1
|
Hunter JJ, Del Valle L, Peruzzi F, Reiss K. Cardiolipin Dysregulation in Glioblastoma-Effects on Mitochondrial Function Tumor Cell Death and Sensitivity to Mitochondria-Targeting Drugs. J Cell Physiol 2025; 240:e70045. [PMID: 40372980 DOI: 10.1002/jcp.70045] [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: 01/07/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/17/2025]
Abstract
Biological systems do not exist in isolation. Analogous to the intricate design of a spider web, the metabolic adaptations propagated by glioblastoma cells are interlaced, creating a "defense mechanism" that increases the likelihood of mutagenesis and proliferation, while mitigating stress-induced tumor cell death and immune evasion. Previous studies have observed the role of cardiolipin (CL) in the electron transport chain (ETC) function and several other intracellular signaling pathways. Our review provides a synopsis of the existing knowledge about CL in glioblastoma and its complex relationship with metabolic reprogramming at the subcellular level. Through a meticulous examination of CL defects due to its biogenesis and stress-induced modifications, we seek to elucidate the multifaceted connections between aberrant CL variants and the metabolic alterations that underlie glioblastoma progression. A comprehensive grasp of these mechanisms could provide future direction in designing chemotherapeutic agents that selectively target glioblastoma, are less harmful to normal cells, and therefore, may extend patient survival.
Collapse
Affiliation(s)
- Jean-Jacques Hunter
- Department of Neuroscience, Tulane University, New Orleans, Louisiana, USA
- Department of Interdisciplinary Oncology, Stanley S Scott Cancer Center, LSU Health Sciences Center, New Orleans, Louisiana, USA
| | - Luis Del Valle
- Department of Pathology, Stanley S Scott Cancer Center, LSU Health Sciences Center, New Orleans, Louisiana, USA
| | - Francesca Peruzzi
- Department of Medicine, Stanley S Scott Cancer Center, LSU Health Sciences Center, New Orleans, Louisiana, USA
| | - Krzysztof Reiss
- Department of Interdisciplinary Oncology, Stanley S Scott Cancer Center, LSU Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
2
|
Milan A, Mioc M, Mioc A, Gogulescu A, Mardale G, Avram Ș, Maksimović T, Mara B, Șoica C. Cytotoxic Potential of Betulinic Acid Fatty Esters and Their Liposomal Formulations: Targeting Breast, Colon, and Lung Cancer Cell Lines. Molecules 2024; 29:3399. [PMID: 39064977 PMCID: PMC11279467 DOI: 10.3390/molecules29143399] [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: 06/19/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Betulinic acid is a lupane-type pentacyclic triterpene mostly found in birch bark and thoroughly explored for its wide range of pharmacological activities. Despite its impressive biological potential, its low bioavailability has challenged many researchers to develop different formulations for achieving better in vitro and in vivo effects. We previously reported the synthesis of fatty acid esters of betulinic acid using butyric, stearic, and palmitic acids (But-BA, St-BA, and Pal-BA) and included them in surfaced-modified liposomes (But-BA-Lip, St-BA-Lip, Pal-BA-Lip). In the current study, we evaluated the cytotoxic effects of both fatty acid esters and their respective liposomal formulations against MCF-7, HT-29, and NCI-H460 cell line. The cytotoxic assessment of BA derivatives revealed that both the fatty esters and their liposomal formulations acted as cytotoxic agents in a dose- and time-dependent manner. But-BA-Lip exerted stronger cytotoxic effects than the parent compound, BA and its liposomal formulation, and even stronger effects than 5-FU against HT-29 cells (IC50 of 30.57 μM) and NCI-H460 cells (IC50 of 30.74 μM). BA's fatty esters and their respective liposomal formulations facilitated apoptosis in cancer cells by inducing nuclear morphological changes and increasing caspase-3/-7 activity. The HET-CAM assay proved that none of the tested compounds induced any irritative effect, suggesting that they can be used safely for local applications.
Collapse
Affiliation(s)
- Andreea Milan
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Marius Mioc
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Alexandra Mioc
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Armand Gogulescu
- Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Gabriel Mardale
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Ștefana Avram
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Tamara Maksimović
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
| | - Bogdan Mara
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Institute of Chemistry Coriolan Drăgulescu, 24 Mihai Viteazu Ave, 300223 Timișoara, Romania
| | - Codruța Șoica
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| |
Collapse
|
3
|
O’Neill KC, Liapis E, Harris BT, Perlin DS, Carter CL. Mass spectrometry imaging discriminates glioblastoma tumor cell subpopulations and different microvascular formations based on their lipid profiles. Sci Rep 2022; 12:17069. [PMID: 36224354 PMCID: PMC9556690 DOI: 10.1038/s41598-022-22093-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 10/10/2022] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma is a prevalent malignant brain tumor and despite clinical intervention, tumor recurrence is frequent and usually fatal. Genomic investigations have provided a greater understanding of molecular heterogeneity in glioblastoma, yet there are still no curative treatments, and the prognosis has remained unchanged. The aggressive nature of glioblastoma is attributed to the heterogeneity in tumor cell subpopulations and aberrant microvascular proliferation. Ganglioside-directed immunotherapy and membrane lipid therapy have shown efficacy in the treatment of glioblastoma. To truly harness these novel therapeutics and develop a regimen that improves clinical outcome, a greater understanding of the altered lipidomic profiles within the glioblastoma tumor microenvironment is urgently needed. In this work, high resolution mass spectrometry imaging was utilized to investigate lipid heterogeneity in human glioblastoma samples. Data presented offers the first insight into the histology-specific accumulation of lipids involved in cell metabolism and signaling. Cardiolipins, phosphatidylinositol, ceramide-1-phosphate, and gangliosides, including the glioblastoma stem cell marker, GD3, were shown to differentially accumulate in tumor and endothelial cell subpopulations. Conversely, a reduction in sphingomyelins and sulfatides were detected in tumor cell regions. Cellular accumulation for each lipid class was dependent upon their fatty acid residue composition, highlighting the importance of understanding lipid structure-function relationships. Discriminating ions were identified and correlated to histopathology and Ki67 proliferation index. These results identified multiple lipids within the glioblastoma microenvironment that warrant further investigation for the development of predictive biomarkers and lipid-based therapeutics.
