51
|
Beaulant A, Dia M, Pillot B, Chauvin MA, Ji-Cao J, Durand C, Bendridi N, Chanon S, Vieille-Marchiset A, Da Silva CC, Patouraux S, Anty R, Iannelli A, Tran A, Gual P, Vidal H, Gomez L, Paillard M, Rieusset J. Endoplasmic reticulum-mitochondria miscommunication is an early and causal trigger of hepatic insulin resistance and steatosis. J Hepatol 2022; 77:710-722. [PMID: 35358616 DOI: 10.1016/j.jhep.2022.03.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 02/18/2022] [Accepted: 03/07/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Hepatic insulin resistance in obesity and type 2 diabetes was recently associated with endoplasmic reticulum (ER)-mitochondria miscommunication. These contact sites (mitochondria-associated membranes: MAMs) are highly dynamic and involved in many functions; however, whether MAM dysfunction plays a causal role in hepatic insulin resistance and steatosis is not clear. Thus, we aimed to determine whether and how organelle miscommunication plays a role in the onset and progression of hepatic metabolic impairment. METHODS We analyzed hepatic ER-mitochondria interactions and calcium exchange in a time-dependent and reversible manner in mice with diet-induced obesity. Additionally, we used recombinant adenovirus to express a specific organelle spacer or linker in mouse livers, to determine the causal impact of MAM dysfunction on hepatic metabolic alterations. RESULTS Disruption of ER-mitochondria interactions and calcium exchange is an early event preceding hepatic insulin resistance and steatosis in mice with diet-induced obesity. Interestingly, an 8-week reversal diet concomitantly reversed hepatic organelle miscommunication and insulin resistance in obese mice. Mechanistically, disrupting structural and functional ER-mitochondria interactions through the hepatic overexpression of the organelle spacer FATE1 was sufficient to impair hepatic insulin action and glucose homeostasis. In addition, FATE1-mediated organelle miscommunication disrupted lipid-related mitochondrial oxidative metabolism and induced hepatic steatosis. Conversely, reinforcement of ER-mitochondria interactions through hepatic expression of a synthetic linker prevented diet-induced glucose intolerance after 4 weeks' overnutrition. Importantly, ER-mitochondria miscommunication was confirmed in the liver of obese patients with type 2 diabetes, and correlated with glycemia, HbA1c and HOMA-IR index. CONCLUSIONS ER-mitochondria miscommunication is an early causal trigger of hepatic insulin resistance and steatosis, and can be reversed by switching to a healthy diet. Thus, targeting MAMs could help to restore metabolic homeostasis. LAY SUMMARY The literature suggests that interactions between the endoplasmic reticulum and mitochondria could play a role in hepatic insulin resistance and steatosis during chronic obesity. In the present study, we reappraised the time-dependent regulation of hepatic endoplasmic reticulum-mitochondria interactions and calcium exchange, investigating reversibility and causality, in mice with diet-induced obesity. We also assessed the relevance of our findings to humans. We show that organelle miscommunication is an early causal trigger of hepatic insulin resistance and steatosis that can be improved by nutritional strategies.
Collapse
Affiliation(s)
- Agathe Beaulant
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Maya Dia
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Bruno Pillot
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Marie-Agnes Chauvin
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Jingwei Ji-Cao
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Christine Durand
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Nadia Bendridi
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Stephanie Chanon
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Aurelie Vieille-Marchiset
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Claire Crola Da Silva
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Stéphanie Patouraux
- Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, INSERM, U1065, C3M, Nice, France
| | - Rodolphe Anty
- Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, INSERM, U1065, C3M, Nice, France
| | - Antonio Iannelli
- Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, INSERM, U1065, C3M, Nice, France
| | - Albert Tran
- Université Côte d'Azur, CHU, INSERM, U1065, C3M, Nice, France; Université Côte d'Azur, INSERM, U1065, C3M, Nice, France
| | - Philippe Gual
- Université Côte d'Azur, INSERM, U1065, C3M, Nice, France
| | - Hubert Vidal
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Ludovic Gomez
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Melanie Paillard
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France
| | - Jennifer Rieusset
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, Université Claude Bernard Lyon1, F-69310 Pierre-Bénite and F-69500 Bron, France.
| |
Collapse
|
52
|
Mitochondria-Associated Endoplasmic Reticulum Membranes: Inextricably Linked with Autophagy Process. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7086807. [PMID: 36052160 PMCID: PMC9427242 DOI: 10.1155/2022/7086807] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/08/2022] [Indexed: 01/18/2023]
Abstract
Mitochondria-associated membranes (MAMs), physical connection sites between the endoplasmic reticulum (ER) and the outer mitochondrial membrane (OMM), are involved in numerous cellular processes, such as calcium ion transport, lipid metabolism, autophagy, ER stress, mitochondria morphology, and apoptosis. Autophagy is a highly conserved intracellular process in which cellular contents are delivered by double-membrane vesicles, called autophagosomes, to the lysosomes for destruction and recycling. Autophagy, typically triggered by stress, eliminates damaged or redundant protein molecules and organelles to maintain regular cellular activity. Dysfunction of MAMs or autophagy is intimately associated with various diseases, including aging, cardiovascular, infections, cancer, multiple toxic agents, and some genetic disorders. Increasing evidence has shown that MAMs play a significant role in autophagy development and maturation. In our study, we concentrated on two opposing functions of MAMs in autophagy: facilitating the formation of autophagosomes and inhibiting autophagy. We recognized the link between MAMs and autophagy in the occurrence and progression of the diseases and therefore collated and summarized the existing intrinsic molecular mechanisms. Furthermore, we draw attention to several crucial data and open issues in the area that may be helpful for further study.
Collapse
|
53
|
Sassano ML, Felipe-Abrio B, Agostinis P. ER-mitochondria contact sites; a multifaceted factory for Ca 2+ signaling and lipid transport. Front Cell Dev Biol 2022; 10:988014. [PMID: 36158205 PMCID: PMC9494157 DOI: 10.3389/fcell.2022.988014] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Membrane contact sites (MCS) between organelles of eukaryotic cells provide structural integrity and promote organelle homeostasis by facilitating intracellular signaling, exchange of ions, metabolites and lipids and membrane dynamics. Cataloguing MCS revolutionized our understanding of the structural organization of a eukaryotic cell, but the functional role of MSCs and their role in complex diseases, such as cancer, are only gradually emerging. In particular, the endoplasmic reticulum (ER)-mitochondria contacts (EMCS) are key effectors of non-vesicular lipid trafficking, thereby regulating the lipid composition of cellular membranes and organelles, their physiological functions and lipid-mediated signaling pathways both in physiological and diseased conditions. In this short review, we discuss key aspects of the functional complexity of EMCS in mammalian cells, with particular emphasis on their role as central hubs for lipid transport between these organelles and how perturbations of these pathways may favor key traits of cancer cells.
Collapse
Affiliation(s)
- Maria Livia Sassano
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Blanca Felipe-Abrio
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| |
Collapse
|
54
|
Ubiquitin-specific peptidase 10 ameliorates hepatic steatosis in nonalcoholic steatohepatitis model by restoring autophagic activity. Dig Liver Dis 2022; 54:1021-1029. [PMID: 35288065 DOI: 10.1016/j.dld.2022.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/18/2022] [Accepted: 02/14/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is a critical event in the progression of nonalcoholic fatty liver disease (NAFLD). Steatosis induces lipotoxicity, driving the transition of simple fatty liver (NAFL) to NASH. Autophagy affects NAFLD by improving steatosis. AIM To investigate whether ubiquitin-specific peptidase (USP)10 alleviates hepatic steatosis through autophagy. METHODS A methionine-choline-deficient diet (MCDD) and choline-deficient diet (CDD) were used to model rodent NASH and NAFL, respectively. HepG2 cells were treated with palmitic acid to model hepatocellular steatosis. A viral carrier was used to regulate the USP10 level. Real-time fluorescence quantitative polymerase chain reaction, western blotting, histology, and electron microscopy were used to detect autophagic activity and steatosis. RESULTS In vivo, a time-dependent correlation of USP10 and autophagic activity in the liver was found during NAFLD (including NAFL and NASH) modeling. After 8 weeks of modeling, the autophagic activity of NASH was lower than that of the healthy controls and those with NAFL. USP10 could promote autophagy-related pathways and molecules and increase the synthesis of autophagosomes in NASH, improving steatosis, inflammation, and fibrosis. In vitro, autophagy inhibitors reversed the lipid-lowering effect of USP10 without decreasing the level of fatty acid β-oxidation. CONCLUSION USP10 ameliorated histological steatosis, inflammation, and fibrosis. USP10 alleviated hepatic steatosis in NASH in an autophagy-dependent manner.
Collapse
|
55
|
Li YE, Sowers JR, Hetz C, Ren J. Cell death regulation by MAMs: from molecular mechanisms to therapeutic implications in cardiovascular diseases. Cell Death Dis 2022; 13:504. [PMID: 35624099 PMCID: PMC9142581 DOI: 10.1038/s41419-022-04942-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/04/2022] [Accepted: 05/12/2022] [Indexed: 12/13/2022]
Abstract
The endoplasmic reticulum (ER) and mitochondria are interconnected intracellular organelles with vital roles in the regulation of cell signaling and function. While the ER participates in a number of biological processes including lipid biosynthesis, Ca2+ storage and protein folding and processing, mitochondria are highly dynamic organelles governing ATP synthesis, free radical production, innate immunity and apoptosis. Interplay between the ER and mitochondria plays a crucial role in regulating energy metabolism and cell fate control under stress. The mitochondria-associated membranes (MAMs) denote physical contact sites between ER and mitochondria that mediate bidirectional communications between the two organelles. Although Ca2+ transport from ER to mitochondria is vital for mitochondrial homeostasis and energy metabolism, unrestrained Ca2+ transfer may result in mitochondrial Ca2+ overload, mitochondrial damage and cell death. Here we summarize the roles of MAMs in cell physiology and its impact in pathological conditions with a focus on cardiovascular disease. The possibility of manipulating ER-mitochondria contacts as potential therapeutic approaches is also discussed.
