, Kyung Hee University, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
42
|
Nitulescu GM, Van De Venter M, Nitulescu G, Ungurianu A, Juzenas P, Peng Q, Olaru OT, Grădinaru D, Tsatsakis A, Tsoukalas D, Spandidos DA, Margina D. The Akt pathway in oncology therapy and beyond (Review). Int J Oncol 2018; 53:2319-2331. [PMID: 30334567 PMCID: PMC6203150 DOI: 10.3892/ijo.2018.4597] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Protein kinase B (Akt), similar to many other protein kinases, is at the crossroads of cell death and survival, playing a pivotal role in multiple interconnected cell signaling mechanisms implicated in cell metabolism, growth and division, apoptosis suppression and angiogenesis. Akt protein kinase displays important metabolic effects, among which are glucose uptake in muscle and fat cells or the suppression of neuronal cell death. Disruptions in the Akt-regulated pathways are associated with cancer, diabetes, cardiovascular and neurological diseases. The regulation of the Akt signaling pathway renders Akt a valuable therapeutic target. The discovery process of Akt inhibitors using various strategies has led to the identification of inhibitors with great selectivity, low side-effects and toxicity. The usefulness of Akt emerges beyond cancer therapy and extends to other major diseases, such as diabetes, heart diseases, or neurodegeneration. This review presents key features of Akt structure and functions, and presents the progress of Akt inhibitors in regards to drug development, and their preclinical and clinical activity in regards to therapeutic efficacy and safety for patients.
Collapse
Affiliation(s)
- George Mihai Nitulescu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Maryna Van De Venter
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth 6031, South Africa
| | - Georgiana Nitulescu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Anca Ungurianu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Petras Juzenas
- Department of Pathology, Radiumhospitalet, Oslo University Hospital, 0379 Oslo, Norway
| | - Qian Peng
- Department of Pathology, Radiumhospitalet, Oslo University Hospital, 0379 Oslo, Norway
| | - Octavian Tudorel Olaru
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Daniela Grădinaru
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Aristides Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dimitris Tsoukalas
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
43
|
Elsaid MY, Shahi A, Wang AR, Baiu DC, Li C, Werner LR, Singhal S, Hall LT, Weichert JP, Armstrong EA, Bednarz BP, Harari PM, Iyer G, Otto M. Enhanced Radiosensitivity in Solid Tumors using a Tumor-selective Alkyl Phospholipid Ether Analog. Mol Cancer Ther 2018; 17:2320-2328. [PMID: 30108133 DOI: 10.1158/1535-7163.mct-17-0897] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 05/08/2018] [Accepted: 08/10/2018] [Indexed: 11/16/2022]
Abstract
Antitumor alkyl phospholipid (APL) analogs comprise a group of structurally related molecules with remarkable tumor selectivity. Some of these compounds have shown radiosensitizing capabilities. CLR127 is a novel, clinical-grade antitumor APL ether analog, a subtype of synthetic APL broadly targeting cancer cells with limited uptake in normal tissues. The purpose of this study was to investigate the effect of CLR127 to modulate radiation response across several adult and pediatric cancer types in vitro as well as in murine xenograft models of human prostate adenocarcinoma, neuroblastoma, Ewing sarcoma, and rhabdomyosarcoma. In vitro, CLR127 demonstrated selective uptake in cancer cells compared to normal cells. In cancer cells, CLR127 treatment prior to radiation significantly decreased clonogenic survival in vitro, and led to increased radiation-induced double-stranded DNA (dsDNA) breakage compared with radiation alone, which was not observed in normal controls. In animal models, CLR127 effectively increased the antitumor response to fractionated radiotherapy and led to delayed tumor regrowth at potentially clinically achievable doses. In conclusion, our study highlights the ability of CLR127 to increase radiation response in several cancer types. Given almost universal uptake of CLR127 in malignant cells, future research should test whether the observed effects can be extended to other tumor types. Our data provide a strong rationale for clinical testing of CLR127 as a tumor-targeted radiosensitizing agent. Mol Cancer Ther; 17(11); 2320-8. ©2018 AACR.