Collapse
Affiliation(s)
- Kelly C. O’Neill
- grid.429392.70000 0004 6010 5947Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110 USA
| | - Evangelos Liapis
- grid.429392.70000 0004 6010 5947Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110 USA
| | - Brent T. Harris
- grid.411667.30000 0001 2186 0438Departments of Neurology and Pathology, Georgetown University Medical Center, Washington, D.C. 20007 USA
| | - David S. Perlin
- grid.429392.70000 0004 6010 5947Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110 USA ,grid.429392.70000 0004 6010 5947Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ 07110 USA
| | - Claire L. Carter
- grid.429392.70000 0004 6010 5947Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110 USA ,grid.429392.70000 0004 6010 5947Department of Pathology, Hackensack Meridian School of Medicine, Nutley, NJ 07110 USA
| |
Collapse
|
4
|
Vesga-Jiménez DJ, Martin C, Barreto GE, Aristizábal-Pachón AF, Pinzón A, González J. Fatty Acids: An Insight into the Pathogenesis of Neurodegenerative Diseases and Therapeutic Potential. Int J Mol Sci 2022; 23:2577. [PMID: 35269720 PMCID: PMC8910658 DOI: 10.3390/ijms23052577] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/12/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
One of the most common lipids in the human body is palmitic acid (PA), a saturated fatty acid with essential functions in brain cells. PA is used by cells as an energy source, besides being a precursor of signaling molecules and protein tilting across the membrane. Although PA plays physiological functions in the brain, its excessive accumulation leads to detrimental effects on brain cells, causing lipotoxicity. This mechanism involves the activation of toll-like receptors (TLR) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways, with the consequent release of pro-inflammatory cytokines, increased production of reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, and autophagy impairment. Importantly, some of the cellular changes induced by PA lead to an augmented susceptibility to the development of Alzheimer's and Parkinson´s diseases. Considering the complexity of the response to PA and the intrinsic differences of the brain, in this review, we provide an overview of the molecular and cellular effects of PA on different brain cells and their possible relationships with neurodegenerative diseases (NDs). Furthermore, we propose the use of other fatty acids, such as oleic acid or linoleic acid, as potential therapeutic approaches against NDs, as these fatty acids can counteract PA's negative effects on cells.
Collapse
Affiliation(s)
- Diego Julián Vesga-Jiménez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA 30329, USA;
| | - Cynthia Martin
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA 30329, USA;
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland;
- Health Research Institute, University of Limerick, V94 T9PX Limerick, Ireland
| | - Andrés Felipe Aristizábal-Pachón
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
| | - Andrés Pinzón
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogota 111321, Colombia;
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
| |
Collapse
|
5
|
Puca F, Yu F, Bartolacci C, Pettazzoni P, Carugo A, Huang-Hobbs E, Liu J, Zanca C, Carbone F, Del Poggetto E, Gumin J, Dasgupta P, Seth S, Srinivasan S, Lang FF, Sulman EP, Lorenzi PL, Tan L, Shan M, Tolstyka ZP, Kachman M, Zhang L, Gao S, Deem AK, Genovese G, Scaglioni PP, Lyssiotis CA, Viale A, Draetta GF. Medium-Chain Acyl-CoA Dehydrogenase Protects Mitochondria from Lipid Peroxidation in Glioblastoma. Cancer Discov 2021; 11:2904-2923. [PMID: 34039636 PMCID: PMC8711129 DOI: 10.1158/2159-8290.cd-20-1437] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/25/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is highly resistant to chemotherapies, immune-based therapies, and targeted inhibitors. To identify novel drug targets, we screened orthotopically implanted, patient-derived glioblastoma sphere-forming cells using an RNAi library to probe essential tumor cell metabolic programs. This identified high dependence on mitochondrial fatty acid metabolism. We focused on medium-chain acyl-CoA dehydrogenase (MCAD), which oxidizes medium-chain fatty acids (MCFA), due to its consistently high score and high expression among models and upregulation in GBM compared with normal brain. Beyond the expected energetics impairment, MCAD depletion in primary GBM models induced an irreversible cascade of detrimental metabolic effects characterized by accumulation of unmetabolized MCFAs, which induced lipid peroxidation and oxidative stress, irreversible mitochondrial damage, and apoptosis. Our data uncover a novel protective role for MCAD to clear lipid molecules that may cause lethal cell damage, suggesting that therapeutic targeting of MCFA catabolism may exploit a key metabolic feature of GBM. SIGNIFICANCE: MCAD exerts a protective role to prevent accumulation of toxic metabolic by-products in glioma cells, actively catabolizing lipid species that would otherwise affect mitochondrial integrity and induce cell death. This work represents a first demonstration of a nonenergetic role for dependence on fatty acid metabolism in cancer.This article is highlighted in the In This Issue feature, p. 2659.
Collapse
Affiliation(s)
- Francesca Puca
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Fei Yu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Caterina Bartolacci
- Department of Internal Medicine Division of Hematology & Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Piergiorgio Pettazzoni
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alessandro Carugo
- The Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emmet Huang-Hobbs
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jintan Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ciro Zanca
- The Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Federica Carbone
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edoardo Del Poggetto
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pushan Dasgupta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sahil Seth
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sanjana Srinivasan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Erik P Sulman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mengrou Shan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Zachary P Tolstyka
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Maureen Kachman
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, Michigan
| | - Li Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Sisi Gao
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Angela K Deem
- The Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pier Paolo Scaglioni
- Department of Internal Medicine Division of Hematology & Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
6
|
Schönfeld P, Reiser G. How the brain fights fatty acids' toxicity. Neurochem Int 2021; 148:105050. [PMID: 33945834 DOI: 10.1016/j.neuint.2021.105050] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/24/2022]
Abstract
Neurons spurn hydrogen-rich fatty acids for energizing oxidative ATP synthesis, contrary to other cells. This feature has been mainly attributed to a lower yield of ATP per reduced oxygen, as compared to glucose. Moreover, the use of fatty acids as hydrogen donor is accompanied by severe β-oxidation-associated ROS generation. Neurons are especially susceptible to detrimental activities of ROS due to their poor antioxidative equipment. It is also important to note that free fatty acids (FFA) initiate multiple harmful activities inside the cells, particularly on phosphorylating mitochondria. Several processes enhance FFA-linked lipotoxicity in the cerebral tissue. Thus, an uptake of FFA from the circulation into the brain tissue takes place during an imbalance between energy intake and energy expenditure in the body, a situation similar to that during metabolic syndrome and fat-rich diet. Traumatic or hypoxic brain injuries increase hydrolytic degradation of membrane phospholipids and, thereby elevate the level of FFA in neural cells. Accumulation of FFA in brain tissue is markedly associated with some inherited neurological disorders, such as Refsum disease or X-linked adrenoleukodystrophy (X-ALD). What are strategies protecting neurons against FFA-linked lipotoxicity? Firstly, spurning the β-oxidation pathway in mitochondria of neurons. Secondly, based on a tight metabolic communication between neurons and astrocytes, astrocytes donate metabolites to neurons for synthesis of antioxidants. Further, neuronal autophagy of ROS-emitting mitochondria combined with the transfer of degradation-committed FFA for their disposal in astrocytes, is a potent protective strategy against ROS and harmful activities of FFA. Finally, estrogens and neurosteroids are protective as triggers of ERK and PKB signaling pathways, consequently initiating the expression of various neuronal survival genes via the formation of cAMP response element-binding protein (CREB).