Collapse
Affiliation(s)
- Yiran E Li
- Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - James R Sowers
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Buck Institute for Research in Aging, Novato, CA, 94945, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
56
|
Seo W, Silwal P, Song IC, Jo EK. The dual role of autophagy in acute myeloid leukemia. J Hematol Oncol 2022; 15:51. [PMID: 35526025 PMCID: PMC9077970 DOI: 10.1186/s13045-022-01262-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/14/2022] [Indexed: 01/18/2023] Open
Abstract
Acute myeloid leukemia (AML) is a severe hematologic malignancy prevalent in older patients, and the identification of potential therapeutic targets for AML is problematic. Autophagy is a lysosome-dependent catabolic pathway involved in the tumorigenesis and/or treatment of various cancers. Mounting evidence has suggested that autophagy plays a critical role in the initiation and progression of AML and anticancer responses. In this review, we describe recent updates on the multifaceted functions of autophagy linking to genetic alterations of AML. We also summarize the latest evidence for autophagy-related genes as potential prognostic predictors and drivers of AML tumorigenesis. We then discuss the crosstalk between autophagy and tumor cell metabolism into the impact on both AML progression and anti-leukemic treatment. Moreover, a series of autophagy regulators, i.e., the inhibitors and activators, are described as potential therapeutics for AML. Finally, we describe the translation of autophagy-modulating therapeutics into clinical practice. Autophagy in AML is a double-edged sword, necessitating a deeper understanding of how autophagy influences dual functions in AML tumorigenesis and anti-leukemic responses.
Collapse
Affiliation(s)
- Wonhyoung Seo
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Korea.,Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Korea.,Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Korea
| | - Prashanta Silwal
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Korea.,Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Korea
| | - Ik-Chan Song
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, 35015, Korea
| | - Eun-Kyeong Jo
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Korea. .,Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Korea. .,Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Korea.
| |
Collapse
|
57
|
Reyes-Castellanos G, Abdel Hadi N, Carrier A. Autophagy Contributes to Metabolic Reprogramming and Therapeutic Resistance in Pancreatic Tumors. Cells 2022; 11:426. [PMID: 35159234 PMCID: PMC8834004 DOI: 10.3390/cells11030426] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic reprogramming is a feature of cancers for which recent research has been particularly active, providing numerous insights into the mechanisms involved. It occurs across the entire cancer process, from development to resistance to therapies. Established tumors exhibit dependencies for metabolic pathways, constituting vulnerabilities that can be targeted in the clinic. This knowledge is of particular importance for cancers that are refractory to any therapeutic approach, such as Pancreatic Ductal Adenocarcinoma (PDAC). One of the metabolic pathways dysregulated in PDAC is autophagy, a survival process that feeds the tumor with recycled intracellular components, through both cell-autonomous (in tumor cells) and nonautonomous (from the local and distant environment) mechanisms. Autophagy is elevated in established PDAC tumors, contributing to aberrant proliferation and growth even in a nutrient-poor context. Critical elements link autophagy to PDAC including genetic alterations, mitochondrial metabolism, the tumor microenvironment (TME), and the immune system. Moreover, high autophagic activity in PDAC is markedly related to resistance to current therapies. In this context, combining autophagy inhibition with standard chemotherapy, and/or drugs targeting other vulnerabilities such as metabolic pathways or the immune response, is an ongoing clinical strategy for which there is still much to do through translational and multidisciplinary research.
Collapse
Affiliation(s)
| | | | - Alice Carrier
- Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS, INSERM, Institut Paoli-Calmettes, Aix Marseille Université, F-13009 Marseille, France; (G.R.-C.); (N.A.H.)
| |
Collapse
|
58
|
Lower RNA expression of ALDH1A1 distinguishes the favorable risk group in acute myeloid leukemia. Mol Biol Rep 2022; 49:3321-3331. [PMID: 35028852 DOI: 10.1007/s11033-021-07073-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023]
Abstract
The expression and activity of enzymes that belong to the aldehyde dehydrogenases is a characteristic of both normal and malignant stem cells. ALDH1A1 is an enzyme critical in cancer stem cells. In acute myeloid leukemia (AML), ALDH1A1 protects leukemia-initiating cells from a number of antineoplastic agents, which include inhibitors of protein tyrosine kinases. Furthermore, ALDH1A1 proves vital for the establishment of human AML xenografts in mice. We review here important studies characterizing the role of ALDH1A1 in AML and its potential as a therapeutic target. We also analyze datasets from leading studies, and show that decreased ALDH1A1 RNA expression consistently characterizes the AML patient risk group with a favorable prognosis, while there is a consistent association of high ALDH1A1 RNA expression with high risk and poor overall survival. Our review and analysis reinforces the notion to employ both novel as well as existing inhibitors of the ALDH1A1 protein against AML.
Collapse
|
59
|
Saliakoura M, Sebastiano MR, Nikdima I, Pozzato C, Konstantinidou G. Restriction of extracellular lipids renders pancreatic cancer dependent on autophagy. J Exp Clin Cancer Res 2022; 41:16. [PMID: 34998392 PMCID: PMC8742413 DOI: 10.1186/s13046-021-02231-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/21/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND KRAS is the predominant oncogene mutated in pancreatic ductal adenocarcinoma (PDAC), the fourth cause of cancer-related deaths worldwide. Mutant KRAS-driven tumors are metabolically programmed to support their growth and survival, which can be used to identify metabolic vulnerabilities. In the present study, we aimed to understand the role of extracellularly derived fatty acids in KRAS-driven pancreatic cancer. METHODS To assess the dependence of PDAC cells on extracellular fatty acids we employed delipidated serum or RNAi-mediated suppression of ACSL3 (to inhibit the activation and cellular retention of extracellular fatty acids) followed by cell proliferation assays, qPCR, apoptosis assays, immunoblots and fluorescence microscopy experiments. To assess autophagy in vivo, we employed the KrasG12D/+;p53flox/flox;Pdx1-CreERT2 (KPC) mice crossed with Acsl3 knockout mice, and to assess the efficacy of the combination therapy of ACSL3 and autophagy inhibition we used xenografted human cancer cell-derived tumors in immunocompromised mice. RESULTS Here we show that depletion of extracellularly derived lipids either by serum lipid restriction or suppression of ACSL3, triggers autophagy, a process that protects PDAC cells from the reduction of bioenergetic intermediates. Combined extracellular lipid deprivation and autophagy inhibition exhibits anti-proliferative and pro-apoptotic effects against PDAC cell lines in vitro and promotes suppression of xenografted human pancreatic cancer cell-derived tumors in mice. Therefore, we propose lipid deprivation and autophagy blockade as a potential co-targeting strategy for PDAC treatment. CONCLUSIONS Our work unravels a central role of extracellular lipid supply in ensuring fatty acid provision in cancer cells, unmasking a previously unappreciated metabolic vulnerability of PDAC cells.
Collapse
Affiliation(s)
- Maria Saliakoura
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | | | - Ioanna Nikdima
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | - Chiara Pozzato
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | | |
Collapse
|
60
|
Sun Q, Wang Z, Liu B, He F, Gai S, Yang P, Yang D, Li C, Lin J. Recent advances on endogenous/exogenous stimuli-triggered nanoplatforms for enhanced chemodynamic therapy. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214267] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
61
|
Drosophila D-idua Reduction Mimics Mucopolysaccharidosis Type I Disease-Related Phenotypes. Cells 2021; 11:cells11010129. [PMID: 35011691 PMCID: PMC8750945 DOI: 10.3390/cells11010129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 01/21/2023] Open
Abstract
Deficit of the IDUA (α-L-iduronidase) enzyme causes the lysosomal storage disorder mucopolysaccharidosis type I (MPS I), a rare pediatric neurometabolic disease, due to pathological variants in the IDUA gene and is characterized by the accumulation of the undegraded mucopolysaccharides heparan sulfate and dermatan sulfate into lysosomes, with secondary cellular consequences that are still mostly unclarified. Here, we report a new fruit fly RNAi-mediated knockdown model of a IDUA homolog (D-idua) displaying a phenotype mimicking some typical molecular features of Lysosomal Storage Disorders (LSD). In this study, we showed that D-idua is a vital gene in Drosophila and that ubiquitous reduction of its expression leads to lethality during the pupal stage, when the precise degradation/synthesis of macromolecules, together with a functional autophagic pathway, are indispensable for the correct development to the adult stage. Tissue-specific analysis of the D-idua model showed an increase in the number and size of lysosomes in the brain and muscle. Moreover, the incorrect acidification of lysosomes led to dysfunctional lysosome-autophagosome fusion and the consequent block of autophagy flux. A concomitant metabolic drift of glycolysis and lipogenesis pathways was observed. After starvation, D-idua larvae showed a quite complete rescue of both autophagy/lysosome phenotypes and metabolic alterations. Metabolism and autophagy are strictly interconnected vital processes that contribute to maintain homeostatic control of energy balance, and little is known about this regulation in LSDs. Our results provide new starting points for future investigations on the disease’s pathogenic mechanisms and possible pharmacological manipulations.
