1
|
Chen N, Li Z, Liu H, Jiang A, Zhang L, Yan S, He W, Yang J, Liu T. Enhancing PD-1 blockade in NSCLC: Reprogramming tumor immune microenvironment with albumin-bound statins targeting lipid rafts and mitochondrial respiration. Bioact Mater 2025; 49:140-153. [PMID: 40124597 PMCID: PMC11930202 DOI: 10.1016/j.bioactmat.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/09/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) has shown limited response to immunotherapy, primarily due to an immunosuppressive tumor microenvironment characterized by hypoxia and lipid raft formation, which together inhibit T-cell infiltration and function, impeding effective immune responses. To address these challenges, we developed Abstatin, an albumin-bound fluvastatin formulation that targets lipid raft disruption and mitochondrial respiration inhibition, aiming to reduce hypoxia and destabilize lipid rafts to enhance T-cell activity within the tumor. Using bioinformatics analysis, in vitro assays, and in vivo studies in both murine and humanized PDX models, we demonstrated that Abstatin reprograms the NSCLC microenvironment by concurrently lowering hypoxia levels and lipid raft integrity, thereby restoring T-cell infiltration, enhancing cytotoxic T-cell function, and ultimately improving response to Anti-PD-1 therapy. Results showed that Abstatin significantly amplifies Anti-PD-1 efficacy with minimal toxicity, indicating a favorable safety profile for clinical use. This study highlights Abstatin as a promising immunotherapy adjuvant that addresses critical barriers in NSCLC by modulating metabolic pathways linked to immune resistance. Abstatin's approach, which combines modulation of cellular metabolism with immune sensitization, broadens the potential of immunotherapy and provides a practical, scalable strategy to enhance treatment outcomes in NSCLC and potentially other tumors, offering insights into combinatory cancer therapies.
Collapse
Affiliation(s)
- Na Chen
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Zhanfeng Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Heyuan Liu
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Aimin Jiang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, PR China
| | - Liqiang Zhang
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Siqi Yan
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Wangxiao He
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Jingyue Yang
- Department of Clinical Oncology, Air Force Medical University, Xi'an, 710032, PR China
| | - Tianya Liu
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| |
Collapse
|
2
|
Akter R, Hogan MF, Esser N, Barrow BM, Castillo JJ, Boyko EJ, Templin AT, Hull RL, Zraika S, Kahn SE. Increased Steroidogenic Acute Regulatory Protein Contributes to Cholesterol-induced β-Cell Dysfunction. Endocrinology 2025; 166:bqaf027. [PMID: 39928527 PMCID: PMC11833471 DOI: 10.1210/endocr/bqaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/13/2025] [Accepted: 02/07/2025] [Indexed: 02/12/2025]
Abstract
Hypercholesterolemia is often observed in individuals with type 2 diabetes. Cholesterol accumulation in subcellular compartments within islet β-cells can result in insulin secretory dysfunction, which is a key pathological feature of diabetes. Previously, we demonstrated that expression of the mitochondrial cholesterol transport protein, steroidogenic acute regulatory protein (StAR), is induced in islets under conditions of β-cell dysfunction. However, whether it contributes to mitochondrial cholesterol accumulation in β-cells and cholesterol-induced β-cell dysfunction has not been determined. Thus, we sought to examine the role of StAR in isolated mouse islets under conditions of excess exogenous cholesterol. Cholesterol treatment of islets upregulated StAR expression, which was associated with cholesterol accumulation in mitochondria, decreased mitochondrial membrane potential and impaired mitochondrial oxidative phosphorylation. Impaired insulin secretion and reduced islet insulin content were also observed in cholesterol-laden islets. To determine the impact of StAR overexpression in β-cells per se, a lentivirus was used to increase StAR expression in INS-1 cells. Under these conditions, StAR overexpression was sufficient to increase mitochondrial cholesterol content, impair mitochondrial oxidative phosphorylation, and reduce insulin secretion. These findings suggest that elevated cholesterol in diabetes may contribute to β-cell dysfunction via increases in StAR-mediated mitochondrial cholesterol transport and accumulation.
Collapse
Affiliation(s)
- Rehana Akter
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Meghan F Hogan
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, CHU Liège, University of Liège, Liège 4000, Belgium
| | - Breanne M Barrow
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Joseph J Castillo
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Edward J Boyko
- Epidemiologic Research and Information Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Andrew T Templin
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Medicine, Roudebush Veterans Affairs Medical Center and Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rebecca L Hull
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington and Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| |
Collapse
|
3
|
Kharrat F, Capaci V, Conti A, Golino V, Campiglia P, Balasan N, Aloisio M, Licastro D, Monasta L, Caponneto F, Beltrami AP, Romano F, di Lorenzo G, Ricci G, Ura B. A Pilot Study of Exosome Proteomic Profiling Reveals Dysregulated Metabolic Pathways in Endometrial Cancer. Biomedicines 2025; 13:95. [PMID: 39857679 PMCID: PMC11759861 DOI: 10.3390/biomedicines13010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/11/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Endometrial cancer (EC) is the second most frequent gynecological malignant tumor in postmenopausal women. Pathogenic mechanisms related to the onset and development of the disease are still unknown. To identify dysregulated proteins associated with EC we exploited a combined in vitro/in silico approach analyzing the proteome of exosomes with advanced MS techniques and annotating their results by using Chymeris1 AI tools. Methods: To this aim in this pilot study, we performed a deep proteomics analysis with high resolution MS (HRMS), advanced computational tools and western blotting for proteomics data validation. Results: That allowed us to identify 3628 proteins in serum albumin-depleted exosomes from 10 patients with EC compared to 10 healthy controls. This is the largest number of proteins identified in EC serum EVs. After quantification and statistical analysis, we identified 373 significantly (p < 0.05) dysregulated proteins involved in neutrophil and platelet degranulation pathways. A more detailed bioinformatics analysis revealed 61 dysregulated enzymes related to metabolic and catabolic pathways linked to tumor invasion. Through this analysis, we identified 49 metabolic and catabolic pathways related to tumor growth. Conclusions: Altogether, data shed light on the metabolic pathways involved in tumors. This is very important for understanding the metabolism of EC and for the development of new therapies.
Collapse
Affiliation(s)
- Feras Kharrat
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
| | - Valeria Capaci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
| | - Andrea Conti
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
| | - Valentina Golino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy; (V.G.); (P.C.)
- National PhD Program in RNA Therapeutics and Gene Therapy, University of Naples Federico II, 80131 Napoli, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy; (V.G.); (P.C.)
| | - Nour Balasan
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
| | - Michelangelo Aloisio
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy;
| | | | - Lorenzo Monasta
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
| | - Federica Caponneto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (F.C.); (A.P.B.)
| | - Antonio Paolo Beltrami
- Department of Medicine, University of Udine, 33100 Udine, Italy; (F.C.); (A.P.B.)
- Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Federico Romano
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
| | - Giovanni di Lorenzo
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Blendi Ura
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (F.K.); (V.C.); (A.C.); (N.B.); (F.R.); (G.R.); (B.U.)
| |
Collapse
|
4
|
Larrañaga-SanMiguel A, Bengoa-Vergniory N, Flores-Romero H. Crosstalk between mitochondria-ER contact sites and the apoptotic machinery as a novel health meter. Trends Cell Biol 2025; 35:33-45. [PMID: 39379268 DOI: 10.1016/j.tcb.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 10/10/2024]
Abstract
Mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) function as transient signaling platforms that regulate essential cellular functions. MERCS are enriched in specific proteins and lipids that connect mitochondria and the ER together and modulate their activities. Dysregulation of MERCS is associated with several human pathologies including Alzheimer's disease (AD), Parkinson's disease (PD), and cancer. BCL-2 family proteins can locate at MERCS and control essential cellular functions such as calcium signaling and autophagy in addition to their role in mitochondrial apoptosis. Moreover, the BCL-2-mediated apoptotic machinery was recently found to trigger cGAS-STING pathway activation and a proinflammatory response, a recognized hallmark of these diseases that requires mitochondria-ER interplay. This review underscores the pivotal role of MERCS in regulating essential cellular functions, focusing on their crosstalk with BCL-2 family proteins, and discusses how their dysregulation is linked to disease.
Collapse
Affiliation(s)
| | - Nora Bengoa-Vergniory
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy, and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Hector Flores-Romero
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
5
|
Chen J, Gu Z. Cholesterol mediates the potential adverse influence of graphene quantum dots on placental lipid membrane model. Sci Rep 2024; 14:31342. [PMID: 39732901 PMCID: PMC11682271 DOI: 10.1038/s41598-024-82805-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
Nanomaterial-biomembrane interactions constitute a critical biological process in assessing the toxicity of such materials in theoretical studies. However, many investigations simplify these interactions by using membrane models containing only one or a few lipid types, deviating significantly from the complexity of real membrane compositions. In particular, cholesterol, a ubiquitous lipid essential for regulating membrane fluidity and closely linked to various diseases, is often overlooked. Consequently, the role of cholesterol in nanomaterial-biomembrane interactions remains poorly understood. In this study, we employ molecular dynamics (MD) simulations to explore the effect of graphene quantum dots (GQDs) on a realistic placental lipid membrane model, aiming to elucidate the role of cholesterol in these interactions. Our MD results reveal that both GQD monomers and clusters can spontaneously insert into the placental lipid membrane model, driven by strong van der Waals interaction energy. Further analyses indicate that cholesterol and POPC lipids primarily contribute to interfacial interactions. Notably, cholesterol can be squeezed into the bilayer interface, forming a unique structure where it is sandwiched between the GQD cluster and the membrane's bottom leaflet. More significantly, cholesterol, together with the GQD cluster, exhibits free lateral movement, suggesting a strong affinity of cholesterol for GQD clusters. These findings highlight the critical role of cholesterol in mediating GQD insertion into the biomembrane. Structural analyses of the membrane further demonstrate deformation of the placental lipid membrane model during GQD penetration. Finally, free energy calculations confirm that the insertion of both GQD monomers and clusters into the placental lipid membrane model is energetically favorable. Overall, this study not only sheds new light on the potential harmful effects of GQDs on realistic placental membranes but also provides the first theoretical evidence of the pivotal role of cholesterol in nanomaterial-biomembrane interactions, contributing to a deeper understanding of nanomaterial-cell membrane interactions.
Collapse
Affiliation(s)
- Jiao Chen
- Department of Medical Genetics and Prenatal Diagnosis, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China.
| | - Zonglin Gu
- College of Physical Science and Technology, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| |
Collapse
|
6
|
Wei J, Liu R, Yang Z, Liu H, Wang Y, Zhang J, Sun M, Shen C, Liu J, Yu P, Tang NJ. Association of metals and bisphenols exposure with lipid profiles and dyslipidemia in Chinese adults: Independent, combined and interactive effects. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174315. [PMID: 38942316 DOI: 10.1016/j.scitotenv.2024.174315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/07/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Although studies have assessed the association of metals and bisphenols with lipid metabolism, the observed results have been controversial, and limited knowledge exists about the combined and interactive effects of metals and bisphenols exposure on lipid metabolism. METHODS Plasma metals and serum bisphenols concentrations were evaluated in 888 participants. Multiple linear regression and logistic regression models were conducted to assess individual associations of 18 metals and 3 bisphenols with 5 lipid profiles and dyslipidemia risk, respectively. The dose-response relationships of targeted contaminants with lipid profiles and dyslipidemia risk were captured by applying a restriction cubic spline (RCS) function. The bayesian kernel machine regression (BKMR) model was used to assess the overall effects of metals and bisphenols mixture on lipid profiles and dyslipidemia risk. The interactive effects of targeted contaminants on interested outcomes were explored by constructing an interaction model. RESULTS Single-contaminant analyses revealed that exposure to iron (Fe), nickel (Ni), copper (Cu), arsenic (As), selenium (Se), strontium (Sr), and tin (Sn) was associated with elevated lipid levels. Cobalt (Co) showed a negative association with high density lipoprotein cholesterol (HDL-C). Bisphenol A (BPA) and bisphenol AF (BPAF) were associated with decreased HDL-C levels, with nonlinear associations observed. Vanadium (V), lead (Pb), and silver (Ag) displayed U-shaped dose-response relationships with most lipid profiles. Multi-contaminant analyses indicated positive trends between contaminants mixture and total cholesterol (TC), triglycerides (TG), low density lipoprotein cholesterol (LDL-C), and non-high-density lipoprotein cholesterol (non-HDL-C). The interaction analyses showed that Se-Fe exhibited synergistic effects on LDL-C and non-HDL-C, and Se-Sn showed a synergistic effect on HDL-C. CONCLUSIONS Our study suggested that exposure to metals and bisphenols was associated with changes in lipid levels, and demonstrated their combined and interactive effects.
Collapse
Affiliation(s)
- Jiemin Wei
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Ruifang Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Ze Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Hongbo Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Yiqing Wang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Jingyun Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Meiqing Sun
- Wuqing District Center for Disease Control and Prevention, Tianjin 301700, China
| | - Changkun Shen
- Wuqing District Center for Disease Control and Prevention, Tianjin 301700, China
| | - Jian Liu
- Wuqing District Center for Disease Control and Prevention, Tianjin 301700, China
| | - Pei Yu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Nai-Jun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition, and Public Health, Tianjin Medical University, Tianjin 300070, China; Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China; Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
7
|
Li N, Wang G, Guo M, Zhu N, Yu W. The mechanism and clinical application of farnesyl diphosphate farnesyltransferase 1 in cancer metabolism. Biochem Biophys Res Commun 2024; 719:150046. [PMID: 38749088 DOI: 10.1016/j.bbrc.2024.150046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 06/05/2024]
Abstract
Cancer poses a significant risk to human well-being. Among the crucial characteristics of cancer is metabolic reprogramming. To meet the relentless metabolic needs, cancer cells enhance cholesterol metabolism within the adverse tumor microenvironment. Reprograming cholesterol metabolism includes a series of modifications in the synthesis, absorption, esterification, and metabolites associated with cholesterol. These adjustments have a strong correlation with the proliferation, invasion, metastasis, and other characteristics of malignant tumors. FDFT1, also known as farnesyl diphosphate farnesyltransferase 1, is an enzyme crucial in the process of cholesterol biosynthesis. Its significant involvement in tumor metabolism has garnered considerable interest. The significance of FDFT1 in cancer metabolism cannot be overstated, as it actively interacts with cancer cells. This paper aims to analyze and consolidate the mechanism of FDFT1 in cancer metabolism and explore its clinical application. The goal is to contribute new strategies and targets for the prevention and treatment of cancer metabolism.
