1
|
Alaouna M, Molefi T, Khanyile R, Chauke-Malinga N, Chatziioannou A, Luvhengo TE, Raletsena M, Penny C, Hull R, Dlamini Z. The potential of the South African plant Tulbaghia Violacea Harv for the treatment of triple negative breast cancer. Sci Rep 2025; 15:5737. [PMID: 39962120 PMCID: PMC11832780 DOI: 10.1038/s41598-025-88417-2] [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: 07/03/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is difficult to treat and has a low five-year survival rate. In South Africa, a large percentage of the population still relies on traditional plant-based medicine. To establish the utility of both methanol and water-soluble extracts from the leaves of Tulbaghia violacea, cytotoxicity assays were carried out to establish the IC50 values against a TNBC cell line. Cell cycle and apoptosis assays were carried out using the extracts. To identify the molecular compounds, present in water-soluble leaf extracts, NMR spectroscopy was performed. Compounds of interest were then used in computational docking studies with the anti-apoptotic protein COX-2. The IC50 values for the water- and methanol-soluble extracts were determined to be 400 and 820 µg/mL, respectively. The water-soluble extract induced apoptosis in the TNBC cell line to a greater extent than in the normal cell line. RNAseq indicated that there was an increase in the transcription of pro-apoptotic genes in the TNBC cell line. The crude extract also caused these cells to stall in the S phase. Of the 61 compounds identified in this extract, five demonstrated a high binding affinity for COX-2. Based on these findings, the compounds within the extract show significant potential for further investigation as candidates for the development of cancer therapeutics, particularly for TNBC.
Collapse
Affiliation(s)
- Mohammed Alaouna
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Thulo Molefi
- Department of Medical Oncology, Steve Biko Academic Hospital, University of Pretoria, Pretoria, 0001, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Richard Khanyile
- Department of Medical Oncology, Steve Biko Academic Hospital, University of Pretoria, Pretoria, 0001, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Nkhensani Chauke-Malinga
- Papillon Aesthetics, Suite 302b Netcare Linksfield Hospital, 24 12th Ave, Linksfield West, Johannesburg, 2192, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Aristotelis Chatziioannou
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa
| | - Thifhelimbilu Emmanuel Luvhengo
- Department of Surgery, Charlotte Maxeke Johannesburg Academic Hospital, University of the Witwatersrand, Parktown, Johannesburg, 2193, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maropeng Raletsena
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemistry, University of South Africa, Florida Campus, Johannesburg, South Africa
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rodney Hull
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa.
| | - Zodwa Dlamini
- Department of Chemical pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- SA-MRC Precision Oncology Research Unit (PORU), DSTI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria Hatfield, Pretoria, 0028, South Africa.
| |
Collapse
|
2
|
Murphy CS, Fairfield H, DeMambro VE, Fadel S, Gartner CA, Karam M, Potts C, Rodriguez P, Qiang YW, Hamidi H, Guan X, Vary CPH, Reagan MR. Inhibition of acyl-CoA synthetase long-chain isozymes decreases multiple myeloma cell proliferation and causes mitochondrial dysfunction. Mol Oncol 2025. [PMID: 39853696 DOI: 10.1002/1878-0261.13794] [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: 11/18/2024] [Revised: 12/14/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
Multiple myeloma (MM) is an incurable cancer of plasma cells with a 5-year survival rate of 59%. Dysregulation of fatty acid (FA) metabolism is associated with MM development and progression; however, the underlying mechanisms remain unclear. Herein, we explore the roles of long-chain fatty acid coenzyme A ligase (ACSL) family members in MM. ACSLs convert free long-chain fatty acids into fatty acyl-CoA esters and play key roles in catabolic and anabolic fatty acid metabolism. Analysis of the Multiple Myeloma Research Foundation (MMRF) CoMMpassSM study showed that high ACSL1 and ACSL4 expression in myeloma cells are both associated with worse clinical outcomes for MM patients. Cancer Dependency Map (DepMap) data showed that all five ACSLs have negative Chronos scores, and ACSL3 and ACSL4 were among the top 25% Hallmark Fatty Acid Metabolism genes that support myeloma cell line fitness. Inhibition of ACSLs in myeloma cell lines in vitro, using the pharmacological inhibitor Triacsin C (TriC), increased apoptosis, decreased proliferation, and decreased cell viability, in a dose- and time-dependent manner. RNA-sequencing analysis of MM.1S cells treated with TriC showed a significant enrichment in apoptosis, ferroptosis, and endoplasmic reticulum (ER) stress, and proteomic analysis of these cells revealed enriched pathways for mitochondrial dysfunction and oxidative phosphorylation. TriC also rewired mitochondrial metabolism by decreasing mitochondrial membrane potential, increasing mitochondrial superoxide levels, decreasing mitochondrial ATP production rates, and impairing cellular respiration. Overall, our data support the hypothesis that suppression of ACSLs in myeloma cells is a novel metabolic target in MM that inhibits their viability, implicating this family as a promising therapeutic target in treating myeloma.