Collapse
|
62
|
Overexpression of Neuroglobin Promotes Energy Metabolism and Autophagy Induction in Human Neuroblastoma SH-SY5Y Cells. Cells 2021; 10:cells10123394. [PMID: 34943907 PMCID: PMC8699457 DOI: 10.3390/cells10123394] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 01/18/2023] Open
Abstract
Neuroglobin (NGB) is an O2-binding globin mainly expressed in the central and peripheral nervous systems and cerebrospinal fluid. Previously, it was demonstrated that NGB overexpression protects cells from hypoxia-induced death. To investigate processes promoted by NGB overexpression, we used a cellular model of neuroblastoma stably overexpressing an NGB-FLAG construct. We used a proteomic approach to identify the specific profile following NGB overexpression. To evaluate the role of NGB overexpression in increasing energetic metabolism, we measured oxygen consumption rate (OCR) and the extracellular acidification rate through Seahorse XF technology. The effect on autophagy induction was evaluated by analyzing SQSTM1/p62 and LC3-II expression. Proteomic analysis revealed several differentially regulated proteins, involved in oxidative phosphorylation and integral mitochondrial proteins linked to energy metabolism. The analysis of mitochondrial metabolism demonstrated that NGB overexpression increases mitochondrial ATP production. Indeed, NGB overexpression enhances bioenergetic metabolism, increasing OCR and oxygen consumption. Analysis of autophagy induction revealed an increase of LC3-II together with a significant decrease of SQSTM1/p62, and NGB-LC3-II association during autophagosome formation. These results highlight the active participation of NGB in several cellular processes that can be upregulated in response to NGB overexpression, playing a role in the adaptive response to stress in neuroblastoma cells.
Collapse
|
63
|
Pandey A, Yadav P, Shukla S. Unfolding the role of autophagy in the cancer metabolism. Biochem Biophys Rep 2021; 28:101158. [PMID: 34754952 PMCID: PMC8564564 DOI: 10.1016/j.bbrep.2021.101158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is considered an indispensable process that scavenges toxins, recycles complex macromolecules, and sustains the essential cellular functions. In addition to its housekeeping role, autophagy plays a substantial role in many pathophysiological processes such as cancer. Certainly, it adapts cancer cells to thrive in the stress conditions such as hypoxia and starvation. Cancer cells indeed have also evolved by exploiting the autophagy process to fulfill energy requirements through the production of metabolic fuel sources and fundamentally altered metabolic pathways. Occasionally autophagy as a foe impedes tumorigenesis and promotes cell death. The complex role of autophagy in cancer makes it a potent therapeutic target and has been actively tested in clinical trials. Moreover, the versatility of autophagy has opened new avenues of effective combinatorial therapeutic strategies. Thereby, it is imperative to comprehend the specificity of autophagy in cancer-metabolism. This review summarizes the recent research and conceptual framework on the regulation of autophagy by various metabolic pathways, enzymes, and their cross-talk in the cancer milieu, including the implementation of altered metabolism and autophagy in clinically approved and experimental therapeutics.
Collapse
Affiliation(s)
- Anchala Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, 462066, Madhya Pradesh, India
| | - Pooja Yadav
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, 462066, Madhya Pradesh, India
| | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, 462066, Madhya Pradesh, India
| |
Collapse
|
64
|
PACS-2 attenuates diabetic kidney disease via the enhancement of mitochondria-associated endoplasmic reticulum membrane formation. Cell Death Dis 2021; 12:1107. [PMID: 34836936 PMCID: PMC8626491 DOI: 10.1038/s41419-021-04408-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023]
Abstract
The altered homeostasis of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM) was closely associated with the pathological process of nervous system diseases and insulin resistance. Here, the exact implication of phosphofurin acidic cluster sorting protein 2 (PCAS-2), an anchor protein in the MAM interface, in diabetic kidney disease was investigated. In the kidneys of type 1 and type 2 diabetes mice and HG-induced HK-2 cells, a notable disruption of ER-mitochondria interactions, accompanied by a decreased PACS-2 expression in all subcellular fractions. Furthermore, PACS-2 knockout mice with diabetes displayed accelerated development of proteinuria, deterioration of kidney function, and aggravated disruption of MAM area, ER stress, mitochondrial dysfunction, renal apoptosis, and fibrosis. However, overexpression of PACS-2 effectively protected diabetic kidneys and HG-treated HK-2 cells from renal tubular impairments. Importantly, experimental uncoupling of ER-mitochondria contacts reversed the protective effects of PACS-2 restoration on HK-2 cells under HG conditions. In summary, our data indicate a pivotal role of PACS-2 in the development of diabetic renal tubular injury via the stabilization of MAM.
Collapse
|
65
|
Ganzleben I, Neurath MF, Becker C. Autophagy in Cancer Therapy-Molecular Mechanisms and Current Clinical Advances. Cancers (Basel) 2021; 13:cancers13215575. [PMID: 34771737 PMCID: PMC8583685 DOI: 10.3390/cancers13215575] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Autophagy is the capability of cells to dismantle and recycle parts of themselves. This process is closely intertwined with other crucial cell functions, such as growth and control of metabolism. Autophagy is oftentimes dysregulated in cancer and offers established and advanced tumors protection against a lack of nutrients and an advantage regarding proliferation. This review will present an overview of the basics of human autophagy, its dysregulation in cancer, and approaches to target autophagy in cancer treatment in recent and current clinical trials as well as new findings of preclinical research. Abstract Autophagy is a crucial general survival tactic of mammalian cells. It describes the capability of cells to disassemble and partially recycle cellular components (e.g., mitochondria) in case they are damaged and pose a risk to cell survival or simply if their resources are urgently needed elsewhere at the time. Autophagy-associated pathomechanisms have been increasingly recognized as important disease mechanisms in non-malignant (neurodegeneration, diffuse parenchymal lung disease) and malignant conditions alike. However, the overall consequences of autophagy for the organism depend particularly on the greater context in which autophagy occurs, such as the cell type or whether the cell is proliferating. In cancer, autophagy sustains cancer cell survival under challenging, i.e., resource-depleted, conditions. However, this leads to situations in which cancer cells are completely dependent on autophagy. Accordingly, autophagy represents a promising yet complex target in cancer treatment with therapeutically induced increase and decrease of autophagic flux as important therapeutic principles.
Collapse
Affiliation(s)
- Ingo Ganzleben
- Department of Medicine 1, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (I.G.); (M.F.N.)
- Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (I.G.); (M.F.N.)
- Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (I.G.); (M.F.N.)
- Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Correspondence:
| |
Collapse
|
66
|
Bosc C, Saland E, Bousard A, Gadaud N, Sabatier M, Cognet G, Farge T, Boet E, Gotanègre M, Aroua N, Mouchel PL, Polley N, Larrue C, Kaphan E, Picard M, Sahal A, Jarrou L, Tosolini M, Rambow F, Cabon F, Nicot N, Poillet-Perez L, Wang Y, Su X, Fovez Q, Kluza J, Argüello RJ, Mazzotti C, Avet-Loiseau H, Vergez F, Tamburini J, Fournié JJ, Tiong IS, Wei AH, Kaoma T, Marine JC, Récher C, Stuani L, Joffre C, Sarry JE. Mitochondrial inhibitors circumvent adaptive resistance to venetoclax and cytarabine combination therapy in acute myeloid leukemia. NATURE CANCER 2021; 2:1204-1223. [PMID: 35122057 DOI: 10.1038/s43018-021-00264-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/31/2021] [Indexed: 04/23/2023]
Abstract
Therapy resistance represents a major clinical challenge in acute myeloid leukemia (AML). Here we define a 'MitoScore' signature, which identifies high mitochondrial oxidative phosphorylation in vivo and in patients with AML. Primary AML cells with cytarabine (AraC) resistance and a high MitoScore relied on mitochondrial Bcl2 and were highly sensitive to venetoclax (VEN) + AraC (but not to VEN + azacytidine). Single-cell transcriptomics of VEN + AraC-residual cell populations revealed adaptive resistance associated with changes in oxidative phosphorylation, electron transport chain complex and the TP53 pathway. Accordingly, treatment of VEN + AraC-resistant AML cells with electron transport chain complex inhibitors, pyruvate dehydrogenase inhibitors or mitochondrial ClpP protease agonists substantially delayed relapse following VEN + AraC. These findings highlight the central role of mitochondrial adaptation during AML therapy and provide a scientific rationale for alternating VEN + azacytidine with VEN + AraC in patients with a high MitoScore and to target mitochondrial metabolism to enhance the sensitivity of AML cells to currently approved therapies.
Collapse
Affiliation(s)
- Claudie Bosc
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Estelle Saland
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Aurélie Bousard
- Department of Oncology, Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
| | - Noémie Gadaud
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- University of Toulouse, Toulouse, France
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Marie Sabatier
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Guillaume Cognet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Thomas Farge
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Emeline Boet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Mathilde Gotanègre
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Nesrine Aroua
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Pierre-Luc Mouchel
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- University of Toulouse, Toulouse, France
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Nathaniel Polley
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Clément Larrue
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Eléonore Kaphan
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Muriel Picard
- Réanimation Polyvalente IUCT-oncopole, CHU de Toulouse, Toulouse, France
| | - Ambrine Sahal
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Latifa Jarrou
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Marie Tosolini
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Florian Rambow
- Department of Oncology, Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
| | - Florence Cabon
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Nathalie Nicot
- LuxGen, Quantitative Biology Unit, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Laura Poillet-Perez
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Yujue Wang
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Xiaoyang Su
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Quentin Fovez
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277, Lille, France
| | - Jérôme Kluza
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277, Lille, France
| | - Rafael José Argüello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Céline Mazzotti
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Hervé Avet-Loiseau
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - François Vergez
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- University of Toulouse, Toulouse, France
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | | | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
| | - Ing S Tiong
- Department of Clinical Haematology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Andrew H Wei
- Department of Clinical Haematology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Tony Kaoma
- Computational Biomedicine Research Group, Quantitative Biology Unit, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Jean-Christophe Marine
- Department of Oncology, Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
| | - Christian Récher
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- University of Toulouse, Toulouse, France
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Lucille Stuani
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Carine Joffre
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, CNRS, Toulouse, France.
- LabEx Toucan, Toulouse, France.