Collapse
Affiliation(s)
- Nanxin Li
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Guojuan Wang
- Department of Oncology, Affiliated Hospital of Jiangxi University of Chinese Medicine, No.445, Bayi Avenue, Nanchang, 330006, China.
| | - Min Guo
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Naicheng Zhu
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Wenyan Yu
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| |
Collapse
|
8
|
Díaz-Grijuela E, Hernández A, Caballero C, Fernandez R, Urtasun R, Gulak M, Astigarraga E, Barajas M, Barreda-Gómez G. From Lipid Signatures to Cellular Responses: Unraveling the Complexity of Melanoma and Furthering Its Diagnosis and Treatment. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1204. [PMID: 39202486 PMCID: PMC11356604 DOI: 10.3390/medicina60081204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024]
Abstract
Recent advancements in mass spectrometry have significantly enhanced our understanding of complex lipid profiles, opening new avenues for oncological diagnostics. This review highlights the importance of lipidomics in the comprehension of certain metabolic pathways and its potential for the detection and characterization of various cancers, in particular melanoma. Through detailed case studies, we demonstrate how lipidomic analysis has led to significant breakthroughs in the identification and understanding of cancer types and its potential for detecting unique biomarkers that are instrumental in its diagnosis. Additionally, this review addresses the technical challenges and future perspectives of these methodologies, including their potential expansion and refinement for clinical applications. The discussion underscores the critical role of lipidomic profiling in advancing cancer diagnostics, proposing a new paradigm in how we approach this devastating disease, with particular emphasis on its application in comparative oncology.
Collapse
Affiliation(s)
| | | | | | - Roberto Fernandez
- IMG Pharma Biotech, Research and Development Division, 48170 Zamudio, Spain;
| | - Raquel Urtasun
- Biochemistry Area, Department of Health Science, Universidad Pública de Navarra, 31006 Pamplona, Spain; (R.U.); (M.B.)
| | | | - Egoitz Astigarraga
- Betternostics SL, 31110 Noáin, Spain; (E.D.-G.); (A.H.); (C.C.)
- IMG Pharma Biotech, Research and Development Division, 48170 Zamudio, Spain;
| | - Miguel Barajas
- Biochemistry Area, Department of Health Science, Universidad Pública de Navarra, 31006 Pamplona, Spain; (R.U.); (M.B.)
| | - Gabriel Barreda-Gómez
- Betternostics SL, 31110 Noáin, Spain; (E.D.-G.); (A.H.); (C.C.)
- IMG Pharma Biotech, Research and Development Division, 48170 Zamudio, Spain;
| |
Collapse
|
9
|
Erazo-Oliveras A, Muñoz-Vega M, Salinas ML, Wang X, Chapkin RS. Dysregulation of cellular membrane homeostasis as a crucial modulator of cancer risk. FEBS J 2024; 291:1299-1352. [PMID: 36282100 PMCID: PMC10126207 DOI: 10.1111/febs.16665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Cellular membranes serve as an epicentre combining extracellular and cytosolic components with membranous effectors, which together support numerous fundamental cellular signalling pathways that mediate biological responses. To execute their functions, membrane proteins, lipids and carbohydrates arrange, in a highly coordinated manner, into well-defined assemblies displaying diverse biological and biophysical characteristics that modulate several signalling events. The loss of membrane homeostasis can trigger oncogenic signalling. More recently, it has been documented that select membrane active dietaries (MADs) can reshape biological membranes and subsequently decrease cancer risk. In this review, we emphasize the significance of membrane domain structure, organization and their signalling functionalities as well as how loss of membrane homeostasis can steer aberrant signalling. Moreover, we describe in detail the complexities associated with the examination of these membrane domains and their association with cancer. Finally, we summarize the current literature on MADs and their effects on cellular membranes, including various mechanisms of dietary chemoprevention/interception and the functional links between nutritional bioactives, membrane homeostasis and cancer biology.
Collapse
Affiliation(s)
- Alfredo Erazo-Oliveras
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Mónica Muñoz-Vega
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Michael L. Salinas
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Xiaoli Wang
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
- Center for Environmental Health Research; Texas A&M University; College Station, Texas, 77843; USA
| |
Collapse
|
10
|
Fazliyeva R, Makhov P, Uzzo RG, Kolenko VM. Targeting NPC1 in Renal Cell Carcinoma. Cancers (Basel) 2024; 16:517. [PMID: 38339268 PMCID: PMC10854724 DOI: 10.3390/cancers16030517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/05/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Rapidly proliferating cancer cells have a greater requirement for cholesterol than normal cells. Tumor cells are largely dependent on exogenous lipids given that their growth requirements are not fully met by endogenous pathways. Our current study shows that ccRCC cells have redundant mechanisms of cholesterol acquisition. We demonstrate that all major lipoproteins (i.e., LDL, HDL, and VLDL) have a comparable ability to support the growth of ccRCC cells and are equally effective in counteracting the antitumor activities of TKIs. The intracellular trafficking of exogenous lipoprotein-derived cholesterol appears to be distinct from the movement of endogenously synthesized cholesterol. De novo synthetized cholesterol is transported from the endoplasmic reticulum directly to the plasma membrane and to the acyl-CoA: cholesterol acyltransferase, whereas lipoprotein-derived cholesterol is distributed through the NPC1-dependent endosomal trafficking system. Expression of NPC1 is increased in ccRCC at mRNA and protein levels, and high expression of NPC1 is associated with poor prognosis. Our current findings show that ccRCC cells are particularly sensitive to the inhibition of endolysosomal cholesterol export and underline the therapeutic potential of targeting NPC1 in ccRCC.
Collapse
Affiliation(s)
- Rushaniya Fazliyeva
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Peter Makhov
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Robert G. Uzzo
- Department of Urology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Vladimir M. Kolenko
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| |
Collapse
|
11
|
Wang W, Mai H, Xu H, Jing B, Yu C, Li X, Chen D, Huang Y, Shao M, Pan T. 4,8-Dicarboxyl-8,9-iridoid-1-glycoside inhibits apoptosis in human osteoarthritis chondrocytes via enhanced c-MYC-mediated cholesterol metabolism in vitro. Arthritis Res Ther 2023; 25:240. [PMID: 38082328 PMCID: PMC10712063 DOI: 10.1186/s13075-023-03217-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative disease related to cholesterol metabolism disorders. However, current therapies for OA are insufficient and no convincing disease-modifying OA drugs exist. Therefore, we aimed to elucidate the mechanism by which borojoa iridoid glycoside (BIG) inhibits chondrocyte apoptosis in OA. METHODS Borojoa pulp was heated to 70 °C, and the main active substance in borojoa, BIG, was extracted by fractionation at an ultraviolet 254-nm absorption peak. Chondrocytes were identified by immunohistochemistry and visualized by immunofluorescence confocal microscopy. The proliferation of chondrocytes cultured with BIG was determined by MTS assay. The apoptosis of chondrocytes cultured with BIG was tested by Annexin V-FITC/PI, and the cytokine, protein, and cholesterol levels in chondrocytes were detected by ELISA, RT‒qPCR, Western blot, and biochemistry analyses. Protein‒protein interactions were verified by a coimmunoprecipitation (Co-IP) assay. RESULTS BIG promoted chondrocyte proliferation and reduced apoptosis in vitro. BIG induced an alteration of the total RNA profiles in chondrocytes, and bioinformatic analysis showed that BIG inhibited chondrocyte apoptosis by promoting c-MYC expression; KEGG analysis confirmed that BIG-inhibited apoptosis was enriched in the cell cycle pathway. Flow cell cycle experiments confirmed that BIG promoted chondrocyte proliferation by significantly increasing the S phase cell number. The c-MYC inhibitor 10058-F4 stimulated the increased expression of IL-1β, IL-6, TNF-α, and AGEs and suppressed the cholesterol metabolism, which promoted chondrocyte apoptosis and autophagy. Co-IP analysis showed that BIG promoted the interaction of c-MYC and CH25H, Bcl-2, which suggests that BIG could inhibit chondrocyte apoptosis in part by enhancing c-MYC-mediated cholesterol metabolism. CONCLUSIONS This study confirmed that BIG promotes chondrocyte proliferation and inhibits apoptosis and autophagy, and BIG improving OA is associated with cholesterol metabolism. The results identify a potential mechanism by which BIG enhances c-MYC-mediated CH25H regulation of cholesterol metabolism in vitro and suggest that BIG might be a promising new drug against OA.
Collapse
Affiliation(s)
- WeiBing Wang
- Department of Anesthesiology, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China
| | - HaiMin Mai
- Department of Orthopedic, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510008, People's Republic of China
| | - Huang Xu
- Department of Anesthesiology, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China
| | - BaoSheng Jing
- Department of Orthopedics, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China
| | - CuiYu Yu
- Department of Anesthesiology, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China
| | - XiaoTing Li
- Department of Anesthesiology, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China
| | - DanGui Chen
- Department of Hematology, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China
| | - Yuan Huang
- Department of Science and Education, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China
| | - MeiMang Shao
- Department of Science and Education, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China
| | - Tao Pan
- Department of Orthopedic, Anqing Municipal Hospitals, Anhui Medical University, Anqing, 246000, People's Republic of China.
| |
Collapse
|
12
|
Torrens-Mas M, Collado-Solé A, Sola-Leyva A, Carrasco-Jiménez MP, Oliver J, Pons DG, Roca P, Sastre-Serra J. Mitochondrial Functionality Is Regulated by Alkylphospholipids in Human Colon Cancer Cells. BIOLOGY 2023; 12:1457. [PMID: 38132283 PMCID: PMC10740929 DOI: 10.3390/biology12121457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023]
Abstract
Alkylphospholipids (APLs) have been studied as anticancer drugs that interfere with biological membranes without targeting DNA. Although their mechanism of action is not fully elucidated yet, it is known that they disrupt the intracellular trafficking of cholesterol and its metabolism. Here, we analyzed whether APLs could also interfere with mitochondrial function. For this purpose, we used HT29 colorectal cancer cells, derived from a primary tumor, and SW620 colorectal cancer cells, derived from a metastasis site. After treatment with the APLs miltefosine and perifosine, we analyzed various mitochondrial parameters, including mitochondrial mass, cardiolipin content, mitochondrial membrane potential, H2O2 production, the levels of oxidative phosphorylation (OXPHOS) complexes, metabolic enzymes activity, the oxygen consumption rate, and the levels of apoptosis and autophagy markers. APLs, especially perifosine, increased mitochondrial mass while OXPHOS complexes levels were decreased without affecting the total oxygen consumption rate. Additionally, we observed an increase in pyruvate dehydrogenase (PDH) and isocitrate dehydrogenase (IDH) levels and a decrease in lactate dehydrogenase (LDH) activity, suggesting a metabolic rewiring induced by perifosine. These alterations led to higher mitochondrial membrane potential, which was potentiated by decreased uncoupling protein 2 (UCP2) levels and increased reactive oxygen species (ROS) production. Consequently, perifosine induced an imbalance in mitochondrial function, resulting in higher ROS production that ultimately impacted cellular viability.
Collapse
Affiliation(s)
- Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
| | - Alejandro Collado-Solé
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
| | - Alberto Sola-Leyva
- Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, University of Granada, Av. Fuentenueva s/n, 18001 Granada, Spain; (A.S.-L.); (M.P.C.-J.)
| | - María Paz Carrasco-Jiménez
- Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, University of Granada, Av. Fuentenueva s/n, 18001 Granada, Spain; (A.S.-L.); (M.P.C.-J.)
| | - Jordi Oliver
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Gabriel Pons
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Jorge Sastre-Serra
- Grupo Multidisciplinar de Oncología Traslacional, Research Institute of Health Sciences (IUNICS), University of Balearic Islands, 07122 Palma de Mallorca, Spain; (M.T.-M.); (J.O.); (D.G.P.); (J.S.-S.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
13
|
Howard E, Hurrell BP, Helou DG, Shafiei-Jahani P, Hasiakos S, Painter J, Srikanth S, Gwack Y, Akbari O. Orai inhibition modulates pulmonary ILC2 metabolism and alleviates airway hyperreactivity in murine and humanized models. Nat Commun 2023; 14:5989. [PMID: 37752127 PMCID: PMC10522697 DOI: 10.1038/s41467-023-41065-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Ca2+ entry via Ca2+ release-activated Ca2+ (CRAC) channels is a predominant mechanism of intracellular Ca2+ elevation in immune cells. Here we show the immunoregulatory role of CRAC channel components Orai1 and Orai2 in Group 2 innate lymphoid cells (ILC2s), that play crucial roles in the induction of type 2 inflammation. We find that blocking or genetic ablation of Orai1 and Orai2 downregulates ILC2 effector function and cytokine production, consequently ameliorating the development of ILC2-mediated airway inflammation in multiple murine models. Mechanistically, ILC2 metabolic and mitochondrial homeostasis are inhibited and lead to the upregulation of reactive oxygen species production. We confirm our findings in human ILC2s, as blocking Orai1 and Orai2 prevents the development of airway hyperreactivity in humanized mice. Our findings have a broad impact on the basic understanding of Ca2+ signaling in ILC2 biology, providing potential insights into the development of therapies for the treatment of allergic and atopic inflammatory diseases.