Collapse
Affiliation(s)
- Connor S Murphy
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Heather Fairfield
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
| | - Victoria E DeMambro
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Samaa Fadel
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
- University of New England, Biddeford, ME, USA
| | - Carlos A Gartner
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Michelle Karam
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | - Christian Potts
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | - Princess Rodriguez
- Vermont Integrative Genomics Resource DNA Facility, University of Vermont, Burlington, VT, USA
| | - Ya-Wei Qiang
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | | | | | - Calvin P H Vary
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
| | - Michaela R Reagan
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
| |
Collapse
|
3
|
Pu KL, Kang H, Li L. Therapeutic targets for age-related macular degeneration: proteome-wide Mendelian randomization and colocalization analyses. Front Neurol 2024; 15:1400557. [PMID: 38903171 PMCID: PMC11187347 DOI: 10.3389/fneur.2024.1400557] [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: 03/13/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Background Currently, effective therapeutic drugs for age-related macular degeneration (AMD) are urgently needed, and it is crucial to explore new treatment targets. The proteome is indispensable for exploring disease targets, so we conducted a Mendelian randomization (MR) of the proteome to identify new targets for AMD and its related subtypes. Methods The plasma protein level data used in this study were obtained from two large-scale studies of protein quantitative trait loci (pQTL), comprising 35,559 and 54,219 samples, respectively. The expression quantitative trait loci (eQTL) data were sourced from eQTLGen and GTEx Version 8. The discovery set for AMD data and subtypes was derived from the FinnGen study, consisting of 9,721 AMD cases and 381,339 controls, 5,239 wet AMD cases and 273,920 controls, and 6,651 dry AMD cases and 272,504 controls. The replication set for AMD data was obtained from the study by Winkler TW et al., comprising 14,034 cases and 91,234 controls. Summary Mendelian randomization (SMR) analysis was employed to assess the association between QTL data and AMD and its subtypes, while colocalization analysis was performed to determine whether they share causal variants. Additionally, chemical exploration and molecular docking were utilized to validate potential drugs targeting the identified proteins. Results SMR and colocalization analysis jointly identified risk-associated proteins for AMD and its subtypes, including 5 proteins (WARS1, BRD2, IL20RB, TGFB1, TNFRSF10A) associated with AMD, 2 proteins (WARS1, IL20RB) associated with Dry-AMD, and 9 proteins (COL10A1, WARS1, VTN, SDF2, LBP, CD226, TGFB1, TNFRSF10A, CSF2) associated with Wet-AMD. The results revealed potential therapeutic chemicals, and molecular docking indicated a good binding between the chemicals and protein structures. Conclusion Proteome-wide MR have identified risk-associated proteins for AMD and its subtypes, suggesting that these proteins may serve as potential therapeutic targets worthy of further clinical investigation.
Collapse
Affiliation(s)
- Kun-Lin Pu
- Pengzhou Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Hong Kang
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Chengdu, China
| | - Li Li
- Pengzhou Hospital of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Safi R, Menéndez P, Pol A. Lipid droplets provide metabolic flexibility for cancer progression. FEBS Lett 2024; 598:1301-1327. [PMID: 38325881 DOI: 10.1002/1873-3468.14820] [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: 09/04/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
A hallmark of cancer cells is their remarkable ability to efficiently adapt to favorable and hostile environments. Due to a unique metabolic flexibility, tumor cells can grow even in the absence of extracellular nutrients or in stressful scenarios. To achieve this, cancer cells need large amounts of lipids to build membranes, synthesize lipid-derived molecules, and generate metabolic energy in the absence of other nutrients. Tumor cells potentiate strategies to obtain lipids from other cells, metabolic pathways to synthesize new lipids, and mechanisms for efficient storage, mobilization, and utilization of these lipids. Lipid droplets (LDs) are the organelles that collect and supply lipids in eukaryotes and it is increasingly recognized that the accumulation of LDs is a new hallmark of cancer cells. Furthermore, an active role of LD proteins in processes underlying tumorigenesis has been proposed. Here, by focusing on three major classes of LD-resident proteins (perilipins, lipases, and acyl-CoA synthetases), we provide an overview of the contribution of LDs to cancer progression and discuss the role of LD proteins during the proliferation, invasion, metastasis, apoptosis, and stemness of cancer cells.
Collapse
Affiliation(s)
- Rémi Safi
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
| |
Collapse
|
5
|
Murphy CS, DeMambro VE, Fadel S, Fairfield H, Garter CA, Rodriguez P, Qiang YW, Vary CPH, Reagan MR. Inhibition of Acyl-CoA Synthetase Long Chain Isozymes Decreases Multiple Myeloma Cell Proliferation and Causes Mitochondrial Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.583708. [PMID: 38559245 PMCID: PMC10979990 DOI: 10.1101/2024.03.13.583708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Multiple myeloma (MM) is an incurable cancer of plasma cells with a 5-year survival rate of 59%. Dysregulation of fatty acid (FA) metabolism is associated with MM development and progression; however, the underlying mechanisms remain unclear. Acyl-CoA synthetase long-chain family members (ACSLs) convert free long-chain fatty acids into fatty acyl-CoA esters and play key roles in catabolic and anabolic fatty acid metabolism. The Cancer Dependency Map data suggested that ACSL3 and ACSL4 were among the top 25% Hallmark Fatty Acid Metabolism genes that support MM fitness. Here, we show that inhibition of ACSLs in human myeloma cell lines using the pharmacological inhibitor Triascin C (TriC) causes apoptosis and decreases proliferation in a dose- and time-dependent manner. RNA-seq of MM.1S cells treated with TriC for 24 h showed a significant enrichment in apoptosis, ferroptosis, and ER stress. Proteomics of MM.1S cells treated with TriC for 48 h revealed that mitochondrial dysfunction and oxidative phosphorylation were significantly enriched pathways of interest, consistent with our observations of decreased mitochondrial membrane potential and increased mitochondrial superoxide levels. Interestingly, MM.1S cells treated with TriC for 24 h also showed decreased mitochondrial ATP production rates and overall lower cellular respiration.