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France.
| |
Collapse
|
67
|
Takahashi N, Hatakeyama K, Nagashima T, Ohshima K, Urakami K, Yamaguchi K, Hirashima Y. Activation of oxidative phosphorylation in TP53-inactive endometrial carcinomas with a poor prognosis. Int J Gynecol Cancer 2021; 31:1557-1563. [PMID: 34725206 DOI: 10.1136/ijgc-2021-002983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/13/2021] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE We aimed to identify pathways for potential therapeutic targets by conducting molecular profiling of endometrial carcinomas in patients with poor prognosis. METHODS The classification of endometrial carcinomas has undergone a paradigm shift with the advent of next generation sequencing based molecular profiling. Although this emerging classification reflects poor prognosis in patients with endometrial carcinoma, knowledge of affected biological pathways is still lacking. In this study, 85 patients with endometrial carcinomas at the Shizuoka Cancer Center were evaluated from January 2014 to March 2019 and classified based on The Cancer Genome Atlas subgroups. The accumulation of germline and somatic mutations was determined using next generation sequencing. Gene expression profiling was used to determine the effect of TP53 inactivation on the recurrence of endometrial carcinoma. Additionally, the biological pathways associated with TP53 inactivation were estimated by pathway analysis based on gene expression. RESULTS Based on The Cancer Genome Atlas classification, the ratio of polymerase-epsilon to copy number-high subgroups and the frequency of PTEN and TP53 mutations differed in patients, and mutations of ARHGAP35 observed in normal endometrium were accumulated in the polymerase-epsilon and microsatellite instability subgroups. We revealed that copy number-high reflects TP53 inactivation in endometrial carcinomas, and that TP53-inactive tumors with or without TP53 mutations have poor prognosis. Furthermore, overexpression of aurora kinase A and activation of oxidative phosphorylation were found in TP53-inactivated endometrial carcinomas, suggesting that the PI3K/mTOR and autophagy pathways are potential drug targets. CONCLUSION Our analysis revealed a relationship between pathways involved in oxidative phosphorylation and poor prognosis and provides insight into potential drug targets.
Collapse
Affiliation(s)
- Nobutaka Takahashi
- Department of Gynecology, Shizuoka Cancer Center, Sunto-gun, Shizuoka, Japan
| | - Keiichi Hatakeyama
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka, Japan
| | - Takeshi Nagashima
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka, Japan.,SRL Inc, Shinjuku-ku, Tokyo, Japan
| | - Keiichi Ohshima
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka, Japan
| | - Kenichi Urakami
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka, Japan
| | | | - Yasuyuki Hirashima
- Department of Gynecology, Shizuoka Cancer Center, Sunto-gun, Shizuoka, Japan
| |
Collapse
|
68
|
Zhang L, Yan F, Li L, Fu H, Song D, Wu D, Wang X. New focuses on roles of communications between endoplasmic reticulum and mitochondria in identification of biomarkers and targets. Clin Transl Med 2021; 11:e626. [PMID: 34841708 PMCID: PMC8562589 DOI: 10.1002/ctm2.626] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
The communication between endoplasmic reticulum (ER) and mitochondria (Mt) plays important roles in maintenance of intra- and extra-cellular microenvironment, metabolisms, signaling activities and cell-cell communication. The present review aims to overview the advanced understanding about roles of ER-Mt structural contacts, molecular interactions and chemical exchanges, signal transmissions and inter-organelle regulations in ER-Mt communication. We address how the ER-Mt communication contributes to the regulation of lipid, amino acid and glucose metabolisms by enzymes, transporters and regulators in the process of biosynthesis. We specially emphasize the importance of deep understanding about molecular mechanisms of ER-Mt communication for identification and development of biology-specific, disease-specific and metabolism-specific biomarkers and therapeutic targets for human diseases. The inhibitors and modulators of the ER-Mt communication are categorized according to therapeutic targets. Rapid development of biotechnologies will provide new insights for spatiotemporally understanding the molecular mechanisms of ER-Mt communication.
Collapse
Affiliation(s)
- Linlin Zhang
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineJinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Furong Yan
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineJinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Liyang Li
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineJinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Huirong Fu
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineJinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Dongli Song
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineJinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Duojiao Wu
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineJinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Xiangdong Wang
- Zhongshan HospitalDepartment of Pulmonary and Critical Care MedicineJinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| |
Collapse
|
69
|
Geldenhuys WJ, Piktel D, Moore JC, Rellick SL, Meadows E, Pinti MV, Hollander JM, Ammer AG, Martin KH, Gibson LF. Loss of the redox mitochondrial protein mitoNEET leads to mitochondrial dysfunction in B-cell acute lymphoblastic leukemia. Free Radic Biol Med 2021; 175:226-235. [PMID: 34496224 PMCID: PMC8478879 DOI: 10.1016/j.freeradbiomed.2021.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/28/2021] [Accepted: 09/04/2021] [Indexed: 01/12/2023]
Abstract
B-cell acute lymphoblastic leukemia (ALL) affects both pediatric and adult patients. Chemotherapy resistant tumor cells that contribute to minimal residual disease (MRD) underlie relapse and poor clinical outcomes in a sub-set of patients. Targeting mitochondrial oxidative phosphorylation (OXPHOS) in the treatment of refractory leukemic cells is a potential novel approach to sensitizing tumor cells to existing standard of care therapeutic agents. In the current study, we have expanded our previous investigation of the mitoNEET ligand NL-1 in the treatment of ALL to interrogate the functional role of the mitochondrial outer membrane protein mitoNEET in B-cell ALL. Knockout (KO) of mitoNEET (gene: CISD1) in REH leukemic cells led to changes in mitochondrial ultra-structure and function. REH cells have significantly reduced OXPHOS capacity in the KO cells coincident with reduction in electron flow and increased reactive oxygen species. In addition, we found a decrease in lipid content in KO cells, as compared to the vector control cells was observed. Lastly, the KO of mitoNEET was associated with decreased proliferation as compared to control cells when exposed to the standard of care agent cytarabine (Ara-C). Taken together, these observations suggest that mitoNEET is essential for optimal function of mitochondria in B-cell ALL and may represent a novel anti-leukemic drug target for treatment of minimal residual disease.
Collapse
Affiliation(s)
- Werner J Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA; Mitochondria Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Debbie Piktel
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA; West Virginia University Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Javohn C Moore
- West Virginia University Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Stephanie L Rellick
- West Virginia University Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Ethan Meadows
- Department of Human Performance, West Virginia University School of Medicine, Morgantown, WV, USA; Mitochondria Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Mark V Pinti
- Department of Human Performance, West Virginia University School of Medicine, Morgantown, WV, USA; Mitochondria Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - John M Hollander
- Department of Human Performance, West Virginia University School of Medicine, Morgantown, WV, USA; Mitochondria Metabolism and Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Amanda G Ammer
- West Virginia University Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Karen H Martin
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA; West Virginia University Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Laura F Gibson
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA; West Virginia University Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
70
|
Mohammadpour H, MacDonald CR, McCarthy PL, Abrams SI, Repasky EA. β2-adrenergic receptor signaling regulates metabolic pathways critical to myeloid-derived suppressor cell function within the TME. Cell Rep 2021; 37:109883. [PMID: 34706232 PMCID: PMC8601406 DOI: 10.1016/j.celrep.2021.109883] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/11/2021] [Accepted: 10/01/2021] [Indexed: 11/20/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) impede antitumor immunity; however, the precise mechanisms that regulate their suppressive function remain unresolved. Identifying these mechanisms could lead to therapeutic interventions to boost cancer immunotherapy efficacy. Here, we reveal that β2 adrenergic receptor (β2-AR) expression on MDSCs increases with tumor growth and that the β2-AR stress pathway drives the immune suppressive activity of MDSCs by altering their metabolism. We show that β2-AR signaling decreases glycolysis and increases oxidative phosphorylation and fatty acid oxidation (FAO). It also increases expression of the fatty acid transporter CPT1A, which is necessary for the FAO-mediated immunosuppressive function of MDSCs. Moreover, we show that β2-AR signaling increases autophagy and activates the arachidonic acid cycle, both required for increasing the release of the immunosuppressive mediator, PGE2. Our data reveal that β2-AR signaling triggered by stress is an important physiological regulator of key metabolic pathways in MDSCs, driving their immunosuppressive function. Mohammadpour et al. show that β2-AR signaling in MDSCs alters their metabolic state and increases their immunosuppressive function. Specific processes found to be increased include fatty acid oxidation, oxidative phosphorylation, and autophagy. In addition, these metabolic alterations facilitate an increase in PGE2 production via elevated COX2 expression.
Collapse
Affiliation(s)
- Hemn Mohammadpour
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Cameron R MacDonald
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Philip L McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
71
|
Balaian E, Wobus M, Bornhäuser M, Chavakis T, Sockel K. Myelodysplastic Syndromes and Metabolism. Int J Mol Sci 2021; 22:ijms222011250. [PMID: 34681910 PMCID: PMC8541058 DOI: 10.3390/ijms222011250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/01/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are acquired clonal stem cell disorders exhibiting ineffective hematopoiesis, dysplastic cell morphology in the bone marrow, and peripheral cytopenia at early stages; while advanced stages carry a high risk for transformation into acute myeloid leukemia (AML). Genetic alterations are integral to the pathogenesis of MDS. However, it remains unclear how these genetic changes in hematopoietic stem and progenitor cells (HSPCs) occur, and how they confer an expansion advantage to the clones carrying them. Recently, inflammatory processes and changes in cellular metabolism of HSPCs and the surrounding bone marrow microenvironment have been associated with an age-related dysfunction of HSPCs and the emergence of genetic aberrations related to clonal hematopoiesis of indeterminate potential (CHIP). The present review highlights the involvement of metabolic and inflammatory pathways in the regulation of HSPC and niche cell function in MDS in comparison to healthy state and discusses how such pathways may be amenable to therapeutic interventions.