Collapse
Affiliation(s)
- Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Spyridon Hasiakos
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jacob Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
14
|
Ben Hassen C, Goupille C, Vigor C, Durand T, Guéraud F, Silvente-Poirot S, Poirot M, Frank PG. Is cholesterol a risk factor for breast cancer incidence and outcome? J Steroid Biochem Mol Biol 2023; 232:106346. [PMID: 37321513 DOI: 10.1016/j.jsbmb.2023.106346] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
Cholesterol plays important roles in many physiological processes, including cell membrane structure and function, hormone synthesis, and the regulation of cellular homeostasis. The role of cholesterol in breast cancer is complex, and some studies have suggested that elevated cholesterol levels may be associated with an increased risk of developing breast cancer, while others have found no significant association. On the other hand, other studies have shown that, for total cholesterol and plasma HDL-associated cholesterol levels, there was inverse association with breast cancer risk. One possible mechanism by which cholesterol may contribute to breast cancer risk is as a key precursor of estrogen. Other potential mechanisms by which cholesterol may contribute to breast cancer risk include its role in inflammation and oxidative stress, which have been linked to cancer progression. Cholesterol has also been shown to play a role in signaling pathways regulating the growth and proliferation of cancer cells. In addition, recent studies have shown that cholesterol metabolism can generate tumor promoters such as cholesteryl esters, oncosterone, 27-hydroxycholesterol but also tumor suppressor metabolites such as dendrogenin A. This review summarizes some of the most important clinical studies that have evaluated the role of cholesterol or its derivatives in breast cancer. It also addresses the role of cholesterol and its derivatives at the cellular level.
Collapse
Affiliation(s)
| | - Caroline Goupille
- INSERM N2C UMR1069, University of Tours, 37032 Tours, France; Department of Gynecology, CHRU Hôpital Bretonneau, boulevard Tonnellé, 37044 Tours, France
| | - Claire Vigor
- Institut des Biomolécules Max Mousseron, IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34293 CEDEX 5 Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, IBMM, Pôle Chimie Balard Recherche, Université de Montpellier, CNRS, ENSCM, 34293 CEDEX 5 Montpellier, France
| | - Françoise Guéraud
- INRAE, Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Sandrine Silvente-Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV:"Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France
| | - Marc Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV:"Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France; Equipe labellisée par la Ligue Nationale contre le Cancer, France
| | - Philippe G Frank
- INSERM N2C UMR1069, University of Tours, 37032 Tours, France; SGS Health and Nutrition, Saint Benoît, France.
| |
Collapse
|
15
|
Mohamed Abdoul-Latif F, Ainane A, Houmed Aboubaker I, Mohamed J, Ainane T. Exploring the Potent Anticancer Activity of Essential Oils and Their Bioactive Compounds: Mechanisms and Prospects for Future Cancer Therapy. Pharmaceuticals (Basel) 2023; 16:1086. [PMID: 37631000 PMCID: PMC10458506 DOI: 10.3390/ph16081086] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide, affecting millions of people each year. Fortunately, the last decades have been marked by considerable advances in the field of cancer therapy. Researchers have discovered many natural substances, some of which are isolated from plants that have promising anti-tumor activity. Among these, essential oils (EOs) and their constituents have been widely studied and shown potent anticancer activities, both in vitro and in vivo. However, despite the promising results, the precise mechanisms of action of EOs and their bioactive compounds are still poorly understood. Further research is needed to better understand these mechanisms, as well as their effectiveness and safety in use. Furthermore, the use of EOs as anticancer drugs is complex, as it requires absolute pharmacodynamic specificity and selectivity, as well as an appropriate formulation for effective administration. In this study, we present a synthesis of recent work on the mechanisms of anticancer action of EOs and their bioactive compounds, examining the results of various in vitro and in vivo studies. We also review future research prospects in this exciting field, as well as potential implications for the development of new cancer drugs.
Collapse
Affiliation(s)
- Fatouma Mohamed Abdoul-Latif
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti City P.O. Box 486, Djibouti;
| | - Ayoub Ainane
- Superior School of Technology of Khenifra, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco; (A.A.); (T.A.)
| | | | - Jalludin Mohamed
- Medicinal Research Institute, Center for Studies and Research of Djibouti, IRM-CERD, Route de l’Aéroport, Haramous, Djibouti City P.O. Box 486, Djibouti;
| | - Tarik Ainane
- Superior School of Technology of Khenifra, University of Sultan Moulay Slimane, P.O. Box 170, Khenifra 54000, Morocco; (A.A.); (T.A.)
| |
Collapse
|
16
|
Wu CC, Chen WC, Hsiao WP, Huang KF, Liao YS, Lin HB, Wu YJ, Kao CH, Chen SL. Reciprocal Regulation of Peroxisome Biogenesis and Myogenic Factors Is Critical for Myogenesis. Int J Mol Sci 2023; 24:12262. [PMID: 37569637 PMCID: PMC10419124 DOI: 10.3390/ijms241512262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Mitochondria (MITO) and peroxisomes (PEXO) are the major organelles involved in the oxidative metabolism of cells, but detailed examination of their dynamics and functional adaptations during skeletal muscle (SKM) development (myogenesis) is still lacking. In this study, we found that during myogenesis, MITO DNA, ROS level, and redox ratio increased in myotubes, but the membrane potential (Δψm) and ATP content reduced, implying that the MITO efficiency might reduce during myogenesis. The PEXO number and density both increased during myogenesis, which probably resulted from the accumulation and increased biogenesis of PEXO. The expression of PEXO biogenesis factors was induced during myogenesis in vitro and in utero, and their promoters were also activated by MyoD. Knockdown of the biogenesis factors Pex3 repressed not only the PEXO density and functions but also the levels of MITO genes and functions, suggesting a close coupling between PEXO biogenesis and MITO functions. Surprisingly, Pex3 knockdown by the CRISPRi system repressed myogenic differentiation, indicating critical involvement of PEXO biogenesis in myogenesis. Taken together, these observations suggest that the dynamics and functions of both MITO and PEXO are coupled with each other and with the metabolic changes that occur during myogenesis, and these metabolic couplings are critical to myogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Shen-Liang Chen
- Department of Life Sciences, College of Health Sciences and Technology, National Central University, Taoyuan 320317, Taiwan; (C.-C.W.); (W.-C.C.); (W.-P.H.); (K.-F.H.); (Y.-S.L.); (H.-B.L.); (Y.-J.W.); (C.-H.K.)
| |
Collapse
|
17
|
Pan GQ, Jiao Y, Meng GX, Dong ZR, Li T. The relationship between the serum lipid profile and hepatocellular carcinoma in east Asian population: A mendelian randomization study. Heliyon 2023; 9:e17126. [PMID: 37484252 PMCID: PMC10361312 DOI: 10.1016/j.heliyon.2023.e17126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Although several studies have found that the serum lipid profile may be correlated with hepatocellular carcinoma (HCC), the causal relationships between the serum lipid profile and HCC have not been determined due to potential confounder. Here, Mendelian randomization (MR) analysis was performed to identify the relationship between the serum lipid profile and HCC in the East Asian population. METHOD Our study made a MR analysis with the validation of two data sets. We obtained genome-wide association study (GWAS) data related to the serum lipid profile from Asian Genetic Epidemiology Network (AGEN). Then, the data from a recent large GWAS of the East Asian ancestry in Japan (BioBank Japan, BBJ) were extracted. Summary-level statistical data for HCC were obtained from a large GWAS of the East Asian ancestry in Japan. Univariable MR analysis were performed to identify whether the genetic evidence of serum lipid profile was significantly associated with HCC risk. Multivariable MR analysis was conducted to estimate the independent effects of exposures on HCC. RESULTS Univariable and multivariable MR analyses indicated that the serum lipid profile was not a risk factor for HCC incidence in either data set based on the East Asian population. Multivariable MR analysis revealed that the hazard ratios of the probability of HCC in AGEN were 1.134 (95% confidence interval (CI), 0.903-1.424) for TG, 1.010 (95% CI: 0.824-1.237) for HDL-C, 0.974 (95% CI: 0.746-1.271) for TC, 0.918 (95% CI: 0.734-1.147) for LDL-C, while the results in BBJ were also non-significant: 1.111 (95% CI: 0.869-1.419) for TG, 0.957 (95% CI: 0.790-1.158) for HDL-C, 0.917 (95% CI: 0.643-1.308) for TC, 0.932 (95% CI: 0.699-1.243) for LDL-C. CONCLUSION Our MR study with the validation of two data sets found no strong evidence to support causal associations between the serum lipid profile and HCC risk.
Collapse
Affiliation(s)
- Guo-Qiang Pan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Guang-Xiao Meng
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Zhao-Ru Dong
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, 250012, China
| |
Collapse
|
18
|
Li J, Yang H, Zhang L, Zhang S, Dai Y. Metabolic reprogramming and interventions in endometrial carcinoma. Biomed Pharmacother 2023; 161:114526. [PMID: 36933381 DOI: 10.1016/j.biopha.2023.114526] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Cancer cells are usually featured by metabolic adaptations that facilitate their growth, invasion, and metastasis. Thus, reprogramming of intracellular energy metabolism is currently one of the hotspots in the field of cancer research. Whereas aerobic glycolysis (known as the Warburg effect) has long been considered a dominant form of energy metabolism in cancer cells, emerging evidence indicates that other metabolic forms, especially oxidative phosphorylation (OXPHOS), may play a critical role at least in some types of cancer. Of note, women with metabolic syndromes (MetS), including obesity, hyperglycemia, dyslipidemia, and hypertension, have an increased risk of developing endometrial carcinoma (EC), suggesting a close link between metabolism and EC. Interestingly, the metabolic preferences vary among EC cell types, particularly cancer stem cells and chemotherapy-resistant cells. Currently, it is commonly accepted that glycolysis is the main energy provider in EC cells, while OXPHOS is reduced or impaired. Moreover, agents specifically targeting the glycolysis and/or OXPHOS pathways can inhibit tumor cell growth and promote chemosensitization. For example, metformin and weight control not only reduce the incidence of EC but also improve the prognosis of EC patients. In this review, we comprehensively overview the current in-depth understanding of the relationship between metabolism and EC and provide up-to-date insights into the development of novel therapies targeting energy metabolism for auxiliary treatment in combination with chemotherapy for EC, especially those resistant to conventional chemotherapy.
Collapse
Affiliation(s)
- Jiajia Li
- The Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin 130061, China; Department of Gynecologic Oncology, Gynecology and Obstetrics Center, the First Hospital of Jilin University, Changchun, Jilin 130012, China
| | - Hongmei Yang
- Department of Critical Care Medicine, the First Hospital of Jilin University, Changchun, Jilin 130012, China
| | - Lingyi Zhang
- Department of Gynecology and Obstetrics, the Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Songling Zhang
- Department of Gynecologic Oncology, Gynecology and Obstetrics Center, the First Hospital of Jilin University, Changchun, Jilin 130012, China.
| | - Yun Dai
- The Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin 130061, China.
| |
Collapse
|
19
|
Nóbrega-Pereira S, Santos F, Oliveira Santos M, Serafim TL, Lopes AP, Coutinho D, Carvalho FS, Domingues RM, Domingues P, Bernardes de Jesus B, Morais VA, Dias S. Mitochondrial Metabolism Drives Low-density Lipoprotein-induced Breast Cancer Cell Migration. CANCER RESEARCH COMMUNICATIONS 2023; 3:709-724. [PMID: 37377750 PMCID: PMC10132314 DOI: 10.1158/2767-9764.crc-22-0394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 06/29/2023]
Abstract
Most cancer-related deaths are due to metastases. Systemic factors, such as lipid-enriched environments [as low-density lipoprotein (LDL)-cholesterol], favor breast cancer, including triple-negative breast cancer (TNBC) metastasis formation. Mitochondria metabolism impacts TNBC invasive behavior but its involvement in a lipid-enriched setting is undisclosed. Here we show that LDL increases lipid droplets, induces CD36 and augments TNBC cells migration and invasion in vivo and in vitro. LDL induces higher mitochondrial mass and network spread in migrating cells, in an actin remodeling-dependent manner, and transcriptomic and energetic analyses revealed that LDL renders TNBC cells dependent on fatty acids (FA) usage for mitochondrial respiration. Indeed, engagement on FA transport into the mitochondria is required for LDL-induced migration and mitochondrial remodeling. Mechanistically, LDL treatment leads to mitochondrial long-chain fatty acid accumulation and increased reactive oxygen species (ROS) production. Importantly, CD36 or ROS blockade abolished LDL-induced cell migration and mitochondria metabolic adaptations. Our data suggest that LDL induces TNBC cells migration by reprogramming mitochondrial metabolism, revealing a new vulnerability in metastatic breast cancer. Significance LDL induces breast cancer cell migration that relies on CD36 for mitochondrial metabolism and network remodeling, providing an antimetastatic metabolic strategy.