Collapse
Affiliation(s)
- Connor S Murphy
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine, University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA
| | - Victoria E DeMambro
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine, University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA
| | - Samaa Fadel
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of New England, Biddeford, ME, USA
| | - Heather Fairfield
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine, University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA
- Tufts University School of Medicine, Boston MA, USA
| | - Carlos A Garter
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine, University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA
| | | | - Ya-Wei Qiang
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | - Calvin P H Vary
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine, University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA
- Tufts University School of Medicine, Boston MA, USA
| | - Michaela R Reagan
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- University of Maine, University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA
- Tufts University School of Medicine, Boston MA, USA
| |
Collapse
|
6
|
Wang W, Wang P, Zhu L, Liu B, Wei Q, Hou Y, Li X, Hu Y, Li W, Wang Y, Jiang C, Yang G, Wang J. An optimized fluorescent biosensor for monitoring long-chain fatty acyl-CoAs metabolism in vivo. Biosens Bioelectron 2024; 247:115935. [PMID: 38128319 DOI: 10.1016/j.bios.2023.115935] [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: 10/17/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Long-chain fatty acyl-CoAs (LCACoAs) are intermediates in lipid metabolism that exert a wide range of cellular functions. However, our knowledge about the subcellular distribution and regulatory impacts of LCACoAs is limited by a lack of methods for detecting LCACoAs in living cells and tissues. Here, we report our development of LACSerHR, a genetically encoded fluorescent biosensor that enables precise measurement of subtle fluctuations in the levels of endogenous LCACoAs in vivo. LACSerHR significantly improve the fluorescent brightness and analyte affinity, in vitro and in vivo testing showcased LACSerHR's large dynamic range. We demonstrate LACSerHR's capacity for real-time evaluation of LCACoA levels in specific subcellular compartments, for example in response to disruption of ACSL enzyme function in HEK293T cells. Moreover, we show the application of LACSerHR for sensitive measurement of elevated LCACoA levels in the livers of mouse models for two common metabolic diseases (NAFLD and type 2 diabetes). Thus, our LACSerHR sensor is a powerful, broadly applicable tool for studying LCACoAs metabolism and disease.
Collapse
Affiliation(s)
- Weibo Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China; National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, 430079, PR China
| | - Pengcheng Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, PR China
| | - Lixin Zhu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Bingjie Liu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Qingpeng Wei
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yongkang Hou
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Xi Li
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yufei Hu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, PR China
| | - Guangfu Yang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, 430079, PR China.
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China.
| |
Collapse
|
7
|
Liu Z, Guo X, Zhang W, Wang J, Zhang L, Jing J, Han L, Gao A. Oxidative stress-affected ACSL1 hydroxymethylation triggered benzene hematopoietic toxicity by inflammation and senescence. Food Chem Toxicol 2023; 180:114030. [PMID: 37689099 DOI: 10.1016/j.fct.2023.114030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/02/2023] [Accepted: 09/05/2023] [Indexed: 09/11/2023]
Abstract
Long-term benzene exposure is harmful and causes hematopoietic dysfunction. However, the mechanism of benzene hematopoietic toxicity is still unclear. Acyl-CoA Synthetase Long-Chain Family Member 1 (ACSL1) has been found to participate in the progress of a variety of benign and malignant diseases, but there is no research about its effect on benzene-induced hematopoietic toxicity. Herein, We exposed C57BL/6J mice to benzene to construct an in vivo model. Human peripheral blood mononuclear cells (THP-1 cells) were treated with benzene metabolite 1, 4-BQ to construct an in vitro model. We observed that the ACSL1 expression was upregulated both in vivo and in vitro. Moreover, inhibition of ACSL1 relieved inflammation and senescence development in vitro, suggesting that ACSL1 mediates inflammation and senescence. As for the regulation mechanism of ACSL1 expression, it is closely related to hydroxymethylation modification. This was proved by hydroxymethylated DNA immunoprecipitation (hMeDIP) experiments. Furthermore, oxidative stress influenced the hydroxymethylation process. These results showed that benzene hematopoietic toxicity occurs through the induction of oxidative stress and thus the regulation of ACSL1 hydroxymethylation, which in turn mediates inflammation and senescence. Thus, this study might be of great significance in identifying and preventing benzene exposure in the early stage.
Collapse
Affiliation(s)
- Ziyan Liu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Xiaoli Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Wei Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Jingyu Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Lei Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Jiaru Jing
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Lin Han
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Ai Gao
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
8
|
Zareinejad M, Mehdipour F, Roshan-Zamir M, Faghih Z, Ghaderi A. Dual Functions of T Lymphocytes in Breast Carcinoma: From Immune Protection to Orchestrating Tumor Progression and Metastasis. Cancers (Basel) 2023; 15:4771. [PMID: 37835465 PMCID: PMC10571747 DOI: 10.3390/cancers15194771] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer (BC) is the most common cancer type in women and the second leading cause of death. Despite recent advances, the mortality rate of BC is still high, highlighting a need to develop new treatment strategies including the modulation of the immune system and immunotherapies. In this regard, understanding the complex function of the involved immune cells and their crosstalk with tumor cells is of great importance. T-cells are recognized as the most important cells in the tumor microenvironment and are divided into several subtypes including helper, cytotoxic, and regulatory T-cells according to their transcription factors, markers, and functions. This article attempts to provide a comprehensive review of the role of T-cell subsets in the prognosis and treatment of patients with BC, and crosstalk between tumor cells and T-cells. The literature overwhelmingly contains controversial findings mainly due to the plasticity of T-cell subsets within the inflammatory conditions and the use of different panels for their phenotyping. However, investigating the role of T-cells in BC immunity depends on a variety of factors including tumor types or subtypes, the stage of the disease, the localization of the cells in the tumor tissue and the presence of different cells or cytokines.