Collapse
Affiliation(s)
- Peter Schönfeld
- Institut für Biochemie und Zellbiologie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120, Magdeburg, Germany
| | - Georg Reiser
- Institut für Inflammation und Neurodegeneration (Neurobiochemie), Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120, Magdeburg, Germany.
| |
Collapse
|
7
|
Linke C, Wösle M, Harder A. Anti-cancer agent 3-bromopyruvate reduces growth of MPNST and inhibits metabolic pathways in a representative in-vitro model. BMC Cancer 2020; 20:896. [PMID: 32948135 PMCID: PMC7501688 DOI: 10.1186/s12885-020-07397-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/09/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Anticancer compound 3-bromopyruvate (3-BrPA) suppresses cancer cell growth via targeting glycolytic and mitochondrial metabolism. The malignant peripheral nerve sheath tumor (MPNST), a very aggressive, therapy resistant, and Neurofibromatosis type 1 associated neoplasia, shows a high metabolic activity and affected patients may therefore benefit from 3-BrPA treatment. To elucidate the specific mode of action, we used a controlled cell model overexpressing proteasome activator (PA) 28, subsequently leading to p53 inactivation and oncogenic transformation and therefore reproducing an important pathway in MPNST and overall tumor pathogenesis. METHODS Viability of MPNST cell lines S462, NSF1, and T265 in response to increasing doses (0-120 μM) of 3-BrPA was analyzed by CellTiter-Blue® assay. Additionally, we investigated viability, reactive oxygen species (ROS) production (dihydroethidium assay), nicotinamide adenine dinucleotide dehydrogenase activity (NADH-TR assay) and lactate production (lactate assay) in mouse B8 fibroblasts overexpressing PA28 in response to 3-BrPA application. For all experiments normal and nutrient deficient conditions were tested. MPNST cell lines were furthermore characterized immunohistochemically for Ki67, p53, bcl2, bcl6, cyclin D1, and p21. RESULTS MPNST significantly responded dose dependent to 3-BrPA application, whereby S462 cells were most responsive. Human control cells showed a reduced sensitivity. In PA28 overexpressing cancer cell model 3-BrPA application harmed mitochondrial NADH dehydrogenase activity mildly and significantly failed to inhibit lactate production. PA28 overexpression was associated with a functional glycolysis as well as a partial resistance to stress provoked by nutrient deprivation. 3-BrPA treatment was not associated with an increase of ROS. Starvation sensitized MPNST to treatment. CONCLUSIONS Aggressive MPNST cells are sensitive to 3-BrPA therapy in-vitro with and without starvation. In a PA28 overexpression cancer cell model leading to p53 inactivation, thereby reflecting a key molecular feature in human NF1 associated MPNST, known functions of 3-BrPA to block mitochondrial activity and glycolysis were reproduced, however oncogenic cells displayed a partial resistance. To conclude, 3-BrPA was sufficient to reduce NF1 associated MPNST viability potentially due inhibition of glycolysis which should lead to the initiation of further studies and promises a potential benefit for NF1 patients.
Collapse
Affiliation(s)
- Christian Linke
- Faculty of Health Sciences, joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Brandenburg an der Havel, Germany
| | - Markus Wösle
- Clinic for Radiotherapy and Radiation Oncology, Dessau City Hospital, Dessau-Roßlau, Germany
| | - Anja Harder
- Faculty of Health Sciences, joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Brandenburg an der Havel, Germany. .,Institute of Neuropathology, University Hospital Münster, Münster, Germany. .,Institute of Pathology, Brandenburg Medical School Theodor Fontane, Dessau City Hospital, Auenweg 38, 06847, Dessau-Roßlau, Germany.
| |
Collapse
|
8
|
Tibolone Ameliorates the Lipotoxic Effect of Palmitic Acid in Normal Human Astrocytes. Neurotox Res 2020; 38:585-595. [DOI: 10.1007/s12640-020-00247-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
|
9
|
El-Hafidi M, Correa F, Zazueta C. Mitochondrial dysfunction in metabolic and cardiovascular diseases associated with cardiolipin remodeling. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165744. [PMID: 32105822 DOI: 10.1016/j.bbadis.2020.165744] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/21/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Cardiolipin (CL) is an acidic phospholipid almost exclusively found in the inner mitochondrial membrane, that not only stabilizes the structure and function of individual components of the mitochondrial electron transport chain, but regulates relevant mitochondrial processes, like mitochondrial dynamics and cristae structure maintenance among others. Alterations in CL due to peroxidation, correlates with loss of such mitochondrial activities and disease progression. In this review it is recapitulated the current state of knowledge of the role of cardiolipin remodeling associated with mitochondrial dysfunction in metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Mohammed El-Hafidi
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología I. Ch. 14080, Ciudad de México, México
| | - Francisco Correa
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología I. Ch. 14080, Ciudad de México, México
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología I. Ch. 14080, Ciudad de México, México.