Collapse
Affiliation(s)
- Ekaterina Balaian
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: (E.B.); (K.S.)
| | - Manja Wobus
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
| | - Martin Bornhäuser
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- National Center for Tumor Diseases, Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Triantafyllos Chavakis
- National Center for Tumor Diseases, Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus Dresden, 01307 Dresden, Germany
| | - Katja Sockel
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- Correspondence: (E.B.); (K.S.)
| |
Collapse
|
72
|
Yan RL, Chen RH. Autophagy and cancer metabolism-The two-way interplay. IUBMB Life 2021; 74:281-295. [PMID: 34652063 DOI: 10.1002/iub.2569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022]
Abstract
Autophagy is an intracellular catabolic process that degrades cytoplasmic components for recycling in response to stressed conditions, such as nutrient deprivation. Dysregulation of autophagy is associated with various diseases, including cancer. Although autophagy plays dichotomous and context-dependent roles in cancer, evidence has emerged that cancer cells exploit autophagy for metabolic adaptation. Autophagy is upregulated in many cancer types through tumor cell-intrinsic proliferation demands and the hypoxic and nutrient-limited tumor microenvironment (TME). Autophagy-induced breakdown products then fuel into various metabolic pathways to supply tumor cells with energy and building blocks for biosynthesis and survival. This bidirectional regulation between autophagy and tumor constitutes a vicious cycle to potentiate tumor growth and therapy resistance. In addition, the pro-tumor functions of autophagy are expanded to host, including cells in TME and distant organs. Thus, inhibition of autophagy or autophagy-mediated metabolic reprogramming may be a promising strategy for anticancer therapy. Better understanding the metabolic rewiring mechanisms of autophagy for its pro-tumor effects will provide insights into patient treatment.
Collapse
Affiliation(s)
- Reui-Liang Yan
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Ruey-Hwa Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
73
|
Abstract
Tumour initiation and progression requires the metabolic reprogramming of cancer cells. Cancer cells autonomously alter their flux through various metabolic pathways in order to meet the increased bioenergetic and biosynthetic demand as well as mitigate oxidative stress required for cancer cell proliferation and survival. Cancer driver mutations coupled with environmental nutrient availability control flux through these metabolic pathways. Metabolites, when aberrantly accumulated, can also promote tumorigenesis. The development and application of new technologies over the last few decades has not only revealed the heterogeneity and plasticity of tumours but also allowed us to uncover new metabolic pathways involved in supporting tumour growth. The tumour microenvironment (TME), which can be depleted of certain nutrients, forces cancer cells to adapt by inducing nutrient scavenging mechanisms to sustain cancer cell proliferation. There is growing appreciation that the metabolism of cell types other than cancer cells within the TME, including endothelial cells, fibroblasts and immune cells, can modulate tumour progression. Because metastases are a major cause of death of patients with cancer, efforts are underway to understand how metabolism is harnessed by metastatic cells. Additionally, there is a new interest in exploiting cancer genetic analysis for patient stratification and/or dietary interventions in combination with therapies that target metabolism. In this Perspective, we highlight these main themes that are currently under investigation in the context of in vivo tumour metabolism, specifically emphasizing questions that remain unanswered.
Collapse
Affiliation(s)
| | - Navdeep S Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
74
|
Porin 1 Modulates Autophagy in Yeast. Cells 2021; 10:cells10092416. [PMID: 34572064 PMCID: PMC8464718 DOI: 10.3390/cells10092416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Autophagy is a cellular recycling program which efficiently reduces the cellular burden of ageing. Autophagy is characterised by nucleation of isolation membranes, which grow in size and further expand to form autophagosomes, engulfing cellular material to be degraded by fusion with lysosomes (vacuole in yeast). Autophagosomal membranes do not bud from a single cell organelle, but are generated de novo. Several lipid sources for autophagosomal membranes have been identified, but the whole process of their generation is complex and not entirely understood. In this study, we investigated how the mitochondrial outer membrane protein porin 1 (Por1), the yeast orthologue of mammalian voltage-dependent anion channel (VDAC), affects autophagy in yeast. We show that POR1 deficiency reduces the autophagic capacity and leads to changes in vacuole and lipid homeostasis. We further investigated whether limited phosphatidylethanolamine (PE) availability in por1∆ was causative for reduced autophagy by overexpression of the PE-generating phosphatidylserine decarboxylase 1 (Psd1). Altogether, our results show that POR1 deficiency is associated with reduced autophagy, which can be circumvented by additional PSD1 overexpression. This suggests a role for Por1 in Psd1-mediated autophagy regulation.
Collapse
|
75
|
Mukhopadhyay S, Mahapatra KK, Praharaj PP, Patil S, Bhutia SK. Recent progress of autophagy signaling in tumor microenvironment and its targeting for possible cancer therapeutics. Semin Cancer Biol 2021; 85:196-208. [PMID: 34500075 DOI: 10.1016/j.semcancer.2021.09.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 02/08/2023]
Abstract
Autophagy, a lysosomal catabolic process, involves degradation of cellular materials, protein aggregate, and dysfunctional organelles to maintain cellular homeostasis. Strikingly, autophagy exhibits a dual-sided role in cancer; on the one hand, it promotes clearance of transformed cells and inhibits tumorigenesis, while cytoprotective autophagy has a role in sustaining cancer. The autophagy signaling in the tumor microenvironment (TME) during cancer growth and therapy is not adequately understood. The review highlights the role of autophagy signaling pathways to support cancer growth and progression in adaptation to the oxidative and hypoxic context of TME. Furthermore, autophagy contributes to regulating the metabolic switch for generating sufficient levels of high-energy metabolites, including amino acids, ketones, glutamine, and free fatty acids for cancer cell survival. Interestingly, autophagy has a critical role in modulating the tumor-associated fibroblast resulting in different cytokines and paracrine signaling mediated angiogenesis and invasion of pre-metastatic niches to secondary tumor sites. Moreover, autophagy promotes immune evasion to inhibit antitumor immunity, and autophagy inhibitors enhance response to immunotherapy with infiltration of immune cells to the TME niche. Furthermore, autophagy in TME maintains and supports the survival of cancer stem cells resulting in chemoresistance and therapy recurrence. Presently, drug repurposing has enabled the use of lysosomal inhibitor-based antimalarial drugs like chloroquine and hydroxychloroquine as clinically available autophagy inhibitors in cancer therapy. We focus on the recent developments of multiple autophagy modulators from pre-clinical trials and the challenges in developing autophagy-based cancer therapy.
Collapse
Affiliation(s)
- Subhadip Mukhopadhyay
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Shankargouda Patil
- Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Saudi Arabia
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India.
| |
Collapse
|
76
|
Poillet-Perez L, Sarry JE, Joffre C. Autophagy is a major metabolic regulator involved in cancer therapy resistance. Cell Rep 2021; 36:109528. [PMID: 34407408 DOI: 10.1016/j.celrep.2021.109528] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/13/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy sustains cellular homeostasis and metabolism in numerous diseases. By regulating cancer metabolism, both tumor and microenvironmental autophagy promote tumor growth. However, autophagy can support cancer progression through other biological functions such as immune response regulation or cytokine/growth factor secretion. Moreover, autophagy is induced in numerous tumor types as a resistance mechanism following therapy, highlighting autophagy inhibition as a promising target for anti-cancer therapy. Thus, better understanding the mechanisms involved in tumor growth and resistance regulation through autophagy, which are not fully understood, will provide insights into patient treatment.
Collapse
Affiliation(s)
- Laura Poillet-Perez
- Cancer Research Centre of Toulouse, UMR1037 Inserm, UMR5077 CNRS, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, 31037 Toulouse, France.
| | - Jean-Emmanuel Sarry
- Cancer Research Centre of Toulouse, UMR1037 Inserm, UMR5077 CNRS, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, 31037 Toulouse, France
| | - Carine Joffre
- Cancer Research Centre of Toulouse, UMR1037 Inserm, UMR5077 CNRS, Université de Toulouse 3 Paul Sabatier, Equipe Labellisée LIGUE 2018, 31037 Toulouse, France.
| |
Collapse
|
77
|
Mikubo M, Inoue Y, Liu G, Tsao MS. Mechanism of Drug Tolerant Persister Cancer Cells: The Landscape and Clinical Implication for Therapy. J Thorac Oncol 2021; 16:1798-1809. [PMID: 34352380 DOI: 10.1016/j.jtho.2021.07.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/30/2021] [Accepted: 07/18/2021] [Indexed: 01/06/2023]
Abstract
A minor population of cancer cells may evade cell death from chemotherapy and targeted therapy by entering a reversible slow proliferation state known as the drug tolerant persister (DTP) state. This DTP state can allow cancer cells to survive drug therapy long enough for additional mechanisms of acquired drug resistance to develop. Thus, cancer persistence is a major obstacle to curing cancers, where insight into the biology of DTP cells and therapeutic strategies targeting this mechanism can have considerable clinical implications. There is emerging evidence that DTP cells adapt to new environments through epigenomic modification, transcriptomic regulation, flexible energy metabolism, and interactions with the tumor microenvironment. Herein, we review and discuss the various proposed mechanisms of cancer persister cells and the molecular features underlying the DTP state, with insights into the potential therapeutic strategies to conquer DTP cells and prevent cancer recurrence or therapeutic failures.