Collapse
Affiliation(s)
- Sandrina Nóbrega-Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Instituto de Biomedicina (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Francisco Santos
- Instituto de Biomedicina (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Miguel Oliveira Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Teresa L. Serafim
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Ana Patrícia Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Diogo Coutinho
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Filipa S. Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Rosário M. Domingues
- Mass Spectrometry Center, QOPNA, University of Aveiro, Aveiro, Portugal
- Department of Chemistry and CESAM&ECOMARE, University of Aveiro, Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Center, QOPNA, University of Aveiro, Aveiro, Portugal
| | - Bruno Bernardes de Jesus
- Instituto de Biomedicina (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Vanessa A. Morais
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Sérgio Dias
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
20
|
Markowski A, Zaremba-Czogalla M, Jaromin A, Olczak E, Zygmunt A, Etezadi H, Boyd BJ, Gubernator J. Novel Liposomal Formulation of Baicalein for the Treatment of Pancreatic Ductal Adenocarcinoma: Design, Characterization, and Evaluation. Pharmaceutics 2023; 15:pharmaceutics15010179. [PMID: 36678808 PMCID: PMC9865389 DOI: 10.3390/pharmaceutics15010179] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/20/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers so there is an urgent need to develop new drugs and therapies to treat it. Liposome-based formulations of naturally-derived bioactive compounds are promising anticancer candidates due to their potential for passive accumulation in tumor tissues, protection against payload degradation, and prevention of non-specific toxicity. We chose the naturally-derived flavonoid baicalein (BAI) due to its promising effect against pancreatic ductal adenocarcinoma (PDAC) and encapsulated it into a liposomal bilayer using the passive loading method, with an almost 90% efficiency. We performed a morphological and stability analysis of the obtained BAI liposomal formulation and evaluated its activity on two-dimensional and three-dimensional pancreatic cell models. As the result, we obtained a stable BAI-encapsulated liposomal suspension with a size of 100.9 nm ± 2.7 and homogeneity PDI = 0.124 ± 0.02, suitable for intravenous administration. Furthermore, this formulation showed high cytotoxic activity towards AsPC-1 and BxPC-3 PDAC cell lines (IC50 values ranging from 21 ± 3.6 µM to 27.6 ± 4.1 µM), with limited toxicity towards normal NHDF cells and a lack of hemolytic activity. Based on these results, this new BAI liposomal formulation is an excellent candidate for potential anti-PDAC therapy.
Collapse
Affiliation(s)
- Adam Markowski
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
- Correspondence: (A.M.); (A.J.)
| | - Magdalena Zaremba-Czogalla
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
- Correspondence: (A.M.); (A.J.)
| | - Ewa Olczak
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Adrianna Zygmunt
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Haniyeh Etezadi
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Ben J. Boyd
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| |
Collapse
|
21
|
Essaouiba A, Jellali R, Gilard F, Gakière B, Okitsu T, Legallais C, Sakai Y, Leclerc E. Investigation of the Exometabolomic Profiles of Rat Islets of Langerhans Cultured in Microfluidic Biochip. Metabolites 2022; 12:metabo12121270. [PMID: 36557308 PMCID: PMC9786643 DOI: 10.3390/metabo12121270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Diabetes mellitus (DM) is a complex disease with high prevalence of comorbidity and mortality. DM is predicted to reach more than 700 million people by 2045. In recent years, several advanced in vitro models and analytical tools were developed to investigate the pancreatic tissue response to pathological situations and identify therapeutic solutions. Of all the in vitro promising models, cell culture in microfluidic biochip allows the reproduction of in-vivo-like micro-environments. Here, we cultured rat islets of Langerhans using dynamic cultures in microfluidic biochips. The dynamic cultures were compared to static islets cultures in Petri. The islets' exometabolomic signatures, with and without GLP1 and isradipine treatments, were characterized by GC-MS. Compared to Petri, biochip culture contributes to maintaining high secretions of insulin, C-peptide and glucagon. The exometabolomic profiling revealed 22 and 18 metabolites differentially expressed between Petri and biochip on Day 3 and 5. These metabolites illustrated the increase in lipid metabolism, the perturbation of the pentose phosphate pathway and the TCA cycle in biochip. After drug stimulations, the exometabolome of biochip culture appeared more perturbed than the Petri exometabolome. The GLP1 contributed to the increase in the levels of glycolysis, pentose phosphate and glutathione pathways intermediates, whereas isradipine led to reduced levels of lipids and carbohydrates.
Collapse
Affiliation(s)
- Amal Essaouiba
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Centre de Recherche Royallieu CS 60319, 60203 Compiègne, France
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Rachid Jellali
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Centre de Recherche Royallieu CS 60319, 60203 Compiègne, France
- Correspondence: (R.J.); (E.L.)
| | - Françoise Gilard
- Plateforme Métabolisme-Métabolome, Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université Evry, Université Paris Cité, Bâtiment 360, Avenue des Sciences, 91190 Gif sur Yvette, France
| | - Bertrand Gakière
- Plateforme Métabolisme-Métabolome, Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université Evry, Université Paris Cité, Bâtiment 360, Avenue des Sciences, 91190 Gif sur Yvette, France
| | - Teru Okitsu
- Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Cécile Legallais
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Centre de Recherche Royallieu CS 60319, 60203 Compiègne, France
| | - Yasuyuki Sakai
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Department of Chemical Engineering, Faculty of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Eric Leclerc
- Biomechanics and Bioengineering, CNRS, Université de Technologie de Compiègne, Centre de Recherche Royallieu CS 60319, 60203 Compiègne, France
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Correspondence: (R.J.); (E.L.)
| |
Collapse
|
22
|
Lazniewska J, Bader C, Hickey SM, Selemidis S, O'Leary J, Simpson PV, Stagni S, Plush SE, Massi M, Brooks D. Rhenium(I) conjugates as tools for tracking cholesterol in cells. Metallomics 2022; 14:mfac040. [PMID: 35657681 PMCID: PMC9344854 DOI: 10.1093/mtomcs/mfac040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/17/2022] [Indexed: 11/15/2022]
Abstract
Cholesterol is vital to control membrane integrity and fluidity, but is also a precursor to produce steroid hormones, bile acids, and vitamin D. Consequently, altered cholesterol biology has been linked to many diseases, including metabolic syndromes and cancer. Defining the intracellular pools of cholesterol and its trafficking within cells is essential to understand both normal cell physiology and mechanisms of pathogenesis. We have synthesized a new cholesterol mimic (ReTEGCholestanol), comprising a luminescent rhenium metal complex and a cholestanol targeting unit, linked using a tetraethylene glycol (TEG) spacer. ReTEGCholestanol demonstrated favourable imaging properties and improved water solubility when compared to a cholesterol derivative, and structurally related probes lacking the TEG linker. A non-malignant and three malignant prostate cell lines were used to characterize the uptake and intracellular distribution of ReTEGCholestanol. The ReTEGCholestanol complex was effectively internalized and mainly localized to late endosomes/lysosomes in non-malignant PNT1a cells, while in prostate cancer cells it also accumulated in early endosomes and multivesicular bodies, suggesting disturbed cholesterol biology in the malignant cells. The ReTEGCholestanol is a novel imaging agent for visualizing endosomal uptake and trafficking, which may be used to define cholesterol related biology including membrane integration and altered lipid trafficking/processing.
Collapse
Affiliation(s)
- Joanna Lazniewska
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Christie Bader
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Shane M Hickey
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Stavros Selemidis
- Department of Human Biosciences, RMIT University, Melbourne, Victoria 3000, Australia
| | - John O'Leary
- Discipline of Histopathology, University of Dublin Trinity College, Dublin 2, Ireland
| | - Peter V Simpson
- School of Molecular and Life Sciences - Curtin University, Bentley, Western Australia 6102, Australia
| | - Stefano Stagni
- Department of Industrial Chemistry Toso Montanari, University of Bologna, Via Zamboni, 33, Bologna I-40136, Italy
| | - Sally E Plush
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Massimiliano Massi
- School of Molecular and Life Sciences - Curtin University, Bentley, Western Australia 6102, Australia
| | - Doug Brooks
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
- Discipline of Histopathology, University of Dublin Trinity College, Dublin 2, Ireland
- School of Molecular and Life Sciences - Curtin University, Bentley, Western Australia 6102, Australia
| |
Collapse
|
23
|
Gu YR, Kim J, Na JC, Han WK. Mitochondrial metabolic reprogramming by SIRT3 regulation ameliorates drug resistance in renal cell carcinoma. PLoS One 2022; 17:e0269432. [PMID: 35671305 PMCID: PMC9173632 DOI: 10.1371/journal.pone.0269432] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/21/2022] [Indexed: 11/18/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) alters metabolic signals frequently, leading to mitochondrial dysfunction, such as increase of glycolysis and accumulation of lipid. Sirtuin3 (SIRT3) is a key factor for the regulation of both mitochondrial integrity and function. SIRT3 is downregulated and contributes in both cancer development and progression in ccRCC. The aim of this study is to investigate SIRT3-regulated mitochondrial biogenesis in ccRCC. SIRT3 overexpression alone reduced glucose uptake rate and enhanced membrane potential in mitochondria. ccRCC with overexpressed SIRT3 further improved the lethal effects when combined with anticancer drugs (Resveratrol, Everolimus and Temsirolimus). Cell viability was markedly decreased in a dose-dependent manner when treated with resveratrol or mTOR inhibitors in SIRT3 overexpressing ccRCC. In conclusion, SIRT3 improved mitochondrial functions in ccRCC through metabolic reprogramming. Mitochondrial reprogramming by SIRT3 regulation improves the sensitivity to anticancer drugs. The combination of SIRT3 and resveratrol functioned synergistically lethal effect in ccRCC.
Collapse
Affiliation(s)
- Young-Ran Gu
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jinu Kim
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Center of Uro-Oncology, Yonsei Cancer Hospital, Seoul, Korea
| | - Joon Chae Na
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Woong Kyu Han
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Center of Uro-Oncology, Yonsei Cancer Hospital, Seoul, Korea
- * E-mail:
| |
Collapse
|
24
|
Li L, Liu Y, Liu X, Zheng N, Gu Y, Song Y, Wang X. Regulatory roles of external cholesterol in human airway epithelial mitochondrial function through STARD3 signalling. Clin Transl Med 2022; 12:e902. [PMID: 35678098 PMCID: PMC9178408 DOI: 10.1002/ctm2.902] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Hypercholesterolemia is found in patients with chronic lung inflammation, during which airway epithelial cells play important roles in maintenance of inflammatory responses to pathogens. The present study aims at molecular mechanisms by which cholesterol changes airway epithelial sensitivity in response to smoking. METHODS Human bronchial epithelial cells (HBEs) were stimulated with cigarette smoke extract (CSE) and mice were exposed to CS/lipopolysaccharide (LPS) as models in vitro and in vivo. Severe COPD patients and healthy volunteers were also enrolled and the level of cholesterol in plasma was detected by metabolomics. Filipin III and elisa kits were used to stain free cholesterol. Mitochondrial function was detected by mitotracker green, mitotracker green, and Seahorse. Mitochondrial morphology was detected by high content screening and electron microscopy. The mRNA and protein levels of mitochondrial dynamics-related proteins were detected by RT-qPCR and Western blot,respectively. BODIPY 493/503 was used to stain lipid droplets. Lipidomics was used to detect intracellular lipid components. The mRNA level of interleukin (IL)-6 and IL-8 were detected by RT-qPCR. RESULTS We found that the cholesterol overload was associated with chronic obstructive pulmonary disease (COPD) and airway epithelia-driven inflammation, evidenced by hypercholesterolemia in patients with COPD and preclinical models, alteration of lipid metabolism-associated genes in CSE-induced airway epithelia and production of ILs. External cholesterol altered airway epithelial sensitivity of inflammation in response to CSE, through the regulation of STARD3-MFN2 pathway, cholesterol re-distribution, altered transport and accumulation of cholesterol, activities of lipid transport regulators and disorder of mitochondrial function and dynamics. MFN2 down-regulation increased airway epithelial sensitivity and production of ILs after smoking, at least partially by injuring fatty acid oxidation and activating mTOR phosphorylation. CONCLUSIONS Our data provide new insights for understanding molecular mechanisms of cholesterol-altered airway epithelial inflammation and for developing diagnostic biomarkers and therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Liyang Li
- Department of Pulmonary and Critical Care Medicine, Zhongshan HospitalFudan University Shanghai Medical CollegeShanghaiChina
| | - Yifei Liu
- Center of Molecular Diagnosis and TherapyThe Second Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Xuanqi Liu
- Shanghai Institute of Clinical BioinformaticsShanghaiChina
| | - Nannan Zheng
- Department of Pulmonary and Critical Care Medicine, Zhongshan HospitalFudan University Shanghai Medical CollegeShanghaiChina
| | - Yutong Gu
- Department of Pulmonary and Critical Care Medicine, Zhongshan HospitalFudan University Shanghai Medical CollegeShanghaiChina
| | - Yuanlin Song
- Department of Pulmonary and Critical Care Medicine, Zhongshan HospitalFudan University Shanghai Medical CollegeShanghaiChina
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan HospitalFudan University Shanghai Medical CollegeShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghaiChina
- Shanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesShanghaiChina
| |
Collapse
|
25
|
Wang G, Lu Y, Di S, Xie M, Jing F, Dai X. miR‑99a‑5p inhibits glycolysis and induces cell apoptosis in cervical cancer by targeting RRAGD. Oncol Lett 2022; 24:228. [PMID: 35720506 PMCID: PMC9185141 DOI: 10.3892/ol.2022.13349] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/03/2022] [Indexed: 11/06/2022] Open
Abstract
Cervical cancer (CC) is one of the most common gynecological malignancies that endangers women's health. A negative effect of glycolysis is that it contributes to abnormal tumor growth. MicroRNA (miR)-99a expression has been found to be decreased in CC. The present study aimed to investigate the role of miR-99a-5p in glycolysis in CC. For this purpose, the association between miR-99a and the prognosis of patients with CC from The Cancer Genome Atlas database was analyzed using Kaplan-Meier analysis. miR-99a-5p expression and Ras-related GTP binding D (RRAGD) expression were assessed using reverse transcription-quantitative PCR and western blot analysis. Cell proliferation and apoptosis were examined using an MTT assay and flow cytometry, respectively. Glucose uptake, lactate concentration and extracellular acidification rate were measured using a glucose uptake colorimetric assay, a lactate colorimetric assay and a Seahorse XFe96 extracellular flux analyzer, respectively. The association between miR-99a-5p and RRAGD was predicted using TargetScan 7.1, and was confirmed using dual luciferase reporter assay. The results revealed that miR-99a-5p expression was decreased and that of RRAGD was increased in CC tissues and cell lines. RRAGD was negatively regulated by miR-99a-5p. The overexpression of miR-99a-5p induced apoptosis and inhibited glycolysis in CC cells by targeting RRAGD. On the whole, the present study revealed a novel mechanism through which miR-99a-5p regulates cell apoptosis and glycolysis in CC, thus providing a potential therapeutic target for CC.