| |
Collapse
|
10
|
Ting HC, Chen LT, Chen JY, Huang YL, Xin RC, Chan JF, Hsu YHH. Double bonds of unsaturated fatty acids differentially regulate mitochondrial cardiolipin remodeling. Lipids Health Dis 2019; 18:53. [PMID: 30764880 PMCID: PMC6376731 DOI: 10.1186/s12944-019-0990-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/28/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Supplemented fatty acids can incorporate into cardiolipin (CL) and affect its remodeling. The change in CL species may alter the mitochondrial membrane composition, potentially disturbing the mitochondrial structure and function during inflammation. METHOD To investigate the effect of the unsaturation of fatty acids on CL, we supplemented macrophage-like RAW264.7 cells with 18-carbon unsaturated fatty acids including oleic acid (OA, 18:1), linoleic acid (LA, 18:2), α-linolenic acid (ALA, 18:3), γ-linolenic acid (GLA, 18:3), and stearidonic acid (SDA, 18:4). Mitochondrial changes in CL were measured through mass spectrometry. RESULT Our data indicated that OA(18:1) was the most efficient fatty acid that incorporated into CL, forming symmetrical CL without fatty acid elongation and desaturation. In addition, LA(18:2) and ALA(18:3) were further elongated before incorporation, significantly increasing the number of double bonds and the chain length of CL. GLA and SDA were not optimal substrates for remodeling enzymes. The findings of RT-qPCR experiments revealed that none of these changes in CL occurred through the regulation of CL remodeling- or synthesis-related genes. The fatty acid desaturase and transportation genes-Fads2 and Cpt1a, respectively-were differentially regulated by the supplementation of five unsaturated 18-carbon fatty acids. CONCLUSIONS The process of fatty acid incorporation to CL was regulated by the fatty acid desaturation and transportation into mitochondria in macrophage. The double bonds of fatty acids significantly affect the incorporation process and preference. Intact OA(18:1) was incorporated to CL; LA(18:2) and ALA(18:3) were desaturated and elongated to long chain fatty acid before the incorporation; GLA(18:3) and SDA(18:4) were unfavorable for the CL incorporation.
Collapse
Affiliation(s)
- Hsiu-Chi Ting
- Department of Chemistry, Tunghai University, No.1727, Sec4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan, Republic of China
| | - Li-Tzu Chen
- Department of Chemistry, Tunghai University, No.1727, Sec4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan, Republic of China
| | - Jo-Yu Chen
- Department of Chemistry, Tunghai University, No.1727, Sec4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan, Republic of China
| | - Yi-Li Huang
- Department of Chemistry, Tunghai University, No.1727, Sec4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan, Republic of China
| | - Rui-Cheng Xin
- Department of Chemistry, Tunghai University, No.1727, Sec4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan, Republic of China
| | - Jui-Fen Chan
- Department of Chemistry, Tunghai University, No.1727, Sec4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan, Republic of China
| | - Yuan-Hao Howard Hsu
- Department of Chemistry, Tunghai University, No.1727, Sec4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan, Republic of China. .,Life Science Research Center, Tunghai University, No.1727, Sec4, Taiwan Boulevard, Xitun District, Taichung, 40704, Taiwan, Republic of China.
| |
Collapse
|
11
|
Macchioni L, Petricciuolo M, Davidescu M, Fettucciari K, Scarpelli P, Vitale R, Gatticchi L, Orvietani PL, Marchegiani A, Marconi P, Bassotti G, Corcelli A, Corazzi L. Palmitate lipotoxicity in enteric glial cells: Lipid remodeling and mitochondrial ROS are responsible for cyt c release outside mitochondria. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:895-908. [PMID: 29729479 DOI: 10.1016/j.bbalip.2018.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/05/2018] [Accepted: 04/29/2018] [Indexed: 02/08/2023]
Abstract
Enteric glial cells (EGCs) are components of the enteric nervous system, an organized structure that controls gut functions. EGCs may be vulnerable to different agents, such as bacterial infections that could alter the intestinal epithelial barrier, allowing bacterial toxins and/or other agents possessing intrinsic toxic effect to access cells. Palmitate, known to exhibit lipotoxicity, is released in the gut during the digestion process. In this study, we investigated the lipotoxic effect of palmitate in cultured EGCs, with particular emphasis on palmitate-dependent intracellular lipid remodeling. Palmitate but not linoleate altered mitochondrial and endoplasmic reticulum lipid composition. In particular, the levels of phosphatidic acid, key precursor of phospholipid synthesis, increased, whereas those of mitochondrial cardiolipin (CL) decreased; in parallel, phospholipid remodeling was induced. CL remodeling (chains shortening and saturation) together with palmitate-triggered mitochondrial burst, caused cytochrome c (cyt c) detachment from its CL anchor and accumulation in the intermembrane space as soluble pool. Palmitate decreased mitochondrial membrane potential and ATP levels, without mPTP opening. Mitochondrial ROS permeation into the cytosol and palmitate-induced ER stress activated JNK and p38, culminating in Bim and Bax overexpression, factors known to increase the outer mitochondrial membrane permeability. Overall, in EGCs palmitate produced weakening of cyt c-CL interactions and favoured the egress of the soluble cyt c pool outside mitochondria to trigger caspase-3-dependent viability loss. Elucidating the mechanisms of palmitate lipotoxicity in EGCs may be relevant in gut pathological conditions occurring in vivo such as those following an insult that may damage the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Lara Macchioni
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Maya Petricciuolo
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Magdalena Davidescu
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Katia Fettucciari
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Paolo Scarpelli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Rita Vitale
- Department of Basic Medical Sciences, Neuroscience and Sense. Organs, University of Bari "A. Moro", 70124 Bari, Italy
| | - Leonardo Gatticchi
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Pier Luigi Orvietani
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica (MC), Italy
| | | | - Gabrio Bassotti
- Department of Medicine, University of Perugia, 06132 Perugia, Italy
| | - Angela Corcelli
- Department of Basic Medical Sciences, Neuroscience and Sense. Organs, University of Bari "A. Moro", 70124 Bari, Italy
| | - Lanfranco Corazzi
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| |
Collapse
|
12
|
Macchioni L, Davidescu M, Fettucciari K, Petricciuolo M, Gatticchi L, Gioè D, Villanacci V, Bellini M, Marconi P, Roberti R, Bassotti G, Corazzi L. Enteric glial cells counteract Clostridium difficile Toxin B through a NADPH oxidase/ROS/JNK/caspase-3 axis, without involving mitochondrial pathways. Sci Rep 2017; 7:45569. [PMID: 28349972 PMCID: PMC5368562 DOI: 10.1038/srep45569] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/27/2017] [Indexed: 02/06/2023] Open
Abstract
Enteric glial cells (EGCs) are components of the intestinal epithelial barrier essential for regulating the enteric nervous system. Clostridium difficile is the most common cause of antibiotic-associated colitis, toxin B (TcdB) being the major virulence factor, due to its ability to breach the intestinal epithelial barrier and to act on other cell types. Here we investigated TcdB effects on EGCs and the activated molecular mechanisms. Already at 2 hours, TcdB triggered ROS formation originating from NADPH-oxidase, as demonstrated by their reduction in the presence of the NADPH-oxidase inhibitor ML171. Although EGCs mitochondria support almost completely the cellular ATP need, TcdB exerted weak effects on EGCs in terms of ATP and mitochondrial functionality, mitochondrial ROS production occurring as a late event. ROS activated the JNK signalling and overexpression of the proapoptotic Bim not followed by cytochrome c or AIF release to activate the downstream apoptotic cascade. EGCs underwent DNA fragmentation through activation of the ROS/JNK/caspase-3 axis, evidenced by the ability of ML171, N-acetylcysteine, and the JNK inhibitor SP600125 to inhibit caspase-3 or to contrast apoptosis. Therefore, TcdB aggressiveness towards EGCs is mainly restricted to the cytosolic compartment, which represents a peculiar feature, since TcdB primarily influences mitochondria in other cellular types.