Collapse
Affiliation(s)
- Mohamed Y Elsaid
- Division of Hematology, Oncology & Bone Marrow Transplant, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ankita Shahi
- Division of Hematology, Oncology & Bone Marrow Transplant, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Albert R Wang
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Dana C Baiu
- Division of Hematology, Oncology & Bone Marrow Transplant, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Chunrong Li
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Lauryn R Werner
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Sorabh Singhal
- Division of Hematology, Oncology & Bone Marrow Transplant, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Lance T Hall
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jamey P Weichert
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Eric A Armstrong
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Bryan P Bednarz
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Gopal Iyer
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mario Otto
- Division of Hematology, Oncology & Bone Marrow Transplant, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
44
|
Luna ACDL, Saraiva GKV, Chierice GO, Hesse H, Maria DA. Antiproliferative and proapoptotic effects of DODAC/synthetic phosphoethanolamine on hepatocellular carcinoma cells. BMC Pharmacol Toxicol 2018; 19:44. [PMID: 29996919 PMCID: PMC6042440 DOI: 10.1186/s40360-018-0225-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 06/20/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Current studies have demonstrated that DODAC/PHO-S (Dioctadecyldimethylammonium Chloride/Synthetic phosphoethanolamine) liposomes induces cytotoxicity in Hepa1c1c7 and B16F10 murine tumor cells, with a higher proportion than PHO-S. Therefore, our aim was to evaluate the potential of DODAC/PHO-S to elucidate the mechanism of cell death whereby the liposomes induces cytotoxicity in hepatocellular carcinoma Hepa1c1c7, compared to the PHO-S alone. METHODS Liposomes (DODAC/PHO-S) were prepared by ultrasonication. The cell cycle phases, protein expression and types of cell's death on Hepa1c1c7 were analyzed by flow cytometry. The internalisation of liposomes, mitochondrial electrical potential and lysosomal stability were also evaluated by confocal laser scanning microscopy. RESULTS After treatment with liposomes (DODAC/PHO-S), we observed a significant increase in the population of Hepa1c1c7 cells experiencing cell cycle arrest in the S and G2/M phases, and this treatment was significantly more effective to promote cell death by apoptosis. There also was a decrease in the mitochondrial electrical potential; changes in the lysosomes; nuclear fragmentation and catastrophic changes in Hepa1c1c7 cells. The liposomes additionally promoted increases in the expression of DR4 receptor, caspases 3 and 8, cytochrome c, p53, p21, p27 and Bax. There was also a decrease in the expression of Bcl-2, cyclin D1, CD90 and CD44 proteins. CONCLUSION The overall results showed that DODAC/PHO-S liposomes were more effective than PHO-S alone, in promoting cytotoxicity Hepa1c1c7 tumor cells, activating the intrinsic and extrinsic pathways of programmed cell death.
Collapse
Affiliation(s)
- Arthur Cássio de Lima Luna
- Department of Biochemistry and Biophysics, Butantan Institute, 1500, Vital Brasil Avenue, Sao Paulo, 05503-900, Brazil. .,Department of Medical Sciences, Medical School, University of Sao Paulo, Sao Paulo, Brazil.
| | | | | | - Henrique Hesse
- Department of Biochemistry and Biophysics, Butantan Institute, 1500, Vital Brasil Avenue, Sao Paulo, 05503-900, Brazil
| | - Durvanei Augusto Maria
- Department of Biochemistry and Biophysics, Butantan Institute, 1500, Vital Brasil Avenue, Sao Paulo, 05503-900, Brazil. .,Department of Medical Sciences, Medical School, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
45
|
Wittkowski KM, Dadurian C, Seybold MP, Kim HS, Hoshino A, Lyden D. Complex polymorphisms in endocytosis genes suggest alpha-cyclodextrin as a treatment for breast cancer. PLoS One 2018; 13:e0199012. [PMID: 29965997 PMCID: PMC6028090 DOI: 10.1371/journal.pone.0199012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 05/17/2018] [Indexed: 02/06/2023] Open
Abstract
Most breast cancer deaths are caused by metastasis and treatment options beyond radiation and cytotoxic drugs, which have severe side effects, and hormonal treatments, which are or become ineffective for many patients, are urgently needed. This study reanalyzed existing data from three genome-wide association studies (GWAS) using a novel computational biostatistics approach (muGWAS), which had been validated in studies of 600-2000 subjects in epilepsy and autism. MuGWAS jointly analyzes several neighboring single nucleotide polymorphisms while incorporating knowledge about genetics of heritable diseases into the statistical method and about GWAS into the rules for determining adaptive genome-wide significance. Results from three independent GWAS of 1000-2000 subjects each, which