Collapse
Affiliation(s)
- Gang Wang
- Department of Gynecology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei 430061, P.R. China
| | - Yu Lu
- Department of Vasculocardiology, Wuhan Wuchang Hospital, Wuhan, Hubei 430061, P.R. China
| | - Shi Di
- Department of Gynecology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei 430061, P.R. China
| | - Maohua Xie
- Department of Obstetrics, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei 430061, P.R. China
| | - Fang Jing
- Department of Gynecology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei 430061, P.R. China
| | - Xiaoyan Dai
- Department of Gynecology, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei 430061, P.R. China
| |
Collapse
|
26
|
Badr H, Blutrich R, Chan K, Tong J, Taylor P, Zhang W, Kafri R, Röst HL, Tsao MS, Moran MF. Proteomic characterization of a candidate polygenic driver of metabolism in non-small cell lung cancer. J Mol Biol 2022; 434:167636. [PMID: 35595168 DOI: 10.1016/j.jmb.2022.167636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/03/2022] [Accepted: 05/08/2022] [Indexed: 11/18/2022]
Abstract
Proteome analysis revealed signatures of co-expressed upregulated metabolism proteins highly conserved between primary and non-small cell lung cancer (NSCLC) patient-derived xenograft tumors (Li et al. 2014, Nat. Communications 5:5469). The C10 signature is encoded by seven genes (ADSS, ATP2A2, CTPS1, IMPDH2, PKM2, PTGES3, SGPL1) and DNA alterations in C10-encoding genes are associated with longer survival in a subset of NSCLC. To explore the C10 signature as an oncogenic driver and address potential mechanisms of action, C10 protein expression and protein-protein interactions were determined. In independent NSCLC cohorts, the coordinated expression of C10 proteins was significant and mutations in C10 genes were associated with better outcome. Affinity purification-mass spectrometry and in vivo proximity-based biotin identification defined a C10 interactome involving 667 proteins including candidate drug targets and clusters associated with glycolysis, calcium homeostasis, and nucleotide and sphingolipid metabolism. DNA alterations in genes encoding C10 interactome components were also found to be associated with better survival. These data support the notion that the coordinated upregulation of the C10 signature impinges metabolic processes that collectively function as an oncogenic driver in NSCLC.
Collapse
Affiliation(s)
- Heba Badr
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Ron Blutrich
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Kaitlin Chan
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Jiefei Tong
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Paul Taylor
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; SPARC BioCentre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Wen Zhang
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Ran Kafri
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Hannes L Röst
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; Departments of Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Michael F Moran
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada; SPARC BioCentre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada.
| |
Collapse
|
27
|
Sallustio V, Chiocchio I, Mandrone M, Cirrincione M, Protti M, Farruggia G, Abruzzo A, Luppi B, Bigucci F, Mercolini L, Poli F, Cerchiara T. Extraction, Encapsulation into Lipid Vesicular Systems, and Biological Activity of Rosa canina L. Bioactive Compounds for Dermocosmetic Use. Molecules 2022; 27:molecules27093025. [PMID: 35566374 PMCID: PMC9104920 DOI: 10.3390/molecules27093025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/01/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022] Open
Abstract
Valorization of wild plants to obtain botanical ingredients could be a strategy for sustainable production of cosmetics. This study aimed to select the rosehip extract containing the greatest amounts of bioactive compounds and to encapsulate it in vesicular systems capable of protecting their own antioxidant activity. Chemical analysis of Rosa canina L. extracts was performed by LC-DAD-MS/MS and 1H-NMR and vitamins, phenolic compounds, sugars, and organic acids were detected as the main compounds of the extracts. Liposomes, prepared by the film hydration method, together with hyalurosomes and ethosomes, obtained by the ethanol injection method, were characterized in terms of vesicle size, polydispersity index, entrapment efficiency, zeta potential, in vitro release and biocompatibility on WS1 fibroblasts. Among all types of vesicular systems, ethosomes proved to be the most promising nanocarriers showing nanometric size (196 ± 1 nm), narrow polydispersity (0.20 ± 0.02), good entrapment efficiency (92.30 ± 0.02%), and negative zeta potential (−37.36 ± 0.55 mV). Moreover, ethosomes showed good stability over time, a slow release of polyphenols compared with free extract, and they were not cytotoxic. In conclusion, ethosomes could be innovative carriers for the encapsulation of rosehip extract.
Collapse
Affiliation(s)
- Valentina Sallustio
- Drug Delivery Research Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (V.S.); (A.A.); (B.L.); (F.B.)
| | - Ilaria Chiocchio
- Pharmaceutical Botany Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio 42, 40127 Bologna, Italy; (I.C.); (M.M.); (F.P.)
| | - Manuela Mandrone
- Pharmaceutical Botany Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio 42, 40127 Bologna, Italy; (I.C.); (M.M.); (F.P.)
| | - Marco Cirrincione
- Pharmaco-Toxicological Analysis (PTA Lab.), Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (M.C.); (M.P.); (L.M.)
| | - Michele Protti
- Pharmaco-Toxicological Analysis (PTA Lab.), Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (M.C.); (M.P.); (L.M.)
| | - Giovanna Farruggia
- Pharmaceutical Biochemistry Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy;
| | - Angela Abruzzo
- Drug Delivery Research Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (V.S.); (A.A.); (B.L.); (F.B.)
| | - Barbara Luppi
- Drug Delivery Research Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (V.S.); (A.A.); (B.L.); (F.B.)
| | - Federica Bigucci
- Drug Delivery Research Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (V.S.); (A.A.); (B.L.); (F.B.)
| | - Laura Mercolini
- Pharmaco-Toxicological Analysis (PTA Lab.), Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (M.C.); (M.P.); (L.M.)
| | - Ferruccio Poli
- Pharmaceutical Botany Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio 42, 40127 Bologna, Italy; (I.C.); (M.M.); (F.P.)
| | - Teresa Cerchiara
- Drug Delivery Research Lab., Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy; (V.S.); (A.A.); (B.L.); (F.B.)
- Correspondence: ; Tel.: +39-0512095615
| |
Collapse
|
28
|
Torres S, Segalés P, García-Ruiz C, Fernández-Checa JC. Mitochondria and the NLRP3 Inflammasome in Alcoholic and Nonalcoholic Steatohepatitis. Cells 2022; 11:1475. [PMID: 35563780 PMCID: PMC9105698 DOI: 10.3390/cells11091475] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Alcoholic (ASH) and nonalcoholic steatohepatitis (NASH) are advanced stages of fatty liver disease and two of the most prevalent forms of chronic liver disease. ASH and NASH are associated with significant risk of further progression to cirrhosis and hepatocellular carcinoma (HCC), the most common type of liver cancer, and a major cause of cancer-related mortality. Despite extensive research and progress in the last decades to elucidate the mechanisms of the development of ASH and NASH, the pathogenesis of both diseases is still poorly understood. Mitochondrial damage and activation of inflammasome complexes have a role in inducing and sustaining liver damage. Mitochondrial dysfunction produces inflammatory factors that activate the inflammasome complexes. NLRP3 inflammasome (nucleotide-binding oligomerization domain-like receptor protein 3) is a multiprotein complex that activates caspase 1 and the release of pro-inflammatory cytokines, including interleukin-1β (IL-1β) and interleukin-18 (IL-18), and contributes to inflammatory pyroptotic cell death. The present review, which is part of the issue "Mitochondria in Liver Pathobiology", provides an overview of the role of mitochondrial dysfunction and NLRP3 activation in ASH and NASH.
Collapse
Affiliation(s)
- Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (S.T.); (P.S.)
- Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, 28029 Madrid, Spain
| | - Paula Segalés
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (S.T.); (P.S.)
- Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, 28029 Madrid, Spain
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (S.T.); (P.S.)
- Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, 28029 Madrid, Spain
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - José C. Fernández-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (S.T.); (P.S.)
- Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, 28029 Madrid, Spain
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
29
|
Bhangu SK, Fernandes S, Beretta GL, Tinelli S, Cassani M, Radziwon A, Wojnilowicz M, Sarpaki S, Pilatis I, Zaffaroni N, Forte G, Caruso F, Ashokkumar M, Cavalieri F. Transforming the Chemical Structure and Bio-Nano Activity of Doxorubicin by Ultrasound for Selective Killing of Cancer Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107964. [PMID: 35100658 DOI: 10.1002/adma.202107964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/24/2022] [Indexed: 06/14/2023]
Abstract
Reconfiguring the structure and selectivity of existing chemotherapeutics represents an opportunity for developing novel tumor-selective drugs. Here, as a proof-of-concept, the use of high-frequency sound waves is demonstrated to transform the nonselective anthracycline doxorubicin into a tumor selective drug molecule. The transformed drug self-aggregates in water to form ≈200 nm nanodrugs without requiring organic solvents, chemical agents, or surfactants. The nanodrugs preferentially interact with lipid rafts in the mitochondria of cancer cells. The mitochondrial localization of the nanodrugs plays a key role in inducing reactive oxygen species mediated selective death of breast cancer, colorectal carcinoma, ovarian carcinoma, and drug-resistant cell lines. Only marginal cytotoxicity (80-100% cell viability) toward fibroblasts and cardiomyocytes is observed, even after administration of high doses of the nanodrug (25-40 µg mL-1 ). Penetration, cytotoxicity, and selectivity of the nanodrugs in tumor-mimicking tissues are validated by using a 3D coculture of cancer and healthy cells and 3D cell-collagen constructs in a perfusion bioreactor. The nanodrugs exhibit tropism for lung and limited accumulation in the liver and spleen, as suggested by in vivo biodistribution studies. The results highlight the potential of this approach to transform the structure and bioactivity of anticancer drugs and antibiotics bearing sono-active moieties.
Collapse
Affiliation(s)
- Sukhvir Kaur Bhangu
- School of Science, RMIT University, Melbourne, Victoria, 3000, Australia
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
- School of Chemistry, The University of Melbourne, Victoria, 3010, Australia
| | - Soraia Fernandes
- International Clinical Research Center (ICRC), St Anne's University Hospital, Brno, 65691, Czechia
| | - Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, Milan, 20133, Italy
| | - Stella Tinelli
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, Milan, 20133, Italy
| | - Marco Cassani
- International Clinical Research Center (ICRC), St Anne's University Hospital, Brno, 65691, Czechia
| | - Agata Radziwon
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Marcin Wojnilowicz
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Sophia Sarpaki
- BIOEMTECH, 27 Neapoleos st., Lefkippos Attica Technology Park - N.C.S.R. Demokritos, Athens, 15341, Greece
| | - Irinaios Pilatis
- BIOEMTECH, 27 Neapoleos st., Lefkippos Attica Technology Park - N.C.S.R. Demokritos, Athens, 15341, Greece
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, Milan, 20133, Italy
| | - Giancarlo Forte
- International Clinical Research Center (ICRC), St Anne's University Hospital, Brno, 65691, Czechia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | - Francesca Cavalieri
- School of Science, RMIT University, Melbourne, Victoria, 3000, Australia
- Dipartimento di Scienze e Tecnologie Chimiche, Università degli Studi di Roma "Tor Vergata", via della ricerca scientifica 1, Rome, 00133, Italy
| |
Collapse
|
30
|
Laka K, Makgoo L, Mbita Z. Cholesterol-Lowering Phytochemicals: Targeting the Mevalonate Pathway for Anticancer Interventions. Front Genet 2022; 13:841639. [PMID: 35391801 PMCID: PMC8981032 DOI: 10.3389/fgene.2022.841639] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/07/2022] [Indexed: 11/15/2022] Open
Abstract
There are a plethora of cancer causes and the road to fully understanding the carcinogenesis process remains a dream that keeps changing. However, a list of role players that are implicated in the carcinogens process is getting lengthier. Cholesterol is known as bad sterol that is heavily linked with cardiovascular diseases; however, it is also comprehensively associated with carcinogenesis. There is an extensive list of strategies that have been used to lower cholesterol; nevertheless, the need to find better and effective strategies remains vastly important. The role played by cholesterol in the induction of the carcinogenesis process has attracted huge interest in recent years. Phytochemicals can be dubbed as magic tramp cards that humans could exploit for lowering cancer-causing cholesterol. Additionally, the mechanisms that are regulated by phytochemicals can be targeted for anticancer drug development. One of the key role players in cancer development and suppression, Tumour Protein 53 (TP53), is crucial in regulating the biogenesis of cholesterol and is targeted by several phytochemicals. This minireview covers the role of p53 in the mevalonate pathway and how bioactive phytochemicals target the mevalonate pathway and promote p53-dependent anticancer activities.