Collapse
Affiliation(s)
- Lara Macchioni
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Magdalena Davidescu
- Scientific and educational center of Terni, University of Perugia, Perugia, Italy
| | - Katia Fettucciari
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maya Petricciuolo
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Leonardo Gatticchi
- Scientific and educational center of Terni, University of Perugia, Perugia, Italy
| | - Davide Gioè
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Massimo Bellini
- Department of Gastroenterology, University of Pisa, Pisa, Italy
| | | | - Rita Roberti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Gabrio Bassotti
- Department of Medicine, University of Perugia, Perugia, Italy
| | - Lanfranco Corazzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
13
|
Vos M, Geens A, Böhm C, Deaulmerie L, Swerts J, Rossi M, Craessaerts K, Leites EP, Seibler P, Rakovic A, Lohnau T, De Strooper B, Fendt SM, Morais VA, Klein C, Verstreken P. Cardiolipin promotes electron transport between ubiquinone and complex I to rescue PINK1 deficiency. J Cell Biol 2017; 216:695-708. [PMID: 28137779 PMCID: PMC5346965 DOI: 10.1083/jcb.201511044] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 11/25/2016] [Accepted: 01/05/2017] [Indexed: 02/08/2023] Open
Abstract
Parkinson’s disease–causing mutations in PINK1 yield mitochondrial defects including inefficient electron transport between complex I and ubiquinone. Vos et al. show that genetic and pharmacological inhibition of fatty acid synthase bypass these complex I defects in fly, mouse, and human Parkinson’s disease models. PINK1 is mutated in Parkinson’s disease (PD), and mutations cause mitochondrial defects that include inefficient electron transport between complex I and ubiquinone. Neurodegeneration is also connected to changes in lipid homeostasis, but how these are related to PINK1-induced mitochondrial dysfunction is unknown. Based on an unbiased genetic screen, we found that partial genetic and pharmacological inhibition of fatty acid synthase (FASN) suppresses toxicity induced by PINK1 deficiency in flies, mouse cells, patient-derived fibroblasts, and induced pluripotent stem cell–derived dopaminergic neurons. Lower FASN activity in PINK1 mutants decreases palmitate levels and increases the levels of cardiolipin (CL), a mitochondrial inner membrane–specific lipid. Direct supplementation of CL to isolated mitochondria not only rescues the PINK1-induced complex I defects but also rescues the inefficient electron transfer between complex I and ubiquinone in specific mutants. Our data indicate that genetic or pharmacologic inhibition of FASN to increase CL levels bypasses the enzymatic defects at complex I in a PD model.
Collapse
Affiliation(s)
- Melissa Vos
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium.,Department of Neurosciences and Leuven Research Institute for Neurodegenerative Disease, KU Leuven, 3000 Leuven, Belgium.,Institute of Neurogenetics, University of Luebeck, 23562 Luebeck, Germany
| | - Ann Geens
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium.,Department of Neurosciences and Leuven Research Institute for Neurodegenerative Disease, KU Leuven, 3000 Leuven, Belgium
| | - Claudia Böhm
- Institute of Neurogenetics, University of Luebeck, 23562 Luebeck, Germany
| | - Liesbeth Deaulmerie
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium.,Department of Neurosciences and Leuven Research Institute for Neurodegenerative Disease, KU Leuven, 3000 Leuven, Belgium
| | - Jef Swerts
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium.,Department of Neurosciences and Leuven Research Institute for Neurodegenerative Disease, KU Leuven, 3000 Leuven, Belgium
| | - Matteo Rossi
- VIB Center for Cancer Biology, 3000 Leuven, Belgium.,Department of Oncology and Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium.,Department of Neurosciences and Leuven Research Institute for Neurodegenerative Disease, KU Leuven, 3000 Leuven, Belgium
| | - Elvira P Leites
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649 Lisboa, Portugal
| | - Philip Seibler
- Institute of Neurogenetics, University of Luebeck, 23562 Luebeck, Germany
| | - Aleksandar Rakovic
- Institute of Neurogenetics, University of Luebeck, 23562 Luebeck, Germany
| | - Thora Lohnau
- Institute of Neurogenetics, University of Luebeck, 23562 Luebeck, Germany
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium.,Department of Neurosciences and Leuven Research Institute for Neurodegenerative Disease, KU Leuven, 3000 Leuven, Belgium
| | - Sarah-Maria Fendt
- VIB Center for Cancer Biology, 3000 Leuven, Belgium.,Department of Oncology and Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium
| | - Vanessa A Morais
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium.,Department of Neurosciences and Leuven Research Institute for Neurodegenerative Disease, KU Leuven, 3000 Leuven, Belgium.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649 Lisboa, Portugal
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, 23562 Luebeck, Germany
| | - Patrik Verstreken
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium .,Department of Neurosciences and Leuven Research Institute for Neurodegenerative Disease, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
14
|
3-Bromopyruvate treatment induces alterations of metabolic and stress-related pathways in glioblastoma cells. J Proteomics 2017; 152:329-338. [DOI: 10.1016/j.jprot.2016.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 12/27/2022]
|
15
|
Matsuzaki W, Takahashi H, Nakamura H, Murayama T. Effects of Glycerophospholipids on Ceramide Kinase Activity: Cardiolipin-Affected Cellular Formation of Ceramide-1-phosphate. Biol Pharm Bull 2016; 39:1708-1717. [DOI: 10.1248/bpb.b16-00535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Wataru Matsuzaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Hiromasa Takahashi