Collapse
Affiliation(s)
| | | | | | - Zahra Faghih
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45505, Iran; (M.Z.); (F.M.); (M.R.-Z.)
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45505, Iran; (M.Z.); (F.M.); (M.R.-Z.)
| |
Collapse
|
9
|
Al-Rashed F, Haddad D, Al Madhoun A, Sindhu S, Jacob T, Kochumon S, Obeid LM, Al-Mulla F, Hannun YA, Ahmad R. ACSL1 is a key regulator of inflammatory and macrophage foaming induced by short-term palmitate exposure or acute high-fat feeding. iScience 2023; 26:107145. [PMID: 37416456 PMCID: PMC10320618 DOI: 10.1016/j.isci.2023.107145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/29/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Foamy and inflammatory macrophages play pathogenic roles in metabolic disorders. However, the mechanisms that promote foamy and inflammatory macrophage phenotypes under acute-high-fat feeding (AHFF) remain elusive. Herein, we investigated the role of acyl-CoA synthetase-1 (ACSL1) in favoring the foamy/inflammatory phenotype of monocytes/macrophages upon short-term exposure to palmitate or AHFF. Palmitate exposure induced a foamy/inflammatory phenotype in macrophages which was associated with increased ACSL1 expression. Inhibition/knockdown of ACSL1 in macrophages suppressed the foamy/inflammatory phenotype through the inhibition of the CD36-FABP4-p38-PPARδ signaling axis. ACSL1 inhibition/knockdown suppressed macrophage foaming/inflammation after palmitate stimulation by downregulating the FABP4 expression. Similar results were obtained using primary human monocytes. As expected, oral administration of ACSL1 inhibitor triacsin-C in mice before AHFF normalized the inflammatory/foamy phenotype of the circulatory monocytes by suppressing FABP4 expression. Our results reveal that targeting ACSL1 leads to the attenuation of the CD36-FABP4-p38-PPARδ signaling axis, providing a therapeutic strategy to prevent the AHFF-induced macrophage foaming and inflammation.
Collapse
Affiliation(s)
- Fatema Al-Rashed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
| | - Dania Haddad
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
| | - Ashraf Al Madhoun
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
| | - Sardar Sindhu
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
| | - Texy Jacob
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
| | - Shihab Kochumon
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
| | - Lina M. Obeid
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
| | - Yusuf A. Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City, Dasman 15462, Kuwait
| |
Collapse
|
10
|
Alqinyah M, Alhamed AS, Alnefaie HO, Algahtani MM, Badr AM, Albogami AM, Mohany M, Alassmrry YA, Alghaith AF, Alhamami HN, Alhazzani K, Alanazi AZ, Alsaidan OA. Targeting Store-Operated Calcium Entry Regulates the Inflammation-Induced Proliferation and Migration of Breast Cancer Cells. Biomedicines 2023; 11:1637. [PMID: 37371732 PMCID: PMC10296208 DOI: 10.3390/biomedicines11061637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/24/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Persistent challenges complicating the treatment of breast cancer remain, despite some recent undeniable successes. Sufficient evidence currently exists demonstrating the crucial role of inflammation, characterized by the enhanced activation of Toll-like receptor 4 (TLR4) and the COX-2/PGE2 pathway, in the migration and proliferation of breast cancer cells. Interestingly, the store-operated calcium entry (SOCE) pathway was shown to be essential for the TLR4 activity and COX-2 expression in immune cells such as macrophages and microglia. However, whether SOCE influences inflammatory signaling and the inflammation-induced proliferation and migration of breast cancer cells is still unknown. Thus, the current study intended to delineate the role of SOCE in the TLR4-induced inflammation, migration, and proliferation of breast cancer cells. To this end, MDA-MB-231 breast cancer cells were treated with lipopolysaccharide (LPS) to activate TLR4, BTP2 to inhibit SOCE, and Thapsigargin to induce SOCE. Following these treatments, several experiments were conducted to evaluate the proliferation and migration rates of the MDA-MB-231 cells and the expression of several inflammatory and oncogenic genes, including COX-2, PGE2, IL-6, IL-8, and VEGF. Different techniques were used to achieve the aims of this study, including qRT-PCR, Western blotting, ELISA, MTT, and wound healing assays. This study shows that SOCE inhibition using BTP2 suppressed the LPS-induced migration and proliferation of breast cancer cells. Additionally, treatment with LPS caused approximately six- and three-fold increases in COX-2 mRNA and protein expression, respectively, compared to the controls. The LPS-induced elevations in the COX-2 mRNA and protein levels were suppressed by BTP2 to the control levels. In addition to its effect on COX-2, BTP2 also suppressed the LPS-induced productions of PGE2, IL-6, IL-8, and VEGF. Conversely, SOCE induction using Thapsigargin enhanced the LPS-induced inflammation, migration, and proliferation of breast cancer cells. Collectively, these results provide evidence for the potentially important role of SOCE in inflammation-induced breast cancer progression processes. Thus, we argue that the current study may provide novel targets for designing new therapeutic approaches for the treatment of breast cancer.