Collapse
Affiliation(s)
| | | | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Sovenga, South Africa
| |
Collapse
|
31
|
It Takes More than Two to Tango: Complex, Hierarchal, and Membrane-Modulated Interactions in the Regulation of Receptor Tyrosine Kinases. Cancers (Basel) 2022; 14:cancers14040944. [PMID: 35205690 PMCID: PMC8869822 DOI: 10.3390/cancers14040944] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 12/18/2022] Open
Abstract
The search for an understanding of how cell fate and motility are regulated is not a purely scientific undertaking, but it can also lead to rationally designed therapies against cancer. The discovery of tyrosine kinases about half a century ago, the subsequent characterization of certain transmembrane receptors harboring tyrosine kinase activity, and their connection to the development of human cancer ushered in a new age with the hope of finding a treatment for malignant diseases in the foreseeable future. However, painstaking efforts were required to uncover the principles of how these receptors with intrinsic tyrosine kinase activity are regulated. Developments in molecular and structural biology and biophysical approaches paved the way towards better understanding of these pathways. Discoveries in the past twenty years first resulted in the formulation of textbook dogmas, such as dimerization-driven receptor association, which were followed by fine-tuning the model. In this review, the role of molecular interactions taking place during the activation of receptor tyrosine kinases, with special attention to the epidermal growth factor receptor family, will be discussed. The fact that these receptors are anchored in the membrane provides ample opportunities for modulatory lipid-protein interactions that will be considered in detail in the second part of the manuscript. Although qualitative and quantitative alterations in lipids in cancer are not sufficient in their own right to drive the malignant transformation, they both contribute to tumor formation and also provide ways to treat cancer. The review will be concluded with a summary of these medical aspects of lipid-protein interactions.
Collapse
|
32
|
Tindall AM, Stallings VA. Sex differences in cardiovascular risk may be related to sex differences in diet patterns: a narrative review. Ann Hum Biol 2022; 48:517-524. [PMID: 35105204 DOI: 10.1080/03014460.2021.1998621] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
CONTEXT Cardiovascular disease (CVD) is the leading cause of death in the United States and globally. The social and biological differences in diet patterns among men and women may play a role in their differential cardiovascular risk. OBJECTIVE To describe the dietary patterns associated with CVD risk and investigate the differences in diet patterns between men and women, if these differences affect cardiovascular risk, and to explore potential mechanisms of action. METHODS Diet patterns associated with CVD risk were described based on epidemiological and experimental trials. Observational and experimental studies together with systematic and non-systematic reviews were synthesised to examine sex differences in diet and cardiovascular risk factors. RESULTS Differences in vasculature and body composition between sexes may be mediated by dissimilarities in adherence to diet patterns and nutrient metabolism. Salt sensitivity and breakdown and storage of lipids may account for some differences in CVD risk between men and women. Sex differences in social norms, cognitive processing, and odour perception may be affected by biological differences and contribute to differences in cardiovascular risk and dietary patterns. CONCLUSION The relation between diet patterns and cardiovascular risk is well-established and sex differences in diet patterns likely differentially affect CVD risk between men and women.
Collapse
Affiliation(s)
- A M Tindall
- Division of Gastroenterology, Hepatology & Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - V A Stallings
- Division of Gastroenterology, Hepatology & Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Patel KK, Kashfi K. Lipoproteins and cancer: The role of HDL-C, LDL-C, and cholesterol-lowering drugs. Biochem Pharmacol 2022; 196:114654. [PMID: 34129857 PMCID: PMC8665945 DOI: 10.1016/j.bcp.2021.114654] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 02/03/2023]
Abstract
Cholesterol is an amphipathic sterol molecule that is vital for maintaining normal physiological homeostasis. It is a relatively complicated molecule with 27 carbons whose synthesis starts with 2-carbon units. This in itself signifies the importance of this molecule. Cholesterol serves as a precursor for vitamin D, bile acids, and hormones, including estrogens, androgens, progestogens, and corticosteroids. Although essential, high cholesterol levels are associated with cardiovascular and kidney diseases and cancer initiation, progression, and metastasis. Although there are some contrary reports, current literature suggests a positive association between serum cholesterol levels and the risk and extent of cancer development. In this review, we first present a brief overview of cholesterol biosynthesis and its transport, then elucidate the role of cholesterol in the progression of some cancers. Suggested mechanisms for cholesterol-mediated cancer progression are plentiful and include the activation of oncogenic signaling pathways and the induction of oxidative stress, among others. The specific roles of the lipoprotein molecules, high-density lipoprotein (HDL) and low-density lipoprotein (LDL), in this pathogenesis, are also reviewed. Finally, we hone on the potential role of some cholesterol-lowering medications in cancer.
Collapse
Affiliation(s)
- Kush K Patel
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, NY, USA.
| |
Collapse
|
34
|
Yu L, Wu M, Zhu G, Xu Y. Emerging Roles of the Tumor Suppressor p53 in Metabolism. Front Cell Dev Biol 2022; 9:762742. [PMID: 35118064 PMCID: PMC8806078 DOI: 10.3389/fcell.2021.762742] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/27/2021] [Indexed: 01/31/2023] Open
Abstract
Metabolism plays critical roles in maintaining the homeostasis of cells. Metabolic abnormalities are often considered as one of the main driving forces for cancer progression, providing energy and substrates of biosynthesis to support neoplastic proliferation effectively. The tumor suppressor p53 is well known for its roles in inducing cell cycle arrest, apoptosis, senescence and ferroptosis. Recently, emerging evidence has shown that p53 is also actively involved in the reprogramming of cellular metabolism. In this review, we focus on recent advances in our understanding of the interplay between p53 and metabolism of glucose, fatty acid as well as amino acid, and discuss how the deregulation of p53 in these processes could lead to cancer.
Collapse
Affiliation(s)
- Lili Yu
- Key Laboratory of Cancer Prevention and Intervention, Department of Medical Oncology, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Meng Wu
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Gaoyang Zhu
- Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
- *Correspondence: Gaoyang Zhu, ; Yang Xu,
| | - Yang Xu
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
- *Correspondence: Gaoyang Zhu, ; Yang Xu,
| |
Collapse
|
35
|
Wilczyński B, Dąbrowska A, Saczko J, Kulbacka J. The Role of Chloride Channels in the Multidrug Resistance. MEMBRANES 2021; 12:38. [PMID: 35054564 PMCID: PMC8781147 DOI: 10.3390/membranes12010038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/23/2021] [Indexed: 12/19/2022]
Abstract
Nowadays, one of medicine's main and most challenging aims is finding effective ways to treat cancer. Unfortunately, although there are numerous anti-cancerous drugs, such as cisplatin, more and more cancerous cells create drug resistance. Thus, it is equally important to find new medicines and research the drug resistance phenomenon and possibilities to avoid this mechanism. Ion channels, including chloride channels, play an important role in the drug resistance phenomenon. Our article focuses on the chloride channels, especially the volume-regulated channels (VRAC) and CLC chloride channels family. VRAC induces multidrug resistance (MDR) by causing apoptosis connected with apoptotic volume decrease (AVD) and VRAC are responsible for the transport of anti-cancerous drugs such as cisplatin. VRACs are a group of heterogenic complexes made from leucine-rich repetition with 8A (LRRC8A) and a subunit LRRC8B-E responsible for the properties. There are probably other subunits, which can create those channels, for example, TTYH1 and TTYH2. It is also known that the ClC family is involved in creating MDR in mainly two mechanisms-by changing the cell metabolism or acidification of the cell. The most researched chloride channel from this family is the CLC-3 channel. However, other channels are playing an important role in inducing MDR as well. In this paper, we review the role of chloride channels in MDR and establish the role of the channels in the MDR phenomenon.
Collapse
Affiliation(s)
- Bartosz Wilczyński
- Faculty of Medicine, Wroclaw Medical University, L. Pasteura 1, 50-367 Wroclaw, Poland; (B.W.); (A.D.)
| | - Alicja Dąbrowska
- Faculty of Medicine, Wroclaw Medical University, L. Pasteura 1, 50-367 Wroclaw, Poland; (B.W.); (A.D.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
36
|
Deng Z, Yang Z, Peng J. Role of bioactive peptides derived from food proteins in programmed cell death to treat inflammatory diseases and cancer. Crit Rev Food Sci Nutr 2021:1-19. [PMID: 34694177 DOI: 10.1080/10408398.2021.1992606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Bioactive peptides are specific peptide which usually contains 2-20 amino acid residues and actively exerts various functions and biological activities and ultimately affect health. Programmed cell deaths are some styles of cell death discovered in recent years, which is the key to tissue development and balance, eliminating excess, damaged or aging cells. More importantly, programmed cell death is a potential way to treat inflammatory diseases and cancer. In this review, through screening references from 2015 to present, we introduce the effect of bioactive peptides derived from food proteins on inflammatory diseases or cancer through regulating programmed cell deaths, including apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis. And this review also introduces the targets of these bioactive peptides to regulate programmed cell death. The purpose of this review is to help to expand the prospective applications of bioactive peptides in the field of inflammatory disease and cancer to provide some guidance.
Collapse
Affiliation(s)
- Zhao Deng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zhipeng Yang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, P. R. China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| |
Collapse
|
37
|
Xi Y, Yani Z, Jing M, Yinhang W, Xiaohui H, Jing Z, Quan Q, Shuwen H. Mechanisms of induction of tumors by cholesterol and potential therapeutic prospects. Biomed Pharmacother 2021; 144:112277. [PMID: 34624674 DOI: 10.1016/j.biopha.2021.112277] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggested that cholesterol is an important integrant of cell membranes, that plays a key role in tumor progression, immune dysregulation, and pathological changes in epigenetic mechanisms. Based on these theories, there is a growing interest on targeting cholesterol in the treatment of cancer. Here, we comprehensively reviewed the major function of cholesterol on oncogenicity, the therapeutic targets of cholesterol and its metabolites in cancer, and provide detailed insight into the essential roles of cholesterol in mediating immune and epigenetic mechanisms of the tumor microenvironment. It is also worth mentioning that the gut microbiome is an indispensable component of cancer mediation because of its role in cholesterol metabolism. Finally, we summarized recent studies on the potential targets of cholesterol and their metabolism, to provide more therapeutic interventions in oncology.
Collapse
Affiliation(s)
- Yang Xi
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| | - Zhou Yani
- Graduate School of Medical College of Zhejiang University, No. 268 Kaixuan Road, Jianggan District, Hangzhou, Zhejiang 310029, China.
| | - Mao Jing
- Graduate School of Medical College of Zhejiang University, No. 268 Kaixuan Road, Jianggan District, Hangzhou, Zhejiang 310029, China.
| | - Wu Yinhang
- Graduate School of Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China.
| | - Hou Xiaohui
- Graduate School of Nursing, Huzhou University, No. 1 Bachelor Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| | - Zhuang Jing
- Department of Nursing, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| | - Qi Quan
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| | - Han Shuwen
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang 313000, China.
| |
Collapse
|
38
|
Abdelwahed KS, Siddique AB, Qusa MH, King JA, Souid S, Abd Elmageed ZY, El Sayed KA. PCSK9 Axis-Targeting Pseurotin A as a Novel Prostate Cancer Recurrence Suppressor Lead. ACS Pharmacol Transl Sci 2021; 4:1771-1781. [DOI: 10.1021/acsptsci.1c00145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Indexed: 01/02/2023]
Affiliation(s)
- Khaldoun S. Abdelwahed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, Louisiana 71201, United States
| | - Abu Bakar Siddique
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, Louisiana 71201, United States
| | - Mohammed H. Qusa
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, Louisiana 71201, United States
| | - Judy Ann King
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, Louisiana 71103, United States
| | - Soumaya Souid
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, Louisiana 71201, United States
| | - Zakaria Y. Abd Elmageed
- Department of Pharmacology, Edward Via College of Osteopathic Medicine, University of Louisiana at Monroe, Monroe, Louisiana 71203, United States
| | - Khalid A. El Sayed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, Louisiana 71201, United States
| |
Collapse
|
39
|
Angles G, Hail A, Dotson RJ, Pias SC. Atomistic simulations modify interpretation of spin-label oximetry data. Part 1: intensified water-lipid interfacial resistances. APPLIED MAGNETIC RESONANCE 2021; 52:1261-1289. [PMID: 37292189 PMCID: PMC10249954 DOI: 10.1007/s00723-021-01398-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 06/10/2023]
Abstract
The role of membrane cholesterol in cellular function and dysfunction has been the subject of much inquiry. A few studies have suggested that cholesterol may slow oxygen diffusive transport, altering membrane physical properties and reducing oxygen permeability. The primary experimental technique used in recent years to study membrane oxygen transport is saturation-recovery electron paramagnetic resonance (EPR) oximetry, using spin-label probes targeted to specific regions of a lipid bilayer. The technique has been used, in particular, to assess the influence of cholesterol on oxygen transport and membrane permeability. The reliability of such EPR recordings at the water-lipid interface near the phospholipid headgroups has been challenged by all-atom molecular dynamics (MD) simulation data that show substantive agreement with spin-label probe measurements throughout much of the bilayer. This work uses further MD simulations, with an updated oxygen model, to determine the location of the maximum resistance to permeation and the rate-limiting barrier to oxygen permeation in 1-palmitoyl,2-oleoylphosphatidylcholine (POPC) and POPC/cholesterol bilayers at 25 and 35°C. The current simulations show a spike of resistance to permeation in the headgroup region that was not detected by EPR but was predicted in early theoretical work by Diamond and Katz. Published experimental nuclear magnetic resonance (NMR) oxygen measurements provide key validation of the MD models and indicate that the positions and relative magnitudes of the phosphatidylcholine resistance peaks are accurate. Consideration of the headgroup-region resistances predicts bilayer permeability coefficients lower than estimated in EPR studies, giving permeabilities lower than the permeability of unstirred water layers of the same thickness. Here, the permeability of POPC at 35°C is estimated to be 13 cm/s, compared with 10 cm/s for POPC/cholesterol and 118 cm/s for simulation water layers of similar thickness. The value for POPC is 12 times lower than estimated from EPR measurements, while the value for POPC/cholesterol is 5 times lower. These findings underscore the value of atomic resolution models for guiding the interpretation of experimental probe-based measurements.