were made available under the National Institute of Health's "Up For A Challenge" (U4C) project, not only confirmed cell-cycle control and receptor/AKT signaling, but, for the first time in breast cancer GWAS, also consistently identified many genes involved in endo-/exocytosis (EEC), most of which had already been observed in functional and expression studies of breast cancer. In particular, the findings include genes that translocate (ATP8A1, ATP8B1, ANO4, ABCA1) and metabolize (AGPAT3, AGPAT4, DGKQ, LPPR1) phospholipids entering the phosphatidylinositol cycle, which controls EEC. These novel findings suggest scavenging phospholipids as a novel intervention to control local spread of cancer, packaging of exosomes (which prepare distant microenvironment for organ-specific metastases), and endocytosis of β1 integrins (which are required for spread of metastatic phenotype and mesenchymal migration of tumor cells). Beta-cyclodextrins (βCD) have already been shown to be effective in in vitro and animal studies of breast cancer, but exhibits cholesterol-related ototoxicity. The smaller alpha-cyclodextrins (αCD) also scavenges phospholipids, but cannot fit cholesterol. An in-vitro study presented here confirms hydroxypropyl (HP)-αCD to be twice as effective as HPβCD against migration of human cells of both receptor negative and estrogen-receptor positive breast cancer. If the previous successful animal studies with βCDs are replicated with the safer and more effective αCDs, clinical trials of adjuvant treatment with αCDs are warranted. Ultimately, all breast cancer are expected to benefit from treatment with HPαCD, but women with triple-negative breast cancer (TNBC) will benefit most, because they have fewer treatment options and their cancer advances more aggressively.
Collapse
Affiliation(s)
- Knut M. Wittkowski
- Center for Clinical and Translational Science, The Rockefeller University, New York, New York, United States of America
| | - Christina Dadurian
- Center for Clinical and Translational Science, The Rockefeller University, New York, New York, United States of America
| | - Martin P. Seybold
- Institut für Formale Methoden der Informatik, Universität Stuttgart, Stuttgart, Germany
| | - Han Sang Kim
- Department of Pediatrics, and Cell and Developmental Biology Weill Medical College of Cornell University, New York, New York, United States of America
| | - Ayuko Hoshino
- Department of Pediatrics, and Cell and Developmental Biology Weill Medical College of Cornell University, New York, New York, United States of America
| | - David Lyden
- Department of Pediatrics, and Cell and Developmental Biology Weill Medical College of Cornell University, New York, New York, United States of America
| |
Collapse
|
46
|
Knific T, Vouk K, Smrkolj Š, Prehn C, Adamski J, Rižner TL. Models including plasma levels of sphingomyelins and phosphatidylcholines as diagnostic and prognostic biomarkers of endometrial cancer. J Steroid Biochem Mol Biol 2018; 178:312-321. [PMID: 29360580 DOI: 10.1016/j.jsbmb.2018.01.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 01/15/2018] [Indexed: 01/05/2023]
Abstract
In endometrial cancer, biomarkers for preoperative identification of patients with low risk for disease progression would enable stratification according to the extent of surgery needed, and would avoid the complications that can be associated with radical surgery. A panel of proteins, amino acids, enzymes, and miRNA has been investigated as potential biomarkers for endometrial cancer. At the time of the manuscript submission targeted metabolomics/lipidomics approaches have not been applied to biomarker research in endometrial cancer. Using electrospray ionization-tandem mass spectrometry we quantified 163 metabolites in 126 plasma samples (61 patients with endometrial cancer, 65 control patients). Three single phosphatidylcholines were identified with significantly decreased levels in patients with endometrial cancer. A diagnostic model was defined as the ratio between acylcarnitine C16 and phosphatidylcholine PCae C40:1, the ratio between proline and tyrosine, and the ratio between the two phosphatidylcholines PCaa C42:0 and PCae C44:5; which provided sensitivity of 85.25%, specificity of 69.23%, and AUC of 0.837. Addition of smoking status further improved the constructed diagnostic model (AUC = 0.855). The presence of the major prognostic factors of deep myometrial invasion and lymphovascular invasion were also associated with altered metabolite concentrations. A prognostic model for deep myometrial invasion included the ratio between two hydroxysphingomyelins SMOH C14:1 and SMOH C24:1, and the ratio between two phosphatidylcholines PCaa C40:2 and PCaa C42:6, which provided sensitivity of 81.25%, specificity of 86.36%, and AUC of 0.857. The model for lymphovascular invasion included the ratio between two phosphatidylcholines PCaa C34:4 and PCae C38:3, and the ratio between acylcarnitine C16:2 and phosphatidylcholine PCaa C38:1, which provided sensitivity of 88.89%, specificity of 84.31%, and AUC of 0.935.