Collapse
Affiliation(s)
- Masashi Mikubo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yoshiaki Inoue
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Medical Biophysics, University Health Network, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
78
|
Li L, Qi R, Zhang L, Yu Y, Hou J, Gu Y, Song D, Wang X. Potential biomarkers and targets of mitochondrial dynamics. Clin Transl Med 2021; 11:e529. [PMID: 34459143 PMCID: PMC8351522 DOI: 10.1002/ctm2.529] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial dysfunction contributes to the imbalance of cellular homeostasis and the development of diseases, which is regulated by mitochondria-associated factors. The present review aims to explore the process of the mitochondrial quality control system as a new source of the potential diagnostic biomarkers and/or therapeutic targets for diseases, including mitophagy, mitochondrial dynamics, interactions between mitochondria and other organelles (lipid droplets, endoplasmic reticulum, endosomes, and lysosomes), as well as the regulation and posttranscriptional modifications of mitochondrial DNA/RNA (mtDNA/mtRNA). The direct and indirect influencing factors were especially illustrated in understanding the interactions among regulators of mitochondrial dynamics. In addition, mtDNA/mtRNAs and proteomic profiles of mitochondria in various lung diseases were also discussed as an example. Thus, alternations of mitochondria-associated regulators can be a new category of biomarkers and targets for disease diagnosis and therapy.
Collapse
Affiliation(s)
- Liyang Li
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Ruixue Qi
- Jinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghaiChina
| | - Linlin Zhang
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Yuexin Yu
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Jiayun Hou
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Yutong Gu
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Dongli Song
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| | - Xiangdong Wang
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Clinical BioinformaticsShanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Jinshan Hospital Centre for Tumor Diagnosis and TherapyFudan University Shanghai Medical CollegeShanghaiChina
| |
Collapse
|
79
|
Wu R, Wang X, Shao Y, Jiang Y, Zhou Y, Lu C. NFATc4 mediates ethanol-triggered hepatocyte senescence. Toxicol Lett 2021; 350:10-21. [PMID: 34192554 DOI: 10.1016/j.toxlet.2021.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Hepatocyte senescence is a core event that mediates the occurrence and development of alcoholic liver disease. Nuclear factor of activated T-cells 4 (NFATc4) is a key driver of nonalcoholic steatohepatitis. However, little was known about the implication of NFATc4 for alcoholic liver disease. This study was aimed to investigate the role of NFATc4 in hepatocyte senescence and further elucidate the underlying mechanism. METHODS Real-time PCR, Western blot, immunofluorescence staining, and enzyme-linked immunosorbent assay were performed to explore the role of NFATc4 in hepatocyte senescence. RESULTS NFATc4 was induced in ethanol-incubated hepatocytes. NFATc4 knockdown recovered cell viability and reduced the release of aspartate transaminase, alanine transaminase, and lactic dehydrogenase from ethanol-incubated hepatocytes. NFATc4 knockdown protected mice from alcoholic liver injury and inflammation. NFATc4 knockdown counteracted ethanol-induced hepatocyte senescence, evidenced by decreased senescence-associated β-galactosidase positivity and reduced p16, p21, HMGA1, and γH2AX, which was validated in in vivo studies. Peroxisome proliferator-activated receptor (PPAR)γ was inhibited by NFATc4 in ethanol-treated hepatocytes. PPARγ deficiency abrogated the inhibitory effects of NFATc4 knockdown on hepatocyte senescence, oxidative stress, and hepatic steatosis in mice with alcoholic liver disease. CONCLUSIONS This work discovered that ethanol enhanced NFATc4 expression, which further triggered hepatocyte senescence via repression of PPARγ.
Collapse
Affiliation(s)
- Ruoman Wu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xinqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yunyun Shao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yiming Jiang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Ying Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
80
|
Dharmalingam P, Venkatakrishnan K, Tan B. Predicting Metastasis from Cues of Metastatic Cancer Stem-like Cells-3D-Ultrasensitive Metasensor at a Single-Cell Level. ACS NANO 2021; 15:9967-9986. [PMID: 34081852 DOI: 10.1021/acsnano.1c01436] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Metastasis is the primary reason for treatment failure and cancer-related deaths. Hence forecasting the disease in its primary state can advance the prognosis. However, existing techniques fail to reveal the tumor heterogeneity or its evolutionary cascades; hence they are not feasible to predict the onset of metastatic cancer. The key to metastasis originates from the primary tumor cells, evolving by inheriting multistep sequential cue signals. We have identified this specific population, termed metastatic cancer stem-like cells (MCSCs), to foresee cancer's ability to metastasize. An invasive property renders MCSCs nonadherent, summoning a powerful technique to forecast metastasis. Thus, we have generated an ultrasensitive 3D-metasensor to efficiently capture and investigate MCSCs and magnify the vital premetastatic signals from a single cell. We developed 3D-metasensor by an ultrafast laser ionization technique, consisting of self-assembled three-dimensionally organized nanoprobes incorporated with dopant functionalities. This distinct methodology establishes attachment with nonadherent MCSCs, elevates Raman activity, and enables probing of consequent signals (metabolic, proliferation, and metastatic) specifically altered in MCSCs. Extensive analysis using prediction tools-the area under the curve (AUC) and principal component analysis (PCA)-revealed high sensitivity (100%) and specificity (80%) to differentiate the MCSCs from other populations. Further, investigation reveals that the cue signal level from MCSCs of primary cancer is analogous to MCSCs from higher-level tumors, disclosing the relative dependence to estimate the primary tumor's capacity to metastasize. Multiple spectrum evaluation using the metasensor pinpoint the dynamic cues in MCSCs predict the onset of metastasis; thus, exploring these metastasis hallmarks can enhance prognosis and revolutionize therapy strategies.
Collapse
Affiliation(s)
- Priya Dharmalingam
- Ultrashort Laser Nano Manufacturing Research Facility, Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Institute for Biomedical Engineering, Science and Technology (I-BEST), Partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Nano Characterization Laboratory, Department of Aerospace Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface Facility, Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Krishnan Venkatakrishnan
- Ultrashort Laser Nano Manufacturing Research Facility, Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Affiliate Scientist, Keenan Research Center, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario M5B 1T8, Canada
| | - Bo Tan
- Nano Characterization Laboratory, Department of Aerospace Engineering, Ryerson University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Affiliate Scientist, Keenan Research Center, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario M5B 1T8, Canada
| |
Collapse
|
81
|
Autophagy a Close Relative of AML Biology. BIOLOGY 2021; 10:biology10060552. [PMID: 34207482 PMCID: PMC8235674 DOI: 10.3390/biology10060552] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary Acute myeloid leukemia (AML) is the most common acute leukemia in adults. Despite a high rate of complete remission following conventional chemotherapy, the prognosis remains poor due to frequent relapses caused by relapse-initiating leukemic cells (RICs), which are resistant to chemotherapies. While the development of new targeted therapies holds great promise (e.g., molecules targeting IDH1/2, FLT3, BCL2), relapses still occur. Therefore, a paramount issue in the elimination of RICs is to decipher the AML resistance mechanisms. Thus, it has been recently shown that AML cells exhibit metabolic changes in response to chemotherapy or targeted therapies. Autophagy is a major regulator of cell metabolism, involved in maintaining cancer state, metastasis, and resistance to anticancer therapy. However, whether autophagy acts as a tumor suppressor or promoter in AML is still a matter of debate. Therefore, depending on molecular AML subtypes or treatments used, a better understanding of the role of autophagy is needed to determine whether its modulation could result in a clinical benefit. Abstract Autophagy, which literally means “eat yourself”, is more than just a lysosomal degradation pathway. It is a well-known regulator of cellular metabolism and a mechanism implicated in tumor initiation/progression and therapeutic resistance in many cancers. However, whether autophagy acts as a tumor suppressor or promoter is still a matter of debate. In acute myeloid leukemia (AML), it is now proven that autophagy supports cell proliferation in vitro and leukemic progression in vivo. Mitophagy, the specific degradation of mitochondria through autophagy, was recently shown to be required for leukemic stem cell functions and survival, highlighting the prominent role of this selective autophagy in leukemia initiation and progression. Moreover, autophagy in AML sustains fatty acid oxidation through lipophagy to support mitochondrial oxidative phosphorylation (OxPHOS), a hallmark of chemotherapy-resistant cells. Nevertheless, in the context of therapy, in AML, as well as in other cancers, autophagy could be either cytoprotective or cytotoxic, depending on the drugs used. This review summarizes the recent findings that mechanistically show how autophagy favors leukemic transformation of normal hematopoietic stem cells, as well as AML progression and also recapitulates its ambivalent role in resistance to chemotherapies and targeted therapies.
Collapse
|
82
|
Small molecule probes for targeting autophagy. Nat Chem Biol 2021; 17:653-664. [PMID: 34035513 DOI: 10.1038/s41589-021-00768-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 02/08/2021] [Indexed: 02/02/2023]
Abstract
Autophagy is implicated in a wide range of (patho)physiological processes including maintenance of cellular homeostasis, neurodegenerative disorders, aging and cancer. As such, small molecule autophagy modulators are in great demand, both for their ability to act as tools to better understand this essential process and as potential therapeutics. Despite substantial advances in the field, major challenges remain in the development and comprehensive characterization of probes that are specific to autophagy. In this Review, we discuss recent developments in autophagy-modulating small molecules, including the specific challenges faced in the development of activators and inhibitors, and recommend guidelines for their use. Finally, we discuss the potential to hijack the process for targeted protein degradation, an area of great importance in chemical biology and drug discovery.
Collapse
|
83
|
Stubbins RJ, Maksakova IA, Sanford DS, Rouhi A, Kuchenbauer F. Mitochondrial metabolism: powering new directions in acute myeloid leukemia. Leuk Lymphoma 2021; 62:2331-2341. [PMID: 34060970 DOI: 10.1080/10428194.2021.1910685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
There has been an explosion of knowledge about the role of metabolism and the mitochondria in acute myeloid leukemia (AML). We have also recently seen several waves of novel therapies change the treatment landscape for AML, such as the selective B-cell lymphoma 2 (BCL-2) inhibitor venetoclax. In this new context, we review the rapidly advancing literature on the role of metabolism and the mitochondria in AML pathogenesis, and how these are interwoven with the mechanisms of action for novel therapeutics in AML. We also review the role of oxidative phosphorylation (OxPhos) in maintaining leukemia stem cells (LSCs), how recurrent genomic alterations in AML alter downstream metabolism, and focus on how the BCL-2 pathway and the mitochondria are inextricably linked in AML. Thus, we provide an overview of the mitochondria and metabolism in the context of our new therapeutic world for AML and outline how targeting these vulnerabilities may produce novel therapeutic strategies.