Collapse
Affiliation(s)
| | | | | | - Sally C. Pias
- Corresponding author: , Department of Chemistry, New Mexico Institute of Mining and Technology (New Mexico Tech), 801 Leroy Place, Socorro, NM 87801, USA
| |
Collapse
|
40
|
Pandey M, Cuddihy G, Gordon JA, Cox ME, Wasan KM. Inhibition of Scavenger Receptor Class B Type 1 (SR-B1) Expression and Activity as a Potential Novel Target to Disrupt Cholesterol Availability in Castration-Resistant Prostate Cancer. Pharmaceutics 2021; 13:1509. [PMID: 34575583 PMCID: PMC8467449 DOI: 10.3390/pharmaceutics13091509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
There have been several studies that have linked elevated scavenger receptor class b type 1 (SR-B1) expression and activity to the development and progression of castration-resistant prostate cancer (CRPC). SR-B1 facilitates the influx of cholesterol to the cell from lipoproteins in systemic circulation. This influx of cholesterol may be important for many cellular functions, including the synthesis of androgens. Castration-resistant prostate cancer tumors can synthesize androgens de novo to supplement the loss of exogenous sources often induced by androgen deprivation therapy. Silencing of SR-B1 may impact the ability of prostate cancer cells, particularly those of the castration-resistant state, to maintain the intracellular supply of androgens by removing a supply of cholesterol. SR-B1 expression is elevated in CRPC models and has been linked to poor survival of patients. The overarching belief has been that cholesterol modulation, through either synthesis or uptake inhibition, will impact essential signaling processes, impeding the proliferation of prostate cancer. The reduction in cellular cholesterol availability can impede prostate cancer proliferation through both decreased steroid synthesis and steroid-independent mechanisms, providing a potential therapeutic target for the treatment of prostate cancer. In this article, we discuss and highlight the work on SR-B1 as a potential novel drug target for CRPC management.
Collapse
Affiliation(s)
- Mitali Pandey
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Grace Cuddihy
- College of Pharmacy and Nutrition, University of Saskatchewan, 104 Clinic Place, Saskatoon, SK S7N 2Z4, Canada;
| | - Jacob A. Gordon
- Oncology Bioscience, Oncology R&D, AstraZeneca, Boston, MA 02451, USA;
| | - Michael E. Cox
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Kishor M. Wasan
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| |
Collapse
|
41
|
Marzocco S, Singla RK, Capasso A. Multifaceted Effects of Lycopene: A Boulevard to the Multitarget-Based Treatment for Cancer. Molecules 2021; 26:molecules26175333. [PMID: 34500768 PMCID: PMC8434243 DOI: 10.3390/molecules26175333] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/29/2021] [Accepted: 08/29/2021] [Indexed: 02/05/2023] Open
Abstract
Lycopene is a pigment belonging to the group of carotenoids and it is among the most carefully studied antioxidants found especially in fruit and vegetables. As a carotenoid, lycopene exerts beneficial effects on human health by protecting lipids, proteins, and DNA from damage by oxidation. Lycopene is a powerful oxygen inactivator in the singlet state. This is suggestive of the fact that lycopene harbors comparatively stronger antioxidant properties over other carotenoids normally present in plasma. Lycopene is also reported to hinder cancer cell proliferation. The uncontrolled, rapid division of cells is a characteristic of the metabolism of cancer cells. Evidently, lycopene causes a delay in the progression of the cell cycle, which explains its antitumor activity. Furthermore, lycopene can block cell transformation by reducing the loss of contact inhibition of cancer cells. This paper collects recent studies of scientific evidence that show the multiple beneficial properties of lycopene, which acts with different molecular and cellular mechanisms.
Collapse
Affiliation(s)
- Stefania Marzocco
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
- Correspondence: ; Tel.: +39-089-96-92-50
| | - Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China;
- iGlobal Research and Publishing Foundation, New Delhi 110059, India
| | - Anna Capasso
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
| |
Collapse
|
42
|
Huang KCY, Chiang SF, Yang PC, Ke TW, Chen TW, Lin CY, Chang HY, Chen WTL, Chao KSC. ATAD3A stabilizes GRP78 to suppress ER stress for acquired chemoresistance in colorectal cancer. J Cell Physiol 2021; 236:6481-6495. [PMID: 33580514 DOI: 10.1002/jcp.30323] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
AAA domain containing 3A (ATAD3A) is a nucleus-encoded mitochondrial protein with vital function in communication between endoplasmic reticulum (ER) and mitochondria which is participated in cancer metastasis. Here we show that elevated ATAD3A expression is clinically associated with poor 5-year disease-free survival in patients with colorectal cancer (CRC), especially high-risk CRC patients who received adjuvant chemotherapy. Our results indicated ATAD3A is significantly upregulated to reduce chemotherapy-induced cancer cell death. We found that knockdown of ATAD3A leads to dysregulation in protein processing for inducing ER stress by RNA sequencing (RNA-seq). In response to chemotherapy-induced ER stress, ATAD3A interacts with elevated GRP78 protein to assist protein folding and alleviate ER stress for cancer cell survival. This reduction of ER stress leads to reduce the surface exposure of calreticulin, which is the initiator of immunogenic cell death and antitumor immunity. However, silencing of ATAD3A enhances cell death, triggers the feasibility of chemotherapy-induced ER stress for antitumor immunity, increases infiltration of T lymphocytes and delays tumor regrowth in vitro and in vivo. Clinically, CRC patients with less ATAD3A have high density of CD45+ intratumoral infiltrating lymphocytes (TILs) and memory CD45RO+ TILs. Taken together, our results suggest that pharmacologic targeting to ATAD3A might be a potential therapeutic strategy to enhance antitumor immunity for CRC patients who received adjuvant chemotherapy.
Collapse
Affiliation(s)
- Kevin Chih-Yang Huang
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Shu-Fen Chiang
- Lab of Precision Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Taichung, Taiwan
- Cancer Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Pei-Chen Yang
- Cancer Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Tao-Wei Ke
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Chinese Medicine & Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Tsung-Wei Chen
- Department of Pathology, Asia University Hospital, Asia University, Taichung, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Chen-Yu Lin
- Cancer Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Hsin-Yu Chang
- Cancer Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - William Tzu-Liang Chen
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Colorectal Surgery, China Medical University HsinChu Hospital, China Medical University, HsinChu, Taiwan
- Department of Surgery, School of Medicine, China Medical University, Taichung, Taiwan
| | - Kun-San Clifford Chao
- Cancer Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Radiotherapy, School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
43
|
Hoogerheide DP, Rostovtseva TK, Bezrukov SM. Exploring lipid-dependent conformations of membrane-bound α-synuclein with the VDAC nanopore. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2021; 1863:183643. [PMID: 33971161 PMCID: PMC8255272 DOI: 10.1016/j.bbamem.2021.183643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023]
Abstract
Regulation of VDAC by α-synuclein (αSyn) is a rich and instructive example of protein-protein interactions catalyzed by a lipid membrane surface. αSyn, a peripheral membrane protein involved in Parkinson's disease pathology, is known to bind to membranes in a transient manner. αSyn's negatively charged C-terminal domain is then available to be electromechanically trapped by the VDAC β-barrel, a process that is observed in vitro as the reversible reduction of ion flow through a single voltage-biased VDAC nanopore. Binding of αSyn to the lipid bilayer is a prerequisite of the channel-protein interaction; surprisingly, however, we find that the strength of αSyn binding to the membrane does not correlate in any simple way with its efficiency of blocking VDAC, suggesting that the lipid-dependent conformations of the membrane-bound αSyn control the interaction. Quantitative models of the free energy landscape governing the capture and release processes allow us to discriminate between several αSyn (sub-) conformations on the membrane surface. These results, combined with known structural features of αSyn on anionic lipid membranes, point to a model in which the lipid composition determines the fraction of αSyn molecules for which the charged C terminal domain is constrained to be close, but not tightly bound, to the membrane surface and thus readily captured by the VDAC nanopore. We speculate that changes in the mitochondrial membrane lipid composition may be key regulators of the αSyn-VDAC interaction and consequently of VDAC-facilitated transport of ions and metabolites in and out of mitochondria and, i.e. mitochondrial metabolism.
Collapse
Affiliation(s)
- David P Hoogerheide
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Tatiana K Rostovtseva
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Sergey M Bezrukov
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
44
|
Dietary and Genetic Cholesterol Loading Rather Than Steatosis Promotes Liver Tumorigenesis and NASH-Driven HCC. Cancers (Basel) 2021; 13:cancers13164091. [PMID: 34439245 PMCID: PMC8393403 DOI: 10.3390/cancers13164091] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary In the present study, which is part of the Special Issue “Theranostic Advances in Hepatobiliary Tumors”, we address whether hepatic steatosis per se or cholesterol sensitizes to NASH-driven HCC. This is a very important health issue, as the incidence of HCC derived from NASH is expected to keep rising due to the association of NASH with the obesity and type 2 diabetes epidemic. Using dietary and genetic models to generate hepatic steatosis with or without cholesterol accumulation, we provide evidence for the tumor promoter role of cholesterol in NASH-HCC associated with an increased expression of the genes involved in immune checkpoints, which suggests that cholesterol favors a milieu prone to T-cell exhaustion. Abstract The association of nonalcoholic steatohepatitis (NASH) with obesity and type 2 diabetes is a major determinant factor for the continued rise of NASH-driven HCC. Unfortunately, the mechanisms underlying the progression from NASH to HCC are not well-understood. Steatosis is characterized by the accumulation of different lipid species, and cholesterol has emerged as an important player in NASH development, which has been shown to promote NASH-driven HCC. However, recent findings indicated a tumor suppressor role of cholesterol in liver carcinogenesis and HCC development. Thus, we examined the contribution of hepatic steatosis with or without cholesterol accumulation induced by dietary or genetic approaches in liver tumorigenesis and whether the role of cholesterol in NASH-driven HCC is species-dependent. While diethylnitrosamine (DEN) treatment to rats or mice fed a choline-deficient diet decreased the hepatic steatosis, feeding an atherogenic diet enriched in cholesterol potentiated the liver tumor markers. Similar effects were observed in DEN-treated transgenic SREBP-2 mice but not wild-type (WT) mice fed a regular chow diet. Remarkably, long-term feeding of a high-fat high-cholesterol diet (HFHC) but not a high-fat diet (HFD) to WT mice caused severe NASH with spontaneous progression to HCC. A similar outcome was observed in MUP-uPA transgenic mice fed a HFHC diet, which resulted in increased liver tumors and expression of the genes involved in the immune checkpoints. Ezetimibe treatment ameliorated chronic liver disease and, more importantly, tumor multiplicity in HFHC-fed MUP-uPA mice or DEN-treated WT mice. Thus, these results revealed a differential role of steatosis and cholesterol in NASH-driven HCC and indicated that the tumor-promoter role of cholesterol is species-independent and associated with impaired immunosurveillance.
Collapse
|
45
|
Zhang E, Chen Y, Bao S, Hou X, Hu J, Mu OYN, Song Y, Shan L. Identification of subgroups along the glycolysis-cholesterol synthesis axis and the development of an associated prognostic risk model. Hum Genomics 2021; 15:53. [PMID: 34384498 PMCID: PMC8359075 DOI: 10.1186/s40246-021-00350-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/26/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Skin cutaneous melanoma (SKCM) is one of the most highly prevalent and complicated malignancies. Glycolysis and cholesterogenesis pathways both play important roles in cancer metabolic adaptations. The main aims of this study are to subtype SKCM based on glycolytic and cholesterogenic genes and to build a clinical outcome predictive algorithm based on the subtypes. METHODS A dataset with 471 SKCM specimens was downloaded from The Cancer Genome Atlas (TCGA) database. We extracted and clustered genes from the Molecular Signatures Database v7.2 and acquired co-expressed glycolytic and cholesterogenic genes. We then subtyped the SKCM samples and validated the efficacy of subtypes with respect to simple nucleotide variations (SNVs), copy number variation (CNV), patients' survival statuses, tumor microenvironment, and proliferation scores. We also constructed a risk score model based on metabolic subclassification and verified the model using validating datasets. Finally, we explored potential drugs for high-risk SKCM patients. RESULTS SKCM patients were divided into four subtype groups: glycolytic, cholesterogenic, mixed, and quiescent subgroups. The glycolytic subtype had the worst prognosis and MGAM SNV extent. Compared with the cholesterogenic subgroup, the glycolytic subgroup had higher rates of DDR2 and TPR CNV and higher proliferation scores and MK167 expression levels, but a lower tumor purity proportion. We constructed a forty-four-gene predictive signature and identified MST-321, SB-743921, Neuronal Differentiation Inducer III, romidepsin, vindesine, and YM-155 as high-sensitive drugs for high-risk SKCM patients. CONCLUSIONS Subtyping SKCM patients via glycolytic and cholesterogenic genes was effective, and patients in the glycolytic-gene enriched group were found to have the worst outcome. A robust prognostic algorithm was developed to enhance clinical decisions in relation to drug administration.
Collapse
Affiliation(s)
- Enchong Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Yijing Chen
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- School of Postgraduate, China Medical University, Shenyang, Liaoning, China
| | - Shurui Bao
- Department of Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xueying Hou
- School of Postgraduate, China Medical University, Shenyang, Liaoning, China
| | - Jing Hu
- School of Postgraduate, China Medical University, Shenyang, Liaoning, China
| | | | - Yongsheng Song
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Liping Shan
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
46
|
Garcia-Ruiz C, Conde de la Rosa L, Ribas V, Fernandez-Checa JC. MITOCHONDRIAL CHOLESTEROL AND CANCER. Semin Cancer Biol 2021; 73:76-85. [PMID: 32805396 PMCID: PMC7882000 DOI: 10.1016/j.semcancer.2020.07.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022]
Abstract
Cholesterol is a crucial component of membrane bilayers that determines their physical and functional properties. Cells largely satisfy their need for cholesterol through the novo synthesis from acetyl-CoA and this demand is particularly critical for cancer cells to sustain dysregulated cell proliferation. However, the association between serum or tissue cholesterol levels and cancer development is not well established as epidemiologic data do not consistently support this link. While most preclinical studies focused on the role of total celular cholesterol, the specific contribution of the mitochondrial cholesterol pool to alterations in cancer cell biology has been less explored. Although low compared to other bilayers, the mitochondrial cholesterol content plays an important physiological function in the synthesis of steroid hormones in steroidogenic tissues or bile acids in the liver and controls mitochondrial function. In addition, mitochondrial cholesterol metabolism generates oxysterols, which in turn, regulate multiple pathways, including cholesterol and lipid metabolism as well as cell proliferation. In the present review, we summarize the regulation of mitochondrial cholesterol, including its role in mitochondrial routine performance, cell death and chemotherapy resistance, highlighting its potential contribution to cancer. Of particular relevance is hepatocellular carcinoma, whose incidence in Western countries had tripled in the past decades due to the obesity and type II diabetes epidemic. A better understanding of the role of mitochondrial cholesterol in cancer development may open up novel opportunities for cancer therapy.