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University
| |
Collapse
|
16
|
Li F, Chen X, Xu B, Zhou H. Curcumin induces p53-independent necrosis in H1299 cells via a mitochondria-associated pathway. Mol Med Rep 2015; 12:7806-14. [PMID: 26460892 DOI: 10.3892/mmr.2015.4395] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 09/09/2015] [Indexed: 11/05/2022] Open
Abstract
Curcumin has been shown to have various therapeutic and/or adjuvant therapeutic effects on human cancers, as it inhibits cancer cell proliferation and induces apoptosis through p53-dependent molecular pathways. However, numerous cancer cell types bear a mutant p53 gene, and whether curcumin has any therapeutic effects on p53-deficient/mutant cancer cells has remained elusive. The present study sought to determine whether curcumin exerts any anti-proliferative and cytotoxic effects on the p53-deficient H1299 human lung cancer cell line via a p53-independent mechanism. An MTT assay and flow cytometric analysis indicated that curcumin significantly decreased cell proliferation and induced necrotic cell death. Western blot analysis of the cytosolic and mitochondrial fractions of H1299 cells as well as a fluorometric caspase assay indicated that curcumin-induced necrosis was mitochondria- and caspase-dependent, and resulted in cytochrome c release. Of note, this necrotic cell death was reduced following inhibition of B-cell lymphoma‑2 (Bcl-2)‑associated X protein (Bax) or Bcl‑2 homologous antagonist killer (Bak) as well as overexpression of Bcl-2. In conclusion, the present study suggested that curcumin-induced necrotic cell death was mediated via a p53-independent molecular pathway, which was associated with Bax and Bak translocation, caspase activation and cytochrome c release.
Collapse
Affiliation(s)
- Feie Li
- Department of Pharmacy, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xi Chen
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Bing Xu
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hua Zhou
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
17
|
Davidescu M, Macchioni L, Scaramozzino G, Cristina Marchetti M, Migliorati G, Vitale R, Corcelli A, Roberti R, Castigli E, Corazzi L. The energy blockers bromopyruvate and lonidamine lead GL15 glioblastoma cells to death by different p53-dependent routes. Sci Rep 2015; 5:14343. [PMID: 26387611 PMCID: PMC4585687 DOI: 10.1038/srep14343] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 08/26/2015] [Indexed: 12/29/2022] Open
Abstract
The energy metabolism of tumor cells relies on aerobic glycolysis rather than mitochondrial oxidation. This difference between normal and cancer cells provides a biochemical basis for new therapeutic strategies aimed to block the energy power plants of cells. The effects produced by the energy blockers bromopyruvate (3BP) and lonidamine (LND) and the underlying biochemical mechanisms were investigated in GL15 glioblastoma cells. 3BP exerts early effects compared to LND, even though both drugs lead cells to death but by different routes. A dramatic decrease of ATP levels occurred after 1 hour treatment with 3BP, followed by cytochrome c and hexokinase II degradation, and by the decrease of both LC3I/LC3II ratio and p62, markers of an autophagic flux. In addition, Akt(Ser(473)) and p53(Ser(15)/Ser(315)) dephosphorylation occurred. In LND treatment, sustained ATP cellular levels were maintained up to 40 hours. The autophagic response of cells was overcome by apoptosis that was preceded by phosphatidylinositol disappearance and pAkt decrease. This last event favored p53 translocation to mitochondria triggering a p53-dependent apoptotic route, as observed at 48 and 72 hours. Adversely, in 3BP treatment, phospho-p53 dephosphorylation targeted p53 to MDM2-dependent proteolysis, thus channeling cells to irreversible autophagy.
Collapse
Affiliation(s)
| | - Lara Macchioni
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Gaetano Scaramozzino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | | | | | - Rita Vitale
- IMM-CNR, Institute for Microelectronics and Microsystems, National Research Council, Lecce, Italy
| | - Angela Corcelli
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari "A. Moro", Bari, Italy
| | - Rita Roberti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Emilia Castigli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Lanfranco Corazzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
18
|
Gunaratnam K, Vidal C, Boadle R, Thekkedam C, Duque G. Mechanisms of palmitate-induced cell death in human osteoblasts. Biol Open 2013; 2:1382-9. [PMID: 24285710 PMCID: PMC3863423 DOI: 10.1242/bio.20136700] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lipotoxicity is an overload of lipids in non-adipose tissues that affects function and induces cell death. Lipotoxicity has been demonstrated in bone cells in vitro using osteoblasts and adipocytes in coculture. In this condition, lipotoxicity was induced by high levels of saturated fatty acids (mostly palmitate) secreted by cultured adipocytes acting in a paracrine manner. In the present study, we aimed to identify the underlying mechanisms of lipotoxicity in human osteoblasts. Palmitate induced autophagy in cultured osteoblasts, which was preceded by the activation of autophagosomes that surround palmitate droplets. Palmitate also induced apoptosis though the activation of the Fas/Jun kinase (JNK) apoptotic pathway. In addition, osteoblasts could be protected from lipotoxicity by inhibiting autophagy with the phosphoinositide kinase inhibitor 3-methyladenine or by inhibiting apoptosis with the JNK inhibitor SP600125. In summary, we have identified two major molecular mechanisms of lipotoxicity in osteoblasts and in doing so we have identified a new potential therapeutic approach to prevent osteoblast dysfunction and death, which are common features of age-related bone loss and osteoporosis.