Collapse
Affiliation(s)
- Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S. Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hajar O. Alnefaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad M. Algahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Amira M. Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah M. Albogami
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yasseen A. Alassmrry
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Adel F. Alghaith
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hussain N. Alhamami
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Z. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| |
Collapse
|
11
|
Xu M, Ye S, Tang Z, Gong S. Bone Marrow Mesenchymal Stem Cells Restrain the Migration and Invasion of Breast Cancer Cells by Up-Regulating miR-2158 and Inactivating RAI2/NLRP3 Pathway. J BIOMATER TISS ENG 2023. [DOI: 10.1166/jbt.2023.3233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Exosomes are the key mediator for intercellular communication and participate in malignancies. Short non-coding RNAs derived from BMSCs-originated exosomes (BMSCs-exosomes) can be employed as biomolecules for tumor treatment. Here to we aim to dissect the function of microRNA-2158 from
BMSCs-exosomes in breast malignant disease. Breast malignant cells received a separated transfection of miR-2158-mimics and miR-2158-inhibitor, and also treated with BMSCoriginated exosomes followed by analysis of cell viability by MTT method, cell invading and migrating capabilities via Transwell
assays and protein levels of EMT-related and RAI2/NLRP3-related proteins by Western-blot. Breast cancer cells exhibited a significantly enhanced miR-2158 expression after transfection with miR-2158-mimics or treatment with BMSC-EXO, while it was reduced by miR-2158-inhibitor. As the miR-2158
was up-regulated, a significant impediment of proliferation and migration was denoted, along with a down-regulation of RAI2/NLRP3 signal transduction pathway and a retarded EMT process. Furthermore, cell proliferating and migrating capabilities were strengthened by miR-2158-inhibitor, together
with an enhanced RAI2/NLRP3 signal and a strengthened EMT process. In conclusion, miR-2158 retarded the in vitro proliferating and migrating activities of breast malignant cells, leading to the inactivation of RAI2/NLRP3 signal transduction pathway, thereby exerting its tumor-suppressing
function.
Collapse
Affiliation(s)
- Meiyu Xu
- Department of General Surgery, South Campus of the Sixth People’s Hospital Affiliated to Shanghai Jiaotong University (Shanghai Fengxian District Central Hospital), Shanghai, 201499, China
| | - Shen Ye
- Department of General Surgery, South Campus of the Sixth People’s Hospital Affiliated to Shanghai Jiaotong University (Shanghai Fengxian District Central Hospital), Shanghai, 201499, China
| | - Zhiqiang Tang
- Department of General Surgery, South Campus of the Sixth People’s Hospital Affiliated to Shanghai Jiaotong University (Shanghai Fengxian District Central Hospital), Shanghai, 201499, China
| | - Shuai Gong
- Department of Radiotherapy, Qingdao Municipal Hospital, Qingdao, Shangdong, 266000, China
| |
Collapse
|
12
|
Xia H, Zhang Z, You F. Inhibiting ACSL1-Related Ferroptosis Restrains Murine Coronavirus Infection. Viruses 2021; 13:2383. [PMID: 34960652 PMCID: PMC8708337 DOI: 10.3390/v13122383] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/02/2022] Open
Abstract
Murine hepatitis virus strain A59 (MHV-A59) was shown to induce pyroptosis, apoptosis, and necroptosis of infected cells, especially in the murine macrophages. However, whether ferroptosis, a recently identified form of lytic cell death, was involved in the pathogenicity of MHV-A59 is unknown. We utilized murine macrophages and a C57BL/6 mice intranasal infection model to address this. In primary macrophages, the ferroptosis inhibitor inhibited viral propagation, inflammatory cytokines released, and cell syncytia formed after MHV-A59 infection. In the mouse model, we found that in vivo administration of liproxstatin-1 ameliorated lung inflammation and tissue injuries caused by MHV-A59 infection. To find how MHV-A59 infection influenced the expression of ferroptosis-related genes, we performed RNA-seq in primary macrophages and found that MHV-A59 infection upregulates the expression of the acyl-CoA synthetase long-chain family member 1 (ACSL1), a novel ferroptosis inducer. Using ferroptosis inhibitors and a TLR4 inhibitor, we showed that MHV-A59 resulted in the NF-kB-dependent, TLR4-independent ACSL1 upregulation. Accordingly, ACSL1 inhibitor Triacsin C suppressed MHV-A59-infection-induced syncytia formation and viral propagation in primary macrophages. Collectively, our study indicates that ferroptosis inhibition protects hosts from MHV-A59 infection. Targeting ferroptosis may serve as a potential treatment approach for dealing with hyper-inflammation induced by coronavirus infection.
Collapse
Affiliation(s)
| | | | - Fuping You
- Department of Systems Biomedicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (H.X.); (Z.Z.)
| |
Collapse
|
13
|
Al-Roub A, Al Madhoun A, Akhter N, Thomas R, Miranda L, Jacob T, Al-Ozairi E, Al-Mulla F, Sindhu S, Ahmad R. IL-1β and TNFα Cooperativity in Regulating IL-6 Expression in Adipocytes Depends on CREB Binding and H3K14 Acetylation. Cells 2021; 10:3228. [PMID: 34831450 PMCID: PMC8619559 DOI: 10.3390/cells10113228] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022] Open
Abstract
IL-6 was found to be overexpressed in the adipose tissue of obese individuals, which may cause insulin resistance. However, the regulation of IL-6 in adipocytes in obesity setting remains to be explored. Since IL-1β and TNFα are increased in obese adipose tissue and promote inflammation, we investigated whether cooperation between IL-1β and TNFα influences the production of IL-6. Our data show that IL-1β and TNFα cooperatively enhance IL-6 expression in 3T3L-1 adipocytes. Similar results were seen in human adipocytes isolated from subcutaneous and visceral fat. Although adipocytes isolated from lean and obese adipose tissues showed similar responses for production of IL-6 when incubated with IL-1β/TNFα, secretion of IL-6 was higher in adipocytes from obese tissue. TNFα treatment enhanced CREB binding at CRE locus, which was further enhanced with IL-1β, and was associated with elevated histone acetylation at CRE locus. On the other hand, IL-1β treatments mediated C/EBPβ binding to NF-IL-6 consensus, but not sufficiently to mediate significant histone acetylation. Interestingly, treatment with both stimulatory factors amplifies CREB binding and H3K14 acetylation. Furthermore, histone acetylation inhibition by anacardic acid or curcumin reduces IL-6 production. Notably, inhibition of histone deacetylase (HDAC) activity by trichostatin A (TSA) resulted in the further elevation of IL-6 expression in response to combined treatment of adipocytes with IL-1β and TNFα. In conclusion, our results show that there is an additive interaction between IL-1β and TNFα that depends on CREB binding and H3K14 acetylation, and leads to the elevation of IL-6 expression in adipocytes, providing interesting pathophysiological connection among IL-1β, TNFα, and IL-6 in settings such as obesity.