Collapse
Affiliation(s)
- Tamara Knific
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Katja Vouk
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Špela Smrkolj
- University Medical Centre, Department of Obstetrics and Gynaecology, 1000 Ljubljana, Slovenia
| | - Cornelia Prehn
- Institute of Experimental Genetics, Genome Analysis Centre, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Centre, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Lehrstuhl für Experimentelle Genetik, Technische Universität München, 85350 Freising, Weihenstephan, Germany; German Center for Diabetes Research (DZD), 85764 München, Neuherberg, Germany
| | - Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia.
| |
Collapse
|
47
|
Messias MCF, Mecatti GC, Priolli DG, de Oliveira Carvalho P. Plasmalogen lipids: functional mechanism and their involvement in gastrointestinal cancer. Lipids Health Dis 2018. [PMID: 29514688 PMCID: PMC5842581 DOI: 10.1186/s12944-018-0685-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The plasmalogens are a class of glycerophospholipids which contain a vinyl-ether and an ester bond at the sn-1 and sn-2 positions, respectively, in the glycerol backbone. They constitute 10 mol% of the total mass of phospholipids in humans, mainly as membrane structure components. Plasmalogens are important for the organization and stability of lipid raft microdomains and cholesterol-rich membrane regions involved in cellular signaling. In addition to their structural roles, a subset of ether lipids are thought to function as endogenous antioxidants and emerging studies suggest that they are involved in cell differentiation and signaling pathways. Although the clinical significance of plasmalogens is linked to peroxisomal disorders, the pathophysiological roles and their possible metabolic pathways are not fully understood since they present unique structural attributes for the different tissue types. Studies suggest that changes in plasmalogen metabolism may contribute to the development of various types of cancer. Here, we review the molecular characteristics of plasmalogens in order to significantly increase our understanding of the plasmalogen molecule and its involvement in gastrointestinal cancers as well as other types of cancers.
Collapse
Affiliation(s)
- Márcia Cristina Fernandes Messias
- Laboratory of Multidisciplinary Research, São Francisco University, USF, São Francisco de Assis Avenue, 218, Bragança Paulista, SP, 12916-900, Brazil.
| | - Giovana Colozza Mecatti
- Laboratory of Multidisciplinary Research, São Francisco University, USF, São Francisco de Assis Avenue, 218, Bragança Paulista, SP, 12916-900, Brazil
| | - Denise Gonçalves Priolli
- Laboratory of Multidisciplinary Research, São Francisco University, USF, São Francisco de Assis Avenue, 218, Bragança Paulista, SP, 12916-900, Brazil
| | - Patrícia de Oliveira Carvalho
- Laboratory of Multidisciplinary Research, São Francisco University, USF, São Francisco de Assis Avenue, 218, Bragança Paulista, SP, 12916-900, Brazil.
| |
Collapse
|
48
|
Escribá PV. Membrane-lipid therapy: A historical perspective of membrane-targeted therapies - From lipid bilayer structure to the pathophysiological regulation of cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1493-1506. [PMID: 28577973 DOI: 10.1016/j.bbamem.2017.05.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our current understanding of membrane lipid composition, structure and functions has led to the investigation of their role in cell signaling, both in healthy and pathological cells. As a consequence, therapies based on the regulation of membrane lipid composition and structure have been recently developed. This novel field, known as Membrane Lipid Therapy, is growing and evolving rapidly, providing treatments that are now in use or that are being studied for their application to oncological disorders, Alzheimer's disease, spinal cord injury, stroke, diabetes, obesity, and neuropathic pain. This field has arisen from relevant discoveries on the behavior of membranes in recent decades, and it paves the way to adopt new approaches in modern pharmacology and nutrition. This innovative area will promote further investigation into membranes and the development of new therapies with molecules that target the cell membrane. Due to the prominent roles of membranes in the cells' physiology and the paucity of therapeutic approaches based on the regulation of the lipids they contain, it is expected that membrane lipid therapy will provide new treatments for numerous pathologies. The first on-purpose rationally designed molecule in this field, minerval, is currently being tested in clinical trials and it is expected to enter the market around 2020. However, it seems feasible that during the next few decades other membrane regulators will also be marketed for the treatment of human pathologies. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
- Pablo V Escribá
- Department of Biology, University of the Balearic Islands, E-07122 Palma de Mallorca, Spain.
| |
Collapse
|