Collapse
Affiliation(s)
- Krishanthi Gunaratnam
- Ageing Bone Research Program, Sydney Medical School Nepean, The University of Sydney, Penrith, NSW 2750, Australia
| | | | | | | | | |
Collapse
|
19
|
Elschami M, Scherr M, Philippens B, Gerardy-Schahn R. Reduction of STAT3 expression induces mitochondrial dysfunction and autophagy in cardiac HL-1 cells. Eur J Cell Biol 2012; 92:21-9. [PMID: 23102833 DOI: 10.1016/j.ejcb.2012.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 09/18/2012] [Accepted: 09/20/2012] [Indexed: 10/27/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is an important mediator of cardiac survival pathways. Reduced levels of STAT3 in patients with end-stage heart failure suggest a clinical relevance of STAT3 deficiency for cardiac disease. The recent identification of STAT3 as a mitochondrial protein which is important for full activity of mitochondrial complex I has opened a new field for the investigation of how STAT3 functions in cardioprotection. The goal of this study was to establish a cell culture model with a reduced STAT3 expression, and to use this model for the investigation of mitochondrial and mitochondrial-associated functions under STAT3 deficiency. In the murine cardiomyogenic cell line HL-1, the expression of STAT3 was silenced by lentiviral transduction with anti-STAT3 shRNA (STAT3 KD cells). STAT3 mRNA and protein levels were significantly reduced in HL-1 STAT3 KD cells compared to HL-1 cells transduced with a control shRNA. Spectrophotometric and polarographic assays with mitochondrial enriched fractions and intact cells showed reduced activities of respiratory chain complexes I, II, III and IV in HL-1 STAT3 KD cells. At ultrastructural level, a severe damage of mitochondrial integrity was observed, combined with a significant increase in autophagolysosomes in STAT3-deficient HL-1 cells. Our results demonstrate that the HL-1 STAT3 KD cell line is a good model to study cellular consequences of STAT3 deficiency. Moreover, this is the first study to show that STAT3 deficiency leads to a disruption of mitochondrial ultrastructure and increased autophagy.
Collapse
Affiliation(s)
- Myriam Elschami
- Institute for Cellular Chemistry, Hannover Medical School, Carl-Neubergstr. 1, D-30625 Hannover, Germany.
| | | | | | | |
Collapse
|
20
|
Davidescu M, Sciaccaluga M, Macchioni L, Angelini R, Lopalco P, Rambotti MG, Roberti R, Corcelli A, Castigli E, Corazzi L. Bromopyruvate mediates autophagy and cardiolipin degradation to monolyso-cardiolipin in GL15 glioblastoma cells. J Bioenerg Biomembr 2012; 44:51-60. [PMID: 22318357 DOI: 10.1007/s10863-012-9411-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 01/18/2012] [Indexed: 01/16/2023]
Abstract
The GL15 glioblastoma cell line undergoes viability loss upon treatment with bromopyruvate. The biochemical mechanisms triggered by the antiglycolytic agent indicate the activation of an autophagic pathway. Acridine orange stains acidic intracellular vesicles already 60 min after bromopyruvate treatment, whereas autophagosomes engulfing electron dense material are well evidenced 18 h later. The autophagic process is accompanied by the expression of the early autophagosomal marker Atg5 and by LC3-II formation, a late biochemical marker associated with autophagosomes. In agreement with the autophagic route activation, the inhibitory and the activator Akt and ERK signaling pathways are depressed and enhanced, respectively. In spite of the energetic collapse suffered by bromopyruvate-treated cells, MALDI-TOF mass spectrometry lipid analysis does not evidence a decrease of the major phospholipids, in accordance with the need of phospholipids for autophagosomal membranes biogenesis. Contrarily, mitochondrial cardiolipin decreases, accompanied by monolyso-cardiolipin formation and complete cytochrome c degradation, events that could target mitochondria to autophagy. However, in our experimental conditions cytochrome c degradation seems to be independent of the autophagic process.
Collapse
Affiliation(s)
- Magdalena Davidescu
- Department of Internal Medicine, Laboratory of Biochemistry, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kim J, Park YJ, Jang Y, Kwon YH. AMPK activation inhibits apoptosis and tau hyperphosphorylation mediated by palmitate in SH-SY5Y cells. Brain Res 2011; 1418:42-51. [PMID: 21937027 DOI: 10.1016/j.brainres.2011.08.059] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 08/16/2011] [Accepted: 08/23/2011] [Indexed: 11/25/2022]
Abstract
Obesity and diabetes have been shown to be associated with cognitive impairment or early neurodegeneration. However, the cellular mechanisms that link between these two pathologies have not been clarified. In this study, we treated SH-SY5Y human neuroblastoma cells with palmitate and observed its effect on cell apoptosis and tau hyperphosphorylation. Dose- and time-dependent effects of palmitate on apoptosis were observed. Palmitate treatment induced endoplasmic reticulum (ER) stress, determined by the expression of spliced X-box binding protein 1 (XBP-1) mRNA and immunoglobin heavy chain-binding protein (BiP). We also observed increases in c-Jun N-terminal kinase (JNK) activation and tau hyperphosphorylation in response to palmitate. Although palmitate did not impair insulin signaling as shown by the immunoblotting analysis of AKT phosphorylation, it did inactivate AMP-activated protein kinase (AMPK). Activation of AMPK by N(1)-(β-d-Ribofuranosyl)-5-aminoimidazole-4-carboxamide (AICAR), significantly reduced the apoptosis of cells treated with palmitate. AICAR also significantly inhibited ER stress, resulting in reduced tau hyperphosphorylation in cells treated with palmitate. Similarly, A769662, a direct activator of AMPK, also abolished the ER stress-mediated apoptosis and tau hyperphosphorylation. Therefore, these data suggest that palmitate triggers ER stress-mediated lipotoxicity and that AMPK activation inhibits apoptosis and tau hyperphosphorylation mediated by palmitate in SH-SY5Y cells.
Collapse
Affiliation(s)
- Juhae Kim
- Department of Food and Nutrition, Seoul National University, 599 Gwanak-ro, Gwanak-Gu, Seoul, 151-742, Republic of Korea
| | | | | | | |
Collapse
|
22
|
Macchioni L, Davidescu M, Sciaccaluga M, Marchetti C, Migliorati G, Coaccioli S, Roberti R, Corazzi L, Castigli E. Mitochondrial dysfunction and effect of antiglycolytic bromopyruvic acid in GL15 glioblastoma cells. J Bioenerg Biomembr 2011; 43:507-18. [PMID: 21833601 DOI: 10.1007/s10863-011-9375-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Accepted: 06/21/2011] [Indexed: 02/06/2023]
Abstract
Most cancer cells, including GL15 glioblastoma cells, rely on glycolysis for energy supply. The effect of antiglycolytic bromopyruvate on respiratory parameters and viability of GL15 cells was investigated. Bromopyruvate caused Δψ(m) and MTT collapse, ATP decrease, and cell viability loss without involving apoptotic or necrotic pathways. The autophagy marker LC3-II was increased. Δψ(m) decrease was accompanied by reactive oxygen species (ROS) increase and cytochrome c (cyt c) disappearance, suggesting a link between free radical generation and intramitochondrial cyt c degradation. Indeed, the free radical inducer menadione caused a decrease in cyt c that was reversed by N-acetylcysteine. Cyt c is tightly bound to the inner mitochondrial membrane in GL15 cells, which may confer protein peroxidase activity, resulting in auto-oxidation and protein targeting to degradation in the presence of ROS. This process is directed towards impairment of the apoptotic cyt c cascade, although cells are committed to die.