Collapse
Affiliation(s)
- Ryan J Stubbins
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Irina A Maksakova
- Terry Fox Laboratory, BC Cancer Research Centre, University of British Columbia, Vancouver, Canada
| | - David S Sanford
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Arefeh Rouhi
- Terry Fox Laboratory, BC Cancer Research Centre, University of British Columbia, Vancouver, Canada
| | - Florian Kuchenbauer
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, Canada.,Terry Fox Laboratory, BC Cancer Research Centre, University of British Columbia, Vancouver, Canada
| |
Collapse
|
84
|
Aoyama-Ishiwatari S, Hirabayashi Y. Endoplasmic Reticulum-Mitochondria Contact Sites-Emerging Intracellular Signaling Hubs. Front Cell Dev Biol 2021; 9:653828. [PMID: 34095118 PMCID: PMC8172986 DOI: 10.3389/fcell.2021.653828] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/06/2021] [Indexed: 01/04/2023] Open
Abstract
It has become apparent that our textbook illustration of singular isolated organelles is obsolete. In reality, organelles form complex cooperative networks involving various types of organelles. Light microscopic and ultrastructural studies have revealed that mitochondria-endoplasmic reticulum (ER) contact sites (MERCSs) are abundant in various tissues and cell types. Indeed, MERCSs have been proposed to play critical roles in various biochemical and signaling functions such as Ca2+ homeostasis, lipid transfer, and regulation of organelle dynamics. While numerous proteins involved in these MERCS-dependent functions have been reported, how they coordinate and cooperate with each other has not yet been elucidated. In this review, we summarize the functions of mammalian proteins that localize at MERCSs and regulate their formation. We also discuss potential roles of the MERCS proteins in regulating multiple organelle contacts.
Collapse
Affiliation(s)
| | - Yusuke Hirabayashi
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
85
|
Pan XJ, Misrani A, Tabassum S, Yang L. Mitophagy pathways and Alzheimer's disease: From pathogenesis to treatment. Mitochondrion 2021; 59:37-47. [PMID: 33872797 DOI: 10.1016/j.mito.2021.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is an age-dependent, incurable mental illness that is associated with the accumulation of aggregates of amyloid-beta (Aβ) and hyperphosphorylated tau fragments (p-tau). Detailed studies on postmortem AD brains, cell lines, and mouse models of AD have shown that numerous cellular alterations, including mitochondrial deficits, synaptic disruption and glial/astrocytic activation, are involved in the disease process. Mitophagy is a cellular process by which damaged/weakened mitochondria are selectively eliminated from the cell. In AD, impairments in mitophagy trigger the gradual accumulation of defective mitochondria. This review will focus on the recent progress in understanding the molecular mechanisms and pathological role of mitophagy and its implications for AD pathogenesis. We will also discuss the novel concept of the regulation of mitophagy as a therapeutic avenue for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Xian-Ji Pan
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Afzal Misrani
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Sidra Tabassum
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Li Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510006, China.
| |
Collapse
|
86
|
Bustos G, Ahumada-Castro U, Silva-Pavez E, Puebla A, Lovy A, Cesar Cardenas J. The ER-mitochondria Ca 2+ signaling in cancer progression: Fueling the monster. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:49-121. [PMID: 34392932 DOI: 10.1016/bs.ircmb.2021.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is a leading cause of death worldwide. All major tumor suppressors and oncogenes are now recognized to have fundamental connections with metabolic pathways. A hallmark feature of cancer cells is a reprogramming of their metabolism even when nutrients are available. Increasing evidence indicates that most cancer cells rely on mitochondrial metabolism to sustain their energetic and biosynthetic demands. Mitochondria are functionally and physically coupled to the endoplasmic reticulum (ER), the major calcium (Ca2+) storage organelle in mammalian cells, through special domains known as mitochondria-ER contact sites (MERCS). In this domain, the release of Ca2+ from the ER is mainly regulated by inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), a family of Ca2+ release channels activated by the ligand IP3. IP3R mediated Ca2+ release is transferred to mitochondria through the mitochondrial Ca2+ uniporter (MCU). Once in the mitochondrial matrix, Ca2+ activates several proteins that stimulate mitochondrial performance. The role of IP3R and MCU in cancer, as well as the other proteins that enable the Ca2+ communication between these two organelles is just beginning to be understood. Here, we describe the function of the main players of the ER mitochondrial Ca2+ communication and discuss how this particular signal may contribute to the rise and development of cancer traits.
Collapse
Affiliation(s)
- Galdo Bustos
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Eduardo Silva-Pavez
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Andrea Puebla
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Alenka Lovy
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Department of Neuroscience, Center for Neuroscience Research, Tufts School of Medicine, Boston, MA, United States.
| | - J Cesar Cardenas
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, United States; Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, United States.
| |
Collapse
|
87
|
Culp-Hill R, D'Alessandro A, Pietras EM. Extinguishing the Embers: Targeting AML Metabolism. Trends Mol Med 2021; 27:332-344. [PMID: 33121874 PMCID: PMC8005405 DOI: 10.1016/j.molmed.2020.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is a cancer derived from the myeloid lineage of blood cells, characterized by overproduction of leukemic blasts. Although therapeutic improvements have made a significant impact on the outcomes of patients with AML, survival rates remain low due to a high incidence of relapse. Similar to how wildfires can reignite from hidden embers not extinguished from an initial round of firefighting, leukemic stem cells (LSCs) are the embers remaining after completion of traditional chemotherapeutic treatments. LSCs exhibit a unique metabolic profile and contain metabolically distinct subpopulations. In this review, we detail the metabolic features of LSCs and how thetse characteristics promote resistance to traditional chemotherapy. We also discuss new therapeutic approaches that target metabolic vulnerabilities of LSC to selectively eradicate them.
Collapse
Affiliation(s)
- Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
88
|
Mooli RGR, Rodriguez J, Takahashi S, Solanki S, Gonzalez FJ, Ramakrishnan SK, Shah YM. Hypoxia via ERK Signaling Inhibits Hepatic PPARα to Promote Fatty Liver. Cell Mol Gastroenterol Hepatol 2021; 12:585-597. [PMID: 33798787 PMCID: PMC8258975 DOI: 10.1016/j.jcmgh.2021.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Fatty liver or nonalcoholic fatty liver disease (NAFLD) is the most common liver disease associated with comorbidities such as insulin resistance and cardiovascular and metabolic diseases. Chronic activation of hypoxic signaling, in particular, hypoxia-inducible factor (HIF)2α, promotes NAFLD progression by repressing genes involved in fatty acid β-oxidation through unclear mechanisms. Therefore, we assessed the precise mechanism by which HIF2α promotes fatty liver and its physiological relevance in metabolic homeostasis. METHODS Primary hepatocytes from VHL (VhlΔHep) and PPARα (Ppara-null) knockout mice that were loaded with fatty acids, murine dietary protocols to induce hepatic steatosis, and fasting-refeeding dietary regimen approaches were used to test our hypothesis. RESULTS Inhibiting autophagy using chloroquine did not decrease lipid contents in VhlΔHep primary hepatocytes. Inhibition of ERK using MEK inhibitor decreased lipid contents in primary hepatocytes from a genetic model of constitutive HIF activation and primary hepatocytes loaded with free fatty acids. Moreover, MEK-ERK inhibition potentiated ligand-dependent activation of PPARα. We also show that MEK-ERK inhibition improved diet-induced hepatic steatosis, which is associated with the induction of PPARα target genes. During fasting, fatty acid β-oxidation is induced by PPARα, and refeeding inhibits β-oxidation. Our data show that ERK is involved in the post-prandial repression of hepatic PPARα signaling. CONCLUSIONS Overall, our results demonstrate that ERK activated by hypoxia signaling plays a crucial role in fatty acid β-oxidation genes by repressing hepatocyte PPARα signaling.
Collapse
Affiliation(s)
- Raja Gopal Reddy Mooli
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessica Rodriguez
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shogo Takahashi
- Departments of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia; National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sumeet Solanki
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Frank J Gonzalez
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sadeesh K Ramakrishnan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
89
|
Giulitti F, Petrungaro S, Mandatori S, Tomaipitinca L, de Franchis V, D'Amore A, Filippini A, Gaudio E, Ziparo E, Giampietri C. Anti-tumor Effect of Oleic Acid in Hepatocellular Carcinoma Cell Lines via Autophagy Reduction. Front Cell Dev Biol 2021; 9:629182. [PMID: 33614661 PMCID: PMC7892977 DOI: 10.3389/fcell.2021.629182] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Oleic acid (OA) is a component of the olive oil. Beneficial health effects of olive oil are well-known, such as protection against liver steatosis and against some cancer types. In the present study, we focused on OA effects in hepatocellular carcinoma (HCC), investigating responses to OA treatment (50–300 μM) in HCC cell lines (Hep3B and Huh7.5) and in a healthy liver-derived human cell line (THLE-2). Upon OA administration higher lipid accumulation, perilipin-2 increase, and autophagy reduction were observed in HCC cells as compared to healthy cells. OA in the presence of 10% FBS significantly reduced viability of HCC cell lines at 300 μM through Alamar Blue staining evaluation, and reduced cyclin D1 expression in a dose-dependent manner while it was ineffective on healthy hepatocytes. Furthermore, OA increased cell death by about 30%, inducing apoptosis and necrosis in HCC cells but not in healthy hepatocytes at 300 μM dosage. Moreover, OA induced senescence in Hep3B, reduced P-ERK in both HCC cell lines and significantly inhibited the antiapoptotic proteins c-Flip and Bcl-2 in HCC cells but not in healthy hepatocytes. All these results led us to conclude that different cell death processes occur in these two HCC cell lines upon OA treatment. Furthermore, 300 μM OA significantly reduced the migration and invasion of both HCC cell lines, while it has no effects on healthy cells. Finally, we investigated autophagy role in OA-dependent effects by using the autophagy inducer torin-1. Combined OA/torin-1 treatment reduced lipid accumulation and cell death as compared to single OA treatment. We therefore concluded that OA effects in HCC cells lines are, at least, in part dependent on OA-induced autophagy reduction. In conclusion, we report for the first time an autophagy dependent relevant anti-cancer effect of OA in human hepatocellular carcinoma cell lines.