Collapse
Affiliation(s)
- Areej Al-Roub
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| | - Ashraf Al Madhoun
- Genetics & Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (L.M.); (F.A.-M.)
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Nadeem Akhter
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| | - Reeby Thomas
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| | - Lavina Miranda
- Genetics & Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (L.M.); (F.A.-M.)
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Texy Jacob
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| | - Ebaa Al-Ozairi
- Medical Division, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Fahd Al-Mulla
- Genetics & Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (L.M.); (F.A.-M.)
| | - Sardar Sindhu
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (R.T.); (T.J.); (S.S.)
| |
Collapse
|
14
|
Haider M, Al-Rashed F, Albaqsumi Z, Alobaid K, Alqabandi R, Al-Mulla F, Ahmad R. Candida albicans Induces Foaming and Inflammation in Macrophages through FABP4: Its Implication for Atherosclerosis. Biomedicines 2021; 9:biomedicines9111567. [PMID: 34829801 PMCID: PMC8615257 DOI: 10.3390/biomedicines9111567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 01/05/2023] Open
Abstract
Atherosclerosis is a chronic degenerative disorder characterized by lipid-dense plaques and low-grade inflammation affecting arterial walls. Foamy macrophages are important in the formation of atherosclerotic plaques and the induction of low-grade inflammation. The presence of lipid-laden macrophages has occurred in infections caused by opportunistic pathogens. Candida albicans is the major cause of candidiasis in immunocompromised patients, including those with diabetes mellitus. However, the role played by C. albicans in macrophage foaming and the associated inflammation is poorly understood. We investigated whether C. albicans induces foaming along with inflammation in macrophages and, if so, by which mechanism(s). We incubated THP-1 macrophages with heat-killed C. albicans (HKCA). HKCA-induced lipid accumulation in macrophages along with increased expression of inflammatory markers, including CD11b and CD11c or expression and secretion of IL-1β. HKCA also increased the expression of PPARγ, CD36, and FABP4 in macrophages. Mechanistically, we found that the foamy and inflammatory macrophage phenotype induced by HKCA requires FABP4 because disruption of FABP4 in macrophages either by chemical inhibitor BMS309404 or small interfering RNA (siRNA) abrogated foam cell formation and expression of inflammatory markers CD11b, CD11c, and IL-1β. Furthermore, HKCA-treated macrophages displayed high expression and secretion of MMP-9. Inhibition of FABP4 resulted in suppression of HCKA-induced MMP-9 production. Overall, our results demonstrate that C. albicans induces foam cell formation, inflammation, and MMP-9 expression in macrophages via the upregulation of FABP4, which may constitute a novel therapeutic target for treating C. albicans-induced atherosclerosis.
Collapse
Affiliation(s)
- Mohammed Haider
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City 15462, Kuwait;
| | - Fatema Al-Rashed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City 15462, Kuwait; (F.A.-R.); (Z.A.); (R.A.)
| | - Zahraa Albaqsumi
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City 15462, Kuwait; (F.A.-R.); (Z.A.); (R.A.)
| | - Khaled Alobaid
- Mycology Reference Laboratory, Medical Laboratory Department, Mubarak Al-Kabeer Hospital, Kuwait City 15462, Kuwait;
| | - Rawan Alqabandi
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City 15462, Kuwait; (F.A.-R.); (Z.A.); (R.A.)
| | - Fahd Al-Mulla
- Genetics & Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Kuwait City 15462, Kuwait; (F.A.-R.); (Z.A.); (R.A.)
- Correspondence: ; Tel.: +965-2224-2999 (ext. 4311)
| |
Collapse
|
15
|
Kochumon S, Al-Sayyar A, Jacob T, Hasan A, Al-Mulla F, Sindhu S, Ahmad R. TNF-α Increases IP-10 Expression in MCF-7 Breast Cancer Cells via Activation of the JNK/c-Jun Pathways. Biomolecules 2021; 11:biom11091355. [PMID: 34572567 PMCID: PMC8464892 DOI: 10.3390/biom11091355] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
IP-10 (also called CXCL10) plays a significant role in leukocyte homing to inflamed tissues, and increased IP-10 levels are associated with the pathologies of various inflammatory disorders, including type 2 diabetes, atherosclerosis, and cancer. TNF-α is a potent activator of immune cells and induces inflammatory cytokine expression in these cells. However, it is unclear whether TNF-α is able to induce IP-10 expression in MCF-7 breast cancer cells. We therefore determined IP-10 expression in TNF-α-treated MCF-7 cells and investigated the mechanism involved. Our data show that TNF-α induced/upregulated the IP-10 expression at both mRNA and protein levels in MCF-7 cells. Inhibition of JNK (SP600125) significantly suppressed the TNF-α-induced IP-10 in MCF-7 cells, while the inhibition of p38 MAPK (SB203580), MEK1/2 (U0126), and ERK1/2 (PD98059) had no significant effect. Furthermore, TNF-α-induced IP-10 expression was abolished in MCF-7 cells deficient in JNK. Similar results were obtained using MCF-7 cells deficient in c-Jun. Moreover, the JNK kinase inhibitor markedly reduced the TNF-α-induced JNK and c-Jun phosphorylation. The kinase activity of JNK induced by TNF-α stimulation of MCF-7 cells was significantly inhibited by SP600125. Altogether, our novel findings provide the evidence that TNF-α induces IP-10 expression in MCF-7 breast cancer cells via activation of the JNK/c-Jun signaling pathway.