Collapse
Affiliation(s)
- Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Vicent Ribas
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Jose C Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
47
|
Razin T, Melamed-Book N, Argaman J, Galin I, Lowy Y, Anuka E, Naftali-Shani N, Kandel-Kfir M, Garfinkel BP, Brielle S, Granot Z, Apte RN, Conway SJ, Molkentin JD, Kamari Y, Leor J, Orly J. Interleukin-1α dependent survival of cardiac fibroblasts is associated with StAR/STARD1 expression and improved cardiac remodeling and function after myocardial infarction. J Mol Cell Cardiol 2021; 155:125-137. [PMID: 33130150 PMCID: PMC11492233 DOI: 10.1016/j.yjmcc.2020.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/18/2020] [Accepted: 10/25/2020] [Indexed: 12/19/2022]
Abstract
AIMS One unaddressed aspect of healing after myocardial infarction (MI) is how non-myocyte cells that survived the ischemic injury, keep withstanding additional cellular damage by stress forms typically arising during the post-infarction inflammation. Here we aimed to determine if cell survival is conferred by expression of a mitochondrial protein novel to the cardiac proteome, known as steroidogenic acute regulatory protein, (StAR/STARD1). Further studies aimed to unravel the regulation and role of the non-steroidogenic cardiac StAR after MI. METHODS AND RESULTS Following permanent ligation of the left anterior descending coronary artery in mouse heart, timeline western blot analyses showed that StAR expression corresponds to the inflammatory response to MI. Following the identification of StAR in mitochondria of cardiac fibroblasts in culture, confocal microscopy immunohistochemistry (IHC) identified StAR expression in left ventricular (LV) activated interstitial fibroblasts, adventitial fibroblasts and endothelial cells. Further work with the primary fibroblasts model revealed that interleukin-1α (IL-1α) signaling via NF-κB and p38 MAPK pathways efficiently upregulates the expression of the Star gene products. At the functional level, IL-1α primed fibroblasts were protected against apoptosis when exposed to cisplatin mimicry of in vivo apoptotic stress; yet, the protective impact of IL-1α was lost upon siRNA mediated StAR downregulation. At the physiological level, StAR expression was nullified during post-MI inflammation in a mouse model with global IL-1α deficiency, concomitantly resulting in a 4-fold elevation of apoptotic fibroblasts. Serial echocardiography and IHC studies of mice examined 24 days after MI revealed aggravation of LV dysfunction, LV dilatation, anterior wall thinning and adverse tissue remodeling when compared with loxP control hearts. CONCLUSIONS This study calls attention to overlooked aspects of cellular responses evolved under the stress conditions associated with the default inflammatory response to MI. Our observations suggest that LV IL-1α is cardioprotective, and at least one mechanism of this action is mediated by induction of StAR expression in border zone fibroblasts, which renders them apoptosis resistant. This acquired survival feature also has long-term ramifications on the heart recovery by diminishing adverse remodeling and improving the heart function after MI.
Collapse
Affiliation(s)
- Talya Razin
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Naomi Melamed-Book
- Bio-Imaging Unit, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Jasmin Argaman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Iris Galin
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Yosef Lowy
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Eli Anuka
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Nili Naftali-Shani
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Tamman Cardiovascular Research Institute, and Sheba Center of Regenerative Medicine, Stem Cells and Tissue Engineering, Sheba Medical Center, Tel-Hashomer, Israel.
| | - Michal Kandel-Kfir
- Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel, Sackler Faculty of Medicine, Tel-Aviv University, Israel. Michal.KandelKfir-@sheba.health.gov.il
| | - Benjamin P Garfinkel
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Shlomi Brielle
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University Medical School, 91120 Jerusalem, Israel.
| | - Ron N Apte
- Shraga Segal Department of Microbiology and Immunology, Ben-Gurion University, Beer-Sheva, Israel.
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, IN 46202, United States.
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH, United States.
| | - Yehuda Kamari
- Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel, Sackler Faculty of Medicine, Tel-Aviv University, Israel.
| | - Jonathan Leor
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Tamman Cardiovascular Research Institute, and Sheba Center of Regenerative Medicine, Stem Cells and Tissue Engineering, Sheba Medical Center, Tel-Hashomer, Israel.
| | - Joseph Orly
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| |
Collapse
|
48
|
Conde de la Rosa L, Garcia-Ruiz C, Vallejo C, Baulies A, Nuñez S, Monte MJ, Marin JJG, Baila-Rueda L, Cenarro A, Civeira F, Fuster J, Garcia-Valdecasas JC, Ferrer J, Karin M, Ribas V, Fernandez-Checa JC. STARD1 promotes NASH-driven HCC by sustaining the generation of bile acids through the alternative mitochondrial pathway. J Hepatol 2021; 74:1429-1441. [PMID: 33515644 PMCID: PMC8573791 DOI: 10.1016/j.jhep.2021.01.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Besides their physiological role in bile formation and fat digestion, bile acids (BAs) synthesised from cholesterol in hepatocytes act as signalling molecules that modulate hepatocellular carcinoma (HCC). Trafficking of cholesterol to mitochondria through steroidogenic acute regulatory protein 1 (STARD1) is the rate-limiting step in the alternative pathway of BA generation, the physiological relevance of which is not well understood. Moreover, the specific contribution of the STARD1-dependent BA synthesis pathway to HCC has not been previously explored. METHODS STARD1 expression was analyzed in a cohort of human non-alcoholic steatohepatitis (NASH)-derived HCC specimens. Experimental NASH-driven HCC models included MUP-uPA mice fed a high-fat high-cholesterol (HFHC) diet and diethylnitrosamine (DEN) treatment in wild-type (WT) mice fed a HFHC diet. Molecular species of BAs and oxysterols were analyzed by mass spectrometry. Effects of NASH-derived BA profiles were investigated in tumour-initiated stem-like cells (TICs) and primary mouse hepatocytes (PMHs). RESULTS Patients with NASH-associated HCC exhibited increased hepatic expression of STARD1 and an enhanced BA pool. Using NASH-driven HCC models, STARD1 overexpression in WT mice increased liver tumour multiplicity, whereas hepatocyte-specific STARD1 deletion (Stard1ΔHep) in WT or MUP-uPA mice reduced tumour burden. These findings mirrored the levels of unconjugated primary BAs, β-muricholic acid and cholic acid, and their tauroconjugates in STARD1-overexpressing and Stard1ΔHep mice. Incubation of TICs or PMHs with a mix of BAs mimicking this profile stimulated expression of genes involved in pluripotency, stemness and inflammation. CONCLUSIONS The study reveals a previously unrecognised role of STARD1 in HCC pathogenesis, wherein it promotes the synthesis of primary BAs through the mitochondrial pathway, the products of which act in TICs to stimulate self-renewal, stemness and inflammation. LAY SUMMARY Effective therapy for hepatocellular carcinoma (HCC) is limited because of our incomplete understanding of its pathogenesis. The contribution of the alternative pathway of bile acid (BA) synthesis to HCC development is unknown. We uncover a key role for steroidogenic acute regulatory protein 1 (STARD1) in non-alcoholic steatohepatitis-driven HCC, wherein it stimulates the generation of BAs in the mitochondrial acidic pathway, the products of which stimulate hepatocyte pluripotency and self-renewal, as well as inflammation.
Collapse
Affiliation(s)
- Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Carmen Vallejo
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Anna Baulies
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Susana Nuñez
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Maria J Monte
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Experimental Hepatology and Drug Targeting (HEVEFARM), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Jose J G Marin
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Experimental Hepatology and Drug Targeting (HEVEFARM), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Lucia Baila-Rueda
- Instituto Investigación Sanitaria Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain; CIBERCV, Madrid, Spain
| | - Ana Cenarro
- Instituto Investigación Sanitaria Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain; CIBERCV, Madrid, Spain
| | - Fernando Civeira
- Instituto Investigación Sanitaria Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain; CIBERCV, Madrid, Spain
| | - Josep Fuster
- HepatoBilioPancreatic Surgery and Liver and Pancreatic Transplantation Unit, Department of Surgery, ICMDiM, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Juan C Garcia-Valdecasas
- HepatoBilioPancreatic Surgery and Liver and Pancreatic Transplantation Unit, Department of Surgery, ICMDiM, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Joana Ferrer
- HepatoBilioPancreatic Surgery and Liver and Pancreatic Transplantation Unit, Department of Surgery, ICMDiM, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Vicent Ribas
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain.
| | - Jose C Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain; Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
49
|
Mollinedo F, Gajate C. Mitochondrial Targeting Involving Cholesterol-Rich Lipid Rafts in the Mechanism of Action of the Antitumor Ether Lipid and Alkylphospholipid Analog Edelfosine. Pharmaceutics 2021; 13:763. [PMID: 34065546 PMCID: PMC8161315 DOI: 10.3390/pharmaceutics13050763] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
The ether lipid edelfosine induces apoptosis selectively in tumor cells and is the prototypic molecule of a family of synthetic antitumor compounds collectively known as alkylphospholipid analogs. Cumulative evidence shows that edelfosine interacts with cholesterol-rich lipid rafts, endoplasmic reticulum (ER) and mitochondria. Edelfosine induces apoptosis in a number of hematological cancer cells by recruiting death receptors and downstream apoptotic signaling into lipid rafts, whereas it promotes apoptosis in solid tumor cells through an ER stress response. Edelfosine-induced apoptosis, mediated by lipid rafts and/or ER, requires the involvement of a mitochondrial-dependent step to eventually elicit cell death, leading to the loss of mitochondrial membrane potential, cytochrome c release and the triggering of cell death. The overexpression of Bcl-2 or Bcl-xL blocks edelfosine-induced apoptosis. Edelfosine induces the redistribution of lipid rafts from the plasma membrane to the mitochondria. The pro-apoptotic action of edelfosine on cancer cells is associated with the recruitment of F1FO-ATP synthase into cholesterol-rich lipid rafts. Specific inhibition of the FO sector of the F1FO-ATP synthase, which contains the membrane-embedded c-subunit ring that constitutes the mitochondrial permeability transcription pore, hinders edelfosine-induced cell death. Taking together, the evidence shown here suggests that the ether lipid edelfosine could modulate cell death in cancer cells by direct interaction with mitochondria, and the reorganization of raft-located mitochondrial proteins that critically modulate cell death or survival. Here, we summarize and discuss the involvement of mitochondria in the antitumor action of the ether lipid edelfosine, pointing out the mitochondrial targeting of this drug as a major therapeutic approach, which can be extrapolated to other alkylphospholipid analogs. We also discuss the involvement of cholesterol transport and cholesterol-rich lipid rafts in the interactions between the organelles as well as in the role of mitochondria in the regulation of apoptosis in cancer cells and cancer therapy.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, C/Ramiro de Maeztu 9, E-28040 Madrid, Spain;
| | | |
Collapse
|
50
|
de Oliveira MF, Medeiros RCA, Mietto BS, Calvo TL, Mendonça APM, Rosa TLSA, da Silva DS, do Carmo de Vasconcelos KG, Pereira AMR, de Macedo CS, Pereira GMB, de Berrêdo Pinho Moreira M, Pessolani MCV, Moraes MO, Lara FA. Reduction of host cell mitochondrial activity as Mycobacterium leprae's strategy to evade host innate immunity. Immunol Rev 2021; 301:193-208. [PMID: 33913182 PMCID: PMC10084840 DOI: 10.1111/imr.12962] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
Leprosy is a much-feared incapacitating infectious disease caused by Mycobacterium leprae or M lepromatosis, annually affecting roughly 200,000 people worldwide. During host-pathogen interaction, M leprae subverts the immune response, leading to development of disease. Throughout the last few decades, the impact of energy metabolism on the control of intracellular pathogens and leukocytic differentiation has become more evident. Mitochondria play a key role in regulating newly-discovered immune signaling pathways by controlling redox metabolism and the flow of energy besides activating inflammasome, xenophagy, and apoptosis. Likewise, this organelle, whose origin is probably an alphaproteobacterium, directly controls the intracellular pathogens attempting to invade its niche, a feature conquered at the expense of billions of years of coevolution. In the present review, we discuss the role of reduced host cell mitochondrial activity during M leprae infection and the consequential fates of M leprae and host innate immunity. Conceivably, inhibition of mitochondrial energy metabolism emerges as an overlooked and novel mechanism developed by M leprae to evade xenophagy and the host immune response.
Collapse
Affiliation(s)
- Marcus Fernandes de Oliveira
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Bruno Siqueira Mietto
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Minas Gerais, Brazil
| | - Thyago Leal Calvo
- Laboratório de Hanseníase, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Ana Paula Miranda Mendonça
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | - Cristiana Santos de Macedo
- Laboratório de Microbiologia Celular, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
- Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | - Flavio Alves Lara
- Laboratório de Microbiologia Celular, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| |
Collapse
|