Collapse
Affiliation(s)
- Lara Macchioni
- Department of Internal Medicine, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Macchioni L, Corazzi T, Davidescu M, Francescangeli E, Roberti R, Corazzi L. Cytochrome c redox state influences the binding and release of cytochrome c in model membranes and in brain mitochondria. Mol Cell Biochem 2010; 341:149-57. [PMID: 20352475 DOI: 10.1007/s11010-010-0446-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2009] [Accepted: 03/16/2010] [Indexed: 11/28/2022]
Abstract
Cytochrome c (cyt c), a component of the respiratory chain, promotes apoptosis when released into the cytosol. Cyt c anchorage within mitochondria depends on cardiolipin (CL). Detachment and release have been related to CL loss and peroxidation. We report that NaN(3)-dependent complex IV inhibition, accompanied by impairment of respiration, resulted in cyt c release. Contrarily, inhibition of respiration upstream cyt c with complex I and III inhibitors was not accompanied by the release of the protein, despite CL decrease and monolyso-CL increase. No CL changes and H(2)O(2) formation were observed by inhibiting complex IV. In cyt c-CL liposomes, breaching cyt c-CL hydrophilic interactions produced a higher release of the reduced, compared to the oxidized form, suggesting that the hydrophobic component of cyt c-CL binding is prevalent in the oxidized form. Free or liposome-reconstituted cyt c was able to form fatty acid-protein complexes (palmitate < linoleate < oleate) only in its reduced form. We hypothesize that reduced cyt c-fatty acid binding favors the dislocation of the protein from anchoring CL. A mechanism for cyt c release independent of CL peroxidation by H(2)O(2) is feasible. It could weaken the hydrophobic component of cyt c-CL interactions and might function following complex IV inhibition or in oxygen lack, both conditions producing accumulation of reduced cyt c and free fatty acids.
Collapse
Affiliation(s)
- Lara Macchioni
- Department of Internal Medicine, Laboratory of Biochemistry, University of Perugia, via del Giochetto, 06122 Perugia, Italy
| | | | | | | | | | | |
Collapse
|
24
|
Dell'Anna ML, Ottaviani M, Bellei B, Albanesi V, Cossarizza A, Rossi L, Picardo M. Membrane lipid defects are responsible for the generation of reactive oxygen species in peripheral blood mononuclear cells from vitiligo patients. J Cell Physiol 2010; 223:187-93. [PMID: 20049874 DOI: 10.1002/jcp.22027] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The pathogenesis of vitiligo, an acquired depigmenting disease of the skin, involves oxidative stress. Based on that, the generation of reactive oxygen species (ROS) by the mitochondria may be relevant in the pathogenesis of vitiligo. Here, we evaluate the biochemical and functional alterations involved in the defective activity that has been previously described both in melanocytes and peripheral blood mononuclear cells (PBMC) from vitiligo patients. Moreover, we used a freeze-thaw test as a mild stress stimulus to disclose any latent defects in the assembly of membrane lipids that may compromise the functionality of the membrane itself. We show that the lipid constitution of the membrane is altered in vitiligo. Specifically, the cardiolipin (CL) level in the mitochondrial inner membrane is reduced and the level of cholesterol is increased. Furthermore, an increase in the expression level of 3-hydroxy-3methyl-glutaryl-CoenzymeA-reductase (HMG-CoA reductase), the rate-limiting enzyme for cholesterol biosynthesis, was also seen. Associated with that, the expression of electron transport chain (ETC) lipid-dependent subunits was also modified, and their expression was further affected by the freeze-thaw stress. The expression of CL-independent mitochondrial proteins, such as porin and Bcl2, were unaffected in vitiligo PBMC. These data confirm that ETC protein expression mainly correlates with lipid arrangement and that loss of their expression is not due to generalized or random oxidative-mediated damage. We suggest that the modification of membrane lipid components in vitiligo cells may be the biochemical basis for the mitochondrial impairment and the subsequent production of intracellular ROS following the exposure to a mild stress.
Collapse
Affiliation(s)
- Maria Lucia Dell'Anna
- Laboratory of Cutaneous Physiopathology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
25
|
García N, Zazueta C, El-Hafidi M, Pavón N, Martínez-Abundis E, Hernández-Esquivel L, Chávez E. Cyclosporin A inhibits UV-radiation-induced membrane damage but is unable to inhibit carboxyatractyloside-induced permeability transition. Radiat Res 2009; 172:575-83. [PMID: 19883225 DOI: 10.1667/rr1799.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
This work was undertaken to gain further information on the chemical characteristics of the membrane entity involved in the formation of the nonspecific pore. Mitochondria were subjected to oxidative stress by exposure to UV radiation. The results indicate that ultraviolet C radiation induces structural modifications in the adenine nucleotide translocase that lead to membrane permeability transition. Membrane leakage was assessed by measuring mitochondrial Ca2+ transport, the transmembrane electric gradient, and mitochondrial swelling. UV-irradiated mitochondria were unable to retain matrix Ca2+ or to maintain a high level of membrane potential when Ca2+ was added; furthermore, UV-irradiated mitochondria underwent large amplitude swelling. Release of cytochrome c and formation of malondialdehyde, owing to lipid peroxidation, were also seen. Structural modifications of the translocase were revealed by an increase in the binding of the fluorescent probe eosin-5-maleimide to thiol residues of the ADP/ATP carrier. These modifications, taken together with findings indicating that cyclosporin resulted unable to inhibit carboxyatractyloside-induced permeability transition, prompted us to conclude that the translocase could constitute the nonspecific pore or at least be an important modulator of it.
Collapse
Affiliation(s)
- Noemí García
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Ignacio Chávez, Mexico, D.F. 014080, Mexico
| | | | | | | | | | | | | |
Collapse
|
26
|
|