Collapse
Affiliation(s)
- Shihab Kochumon
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Amnah Al-Sayyar
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Texy Jacob
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Amal Hasan
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
| | - Fahd Al-Mulla
- Genetics & Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Sardar Sindhu
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (A.A.-S.); (T.J.); (A.H.); (S.S.)
- Correspondence:
| |
Collapse
|
16
|
Quan J, Bode AM, Luo X. ACSL family: The regulatory mechanisms and therapeutic implications in cancer. Eur J Pharmacol 2021; 909:174397. [PMID: 34332918 DOI: 10.1016/j.ejphar.2021.174397] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/14/2021] [Accepted: 07/28/2021] [Indexed: 12/29/2022]
Abstract
Accumulating evidence shows that deregulation of fatty acid (FA) metabolism is associated with the development of cancer. Long-chain acyl-coenzyme A synthases (ACSLs) are responsible for activating long-chain FAs and are frequently deregulated in cancers. Among the five mammalian ACSL family members, ACSL1 is involved in the TNFα-mediated pro-inflammatory phenotype and mainly facilitates cancer progression. ACSL3 is an androgen-responsive gene. High ACSL3 expression has been detected in a variety of cancers, including melanoma, triple-negative breast cancer (TNBC) and high-grade non-small cell lung carcinoma (NSCLC), and correlates with worse prognosis of patients with these diseases. ACSL4 can exert opposing roles acting as a tumor suppressor or as an oncogene depending on the specific cancer type and tissue environment. Moreover, ACSL4 behaves as a crucial regulator in ferroptosis that is defined as a cell death process caused by iron-dependent peroxidation of lipids. ACSL5 is nuclear-coded and expressed in the mitochondria and physiologically participates in the pro-apoptotic sensing of cells. ACSL5 mainly acts as a tumor suppressor in cancers. ACSL6 downregulation has been observed in many forms of cancers, except in colorectal cancer (CRC). Here, we address the differential regulatory mechanisms of the ACSL family members as well as their functions in carcinogenesis. Moreover, we enumerate the clinical therapeutic implications of ACSLs, which might serve as valuable biomarkers and therapeutic targets for precision cancer treatment.
Collapse
Affiliation(s)
- Jing Quan
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, PR China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, 410078, PR China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan, 410078, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, PR China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, 410078, PR China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, Hunan, 410078, China; Molecular Imaging Research Center of Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
17
|
Al-Roub A, Akhter N, Al-Sayyar A, Wilson A, Thomas R, Kochumon S, Al-Rashed F, Al-Mulla F, Sindhu S, Ahmad R. Short Chain Fatty Acid Acetate Increases TNFα-Induced MCP-1 Production in Monocytic Cells via ACSL1/MAPK/NF-κB Axis. Int J Mol Sci 2021; 22:ijms22147683. [PMID: 34299302 PMCID: PMC8304091 DOI: 10.3390/ijms22147683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
Short-chain fatty acid (SCFA) acetate, a byproduct of dietary fiber metabolism by gut bacteria, has multiple immunomodulatory functions. The anti-inflammatory role of acetate is well documented; however, its effect on monocyte chemoattractant protein-1 (MCP-1) production is unknown. Similarly, the comparative effect of SCFA on MCP-1 expression in monocytes and macrophages remains unclear. We investigated whether acetate modulates TNFα-mediated MCP-1/CCL2 production in monocytes/macrophages and, if so, by which mechanism(s). Monocytic cells were exposed to acetate with/without TNFα for 24 h, and MCP-1 expression was measured. Monocytes treated with acetate in combination with TNFα resulted in significantly greater MCP-1 production compared to TNFα treatment alone, indicating a synergistic effect. On the contrary, treatment with acetate in combination with TNFα suppressed MCP-1 production in macrophages. The synergistic upregulation of MCP-1 was mediated through the activation of long-chain fatty acyl-CoA synthetase 1 (ACSL1). However, the inhibition of other bioactive lipid enzymes [carnitine palmitoyltransferase I (CPT I) or serine palmitoyltransferase (SPT)] did not affect this synergy. Moreover, MCP-1 expression was significantly reduced by the inhibition of p38 MAPK, ERK1/2, and NF-κB signaling. The inhibition of ACSL1 attenuated the acetate/TNFα-mediated phosphorylation of p38 MAPK, ERK1/2, and NF-κB. Increased NF-κB/AP-1 activity, resulting from acetate/TNFα co-stimulation, was decreased by ACSL1 inhibition. In conclusion, this study demonstrates the proinflammatory effects of acetate on TNF-α-mediated MCP-1 production via the ACSL1/MAPK/NF-κB axis in monocytic cells, while a paradoxical effect was observed in THP-1-derived macrophages.