Collapse
Affiliation(s)
- Federico Giulitti
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Simonetta Petrungaro
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Sara Mandatori
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Luana Tomaipitinca
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Valerio de Franchis
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonella D'Amore
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonio Filippini
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Elio Ziparo
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Claudia Giampietri
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
90
|
Tumor Metabolic Reprogramming by Adipokines as a Critical Driver of Obesity-Associated Cancer Progression. Int J Mol Sci 2021; 22:ijms22031444. [PMID: 33535537 PMCID: PMC7867092 DOI: 10.3390/ijms22031444] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Adiposity is associated with an increased risk of various types of carcinoma. One of the plausible mechanisms underlying the tumor-promoting role of obesity is an aberrant secretion of adipokines, a group of hormones secreted from adipose tissue, which have exhibited both oncogenic and tumor-suppressing properties in an adipokine type- and context-dependent manner. Increasing evidence has indicated that these adipose tissue-derived hormones differentially modulate cancer cell-specific metabolism. Some adipokines, such as leptin, resistin, and visfatin, which are overproduced in obesity and widely implicated in different stages of cancer, promote cellular glucose and lipid metabolism. Conversely, adiponectin, an adipokine possessing potent anti-tumor activities, is linked to a more favorable metabolic phenotype. Adipokines may also play a pivotal role under the reciprocal regulation of metabolic rewiring of cancer cells in tumor microenvironment. Given the fact that metabolic reprogramming is one of the major hallmarks of cancer, understanding the modulatory effects of adipokines on alterations in cancer cell metabolism would provide insight into the crosstalk between obesity, adipokines, and tumorigenesis. In this review, we summarize recent insights into putative roles of adipokines as mediators of cellular metabolic rewiring in obesity-associated tumors, which plays a crucial role in determining the fate of tumor cells.
Collapse
|
91
|
Mitochondria Associated Membranes (MAMs): Architecture and physiopathological role. Cell Calcium 2021; 94:102343. [PMID: 33418313 DOI: 10.1016/j.ceca.2020.102343] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/27/2020] [Accepted: 12/27/2020] [Indexed: 12/17/2022]
Abstract
In the last decades, the communication between the Endoplasmic reticulum (ER) and mitochondria has obtained great attention: mitochondria-associated membranes (MAMs), which represent the contact sites between the two organelles, have indeed emerged as central hub involved in different fundamental cell processes, such as calcium signalling, apoptosis, autophagy and lipid biosynthesis. Consistently, dysregulation of ER-mitochondria crosstalk has been associated with different pathological conditions, ranging from diabetes to cancer and neurodegenerative diseases. In this review, we will try to summarize the current knowledge on MAMs' structure and functions in health and their relevance for human diseases.
Collapse
|
92
|
Lechuga-Vieco AV, Justo-Méndez R, Enríquez JA. Not all mitochondrial DNAs are made equal and the nucleus knows it. IUBMB Life 2020; 73:511-529. [PMID: 33369015 PMCID: PMC7985871 DOI: 10.1002/iub.2434] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/06/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
The oxidative phosphorylation (OXPHOS) system is the only structure in animal cells with components encoded by two genomes, maternally transmitted mitochondrial DNA (mtDNA), and biparentally transmitted nuclear DNA (nDNA). MtDNA‐encoded genes have to physically assemble with their counterparts encoded in the nucleus to build together the functional respiratory complexes. Therefore, structural and functional matching requirements between the protein subunits of these molecular complexes are rigorous. The crosstalk between nDNA and mtDNA needs to overcome some challenges, as the nuclear‐encoded factors have to be imported into the mitochondria in a correct quantity and match the high number of organelles and genomes per mitochondria that encode and synthesize their own components locally. The cell is able to sense the mito‐nuclear match through changes in the activity of the OXPHOS system, modulation of the mitochondrial biogenesis, or reactive oxygen species production. This implies that a complex signaling cascade should optimize OXPHOS performance to the cellular‐specific requirements, which will depend on cell type, environmental conditions, and life stage. Therefore, the mitochondria would function as a cellular metabolic information hub integrating critical information that would feedback the nucleus for it to respond accordingly. Here, we review the current understanding of the complex interaction between mtDNA and nDNA.
Collapse
Affiliation(s)
- Ana Victoria Lechuga-Vieco
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Raquel Justo-Méndez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | | |
Collapse
|
93
|
Mediratta K, El-Sahli S, D’Costa V, Wang L. Current Progresses and Challenges of Immunotherapy in Triple-Negative Breast Cancer. Cancers (Basel) 2020; 12:E3529. [PMID: 33256070 PMCID: PMC7761500 DOI: 10.3390/cancers12123529] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
With improved understanding of the immunogenicity of triple-negative breast cancer (TNBC), immunotherapy has emerged as a promising candidate to treat this lethal disease owing to the lack of specific targets and effective treatments. While immune checkpoint inhibition (ICI) has been effectively used in immunotherapy for several types of solid tumor, monotherapies targeting programmed death 1 (PD-1), its ligand PD-L1, or cytotoxic T lymphocyte-associated protein 4 (CTLA-4) have shown little efficacy for TNBC patients. Over the past few years, various therapeutic candidates have been reviewed, attempting to improve ICI efficacy on TNBC through combinatorial treatment. In this review, we describe the clinical limitations of ICI and illustrate candidates from an immunological, pharmacological, and metabolic perspective that may potentiate therapy to improve the outcomes of TNBC patients.
Collapse
Affiliation(s)
- Karan Mediratta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Sara El-Sahli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Vanessa D’Costa
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
94
|
Bioenergetic Profiling of the Differentiating Human MDS Myeloid Lineage with Low and High Bone Marrow Blast Counts. Cancers (Basel) 2020; 12:cancers12123520. [PMID: 33255926 PMCID: PMC7759906 DOI: 10.3390/cancers12123520] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Myelodysplastic syndromes (MDS) encompass a very heterogeneous group of clonal hematopoietic stem cell differentiation disorders with malignant potential, an elusive pathobiology, and a poor prognosis. Given that the bioenergetic profile of the hematopoietic precursors is central to their effective differentiation, we investigated the metabolic status of a human differentiating MDS bone marrow-derived myeloid lineage. Our findings suggest that a perturbed metabolism underlies the syndrome’s pathogenesis and also determines the disease severity. We also propose that these bioenergetic alterations are essentially featured in and indeed drive the process of leukemic transformation. Our data not only offer novel insight into the elusive MDS pathophysiology, but also change our viewpoint on MDS-related acute myeloid leukemia biology. Abstract Myelodysplastic syndromes (MDS) encompass a very heterogeneous group of clonal hematopoietic stem cell differentiation disorders with malignant potential and an elusive pathobiology. Given the central role of metabolism in effective differentiation, we performed an untargeted metabolomic analysis of differentiating myeloid lineage cells from MDS bone marrow aspirates that exhibited <5% (G1) or ≥5% (G2) blasts, in order to delineate its role in MDS severity and malignant potential. Bone marrow aspirates were collected from 14 previously untreated MDS patients (G1, n = 10 and G2, n = 4) and age matched controls (n = 5). Following myeloid lineage cell isolation, untargeted mass spectrometry-based metabolomics analysis was performed. Data were processed and analyzed using Metabokit. Enrichment analysis was performed using Metaboanalyst v4 employing pathway-associated metabolite sets. We established a bioenergetic profile coordinated by the Warburg phenomenon in both groups, but with a massively different outcome that mainly depended upon its group mitochondrial function and redox state. G1 cells are overwhelmed by glycolytic intermediate accumulation due to failing mitochondria, while the functional electron transport chain and improved redox in G2 compensate for Warburg disruption. Both metabolomes reveal the production and abundance of epigenetic modifiers. G1 and G2 metabolomes differ and eventually determine the MDS clinical phenotype, as well as the potential for malignant transformation.
Collapse
|
95
|
Balgoma D, Gil-de-Gómez L, Montero O. Lipidomics Issues on Human Positive ssRNA Virus Infection: An Update. Metabolites 2020; 10:E356. [PMID: 32878290 PMCID: PMC7569815 DOI: 10.3390/metabo10090356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/29/2022] Open
Abstract
The pathogenic mechanisms underlying the Biology and Biochemistry of viral infections are known to depend on the lipid metabolism of infected cells. From a lipidomics viewpoint, there are a variety of mechanisms involving virus infection that encompass virus entry, the disturbance of host cell lipid metabolism, and the role played by diverse lipids in regard to the infection effectiveness. All these aspects have currently been tackled separately as independent issues and focused on the function of proteins. Here, we review the role of cholesterol and other lipids in ssRNA+ infection.
Collapse
Affiliation(s)
- David Balgoma
- Analytical Pharmaceutical Chemistry, Department of Medicinal Chemistry, Uppsala University, Husarg. 3, 75123 Uppsala, Sweden;
| | - Luis Gil-de-Gómez
- Center of Childhood Cancer Center, Children’s Hospital of Philadelphia, Colket Translational Research Center, 3501 Civic Center Blvd, Philadelphia, PA 19104, USA;
| | - Olimpio Montero
- Spanish National Research Council (CSIC), Boecillo’s Technological Park Bureau, Av. Francisco Vallés 8, 47151 Boecillo, Spain
| |
Collapse
|