Collapse
Affiliation(s)
- Areej Al-Roub
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (A.A.-S.); (A.W.); (R.T.); (S.K.); (F.A.-R.)
| | - Nadeem Akhter
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (A.A.-S.); (A.W.); (R.T.); (S.K.); (F.A.-R.)
| | - Amnah Al-Sayyar
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (A.A.-S.); (A.W.); (R.T.); (S.K.); (F.A.-R.)
| | - Ajit Wilson
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (A.A.-S.); (A.W.); (R.T.); (S.K.); (F.A.-R.)
| | - Reeby Thomas
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (A.A.-S.); (A.W.); (R.T.); (S.K.); (F.A.-R.)
| | - Shihab Kochumon
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (A.A.-S.); (A.W.); (R.T.); (S.K.); (F.A.-R.)
| | - Fatema Al-Rashed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (A.A.-S.); (A.W.); (R.T.); (S.K.); (F.A.-R.)
| | - Fahd Al-Mulla
- Genetics & Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Sardar Sindhu
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.-R.); (N.A.); (A.A.-S.); (A.W.); (R.T.); (S.K.); (F.A.-R.)
- Correspondence: ; Tel.: +965-2224-2999 (ext. 4311)
| |
Collapse
|
18
|
Wilkie T, Verma AK, Zhao H, Charan M, Ahirwar DK, Kant S, Pancholi V, Mishra S, Ganju RK. Lipopolysaccharide from the commensal microbiota of the breast enhances cancer growth: role of S100A7 and TLR4. Mol Oncol 2021; 16:1508-1522. [PMID: 33969603 PMCID: PMC8978520 DOI: 10.1002/1878-0261.12975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 03/30/2021] [Accepted: 04/23/2021] [Indexed: 11/19/2022] Open
Abstract
The role of commensal bacterial microbiota in the pathogenesis of human malignancies has been a research field of incomparable progress in recent years. Although breast tissue is commonly assumed to be sterile, recent studies suggest that human breast tissue may contain a bacterial microbiota. In this study, we used an immune‐competent orthotopic breast cancer mouse model to explore the existence of a unique and independent bacterial microbiota in breast tumors. We observed some similarities in breast cancer microbiota with skin; however, breast tumor microbiota was mainly enriched with Gram‐negative bacteria, serving as a primary source of lipopolysaccharide (LPS). In addition, dextran sulfate sodium (DSS) treatment in late‐stage tumor lesions increased LPS levels in the breast tissue environment. We also discovered an increased expression of S100A7 and low level of TLR4 in late‐stage tumors with or without DSS as compared to early‐stage tumor lesions. The treatment of breast cancer cells with LPS increased the expression of S100A7 in breast cancer cells in vitro. Furthermore, S100A7 overexpression downregulated TLR4 and upregulated RAGE expression in breast cancer cells. Analysis of human breast cancer samples also highlighted the inverse correlation between S100A7 and TLR4 expression. Overall, these findings suggest that the commensal microbiota of breast tissue may enhance breast tumor burden through a novel LPS/S100A7/TLR4/RAGE signaling axis.
Collapse
Affiliation(s)
- Tasha Wilkie
- Department of Pathology, The Ohio State University, Wexner Medical Center
| | - Ajeet K Verma
- Department of Pathology, The Ohio State University, Wexner Medical Center
| | - Helong Zhao
- Department of Pathology, The Ohio State University, Wexner Medical Center
| | - Manish Charan
- Department of Pathology, The Ohio State University, Wexner Medical Center
| | - Dinesh K Ahirwar
- Department of Pathology, The Ohio State University, Wexner Medical Center
| | - Sashi Kant
- Department of Pathology, The Ohio State University, Wexner Medical Center
| | - Vijay Pancholi
- Department of Pathology, The Ohio State University, Wexner Medical Center
| | - Sanjay Mishra
- Department of Pathology, The Ohio State University, Wexner Medical Center
| | - Ramesh K Ganju
- Department of Pathology, The Ohio State University, Wexner Medical Center
| |
Collapse
|
19
|
Ceramide kinase regulates TNF-α-induced immune responses in human monocytic cells. Sci Rep 2021; 11:8259. [PMID: 33859296 PMCID: PMC8050074 DOI: 10.1038/s41598-021-87795-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022] Open
Abstract
Ceramide kinase (CERK) phosphorylates ceramide to produce ceramide-1-phosphate (C1P), which is involved in the development of metabolic inflammation. TNF-α modulates inflammatory responses in monocytes associated with various inflammatory disorders; however, the underlying mechanisms remain not fully understood. Here, we investigated the role of CERK in TNF-α-induced inflammatory responses in monocytes. Our results show that disruption of CERK activity in monocytes, either by chemical inhibitor NVP-231 or by small interfering RNA (siRNA), results in the defective expression of inflammatory markers including CD11c, CD11b and HLA-DR in response to TNF-α. Our data show that TNF-α upregulates ceramide phosphorylation. Inhibition of CERK in monocytes significantly reduced the secretion of IL-1β and MCP-1. Similar results were observed in CERK-downregulated cells. TNF-α-induced phosphorylation of JNK, p38 and NF-κB was reduced by inhibition of CERK. Additionally, NF-κB/AP-1 activity was suppressed by the inhibition of CERK. Clinically, obese individuals had higher levels of CERK expression in PBMCs compared to lean individuals, which correlated with their TNF-α levels. Taken together, these results suggest that CERK plays a key role in regulating inflammatory responses in human monocytes during TNF-α stimulation. CERK may be a relevant target for developing novel therapies for chronic inflammatory diseases.
Collapse
|