1
|
O’Brien C, Jones CL. Unraveling lipid metabolism for acute myeloid leukemia therapy. Curr Opin Hematol 2025; 32:77-86. [PMID: 39585293 PMCID: PMC11789610 DOI: 10.1097/moh.0000000000000853] [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] [Indexed: 11/26/2024]
Abstract
PURPOSE OF REVIEW The aim of this review is to highlight the importance of lipids' intricate and interwoven role in mediating diverse acute myeloid leukemia (AML) processes, as well as potentially novel lipid targeting strategies. This review will focus on new studies of lipid metabolism in human leukemia, particularly highlighting work in leukemic stem cells (LSCs), where lipids were assessed directly as a metabolite. RECENT FINDINGS Lipid metabolism is essential to support LSC function and AML survival through diverse mechanisms including supporting energy production, membrane composition, signaling pathways, and ferroptosis. Recent work has highlighted the role of lipid rewiring in metabolic plasticity which can underlie therapy response, the impact of cellular and genetic heterogeneity in AML on lipid metabolism, and the discovery of noncanonical roles of lipid related proteins in AML. SUMMARY Recent findings around lipid metabolism clearly demonstrates their importance to our understanding and therapeutic targeting of AML. We have only begun to unravel the regulation and utilization of lipids in this disease. Further, understanding the layered dynamics of lipid homeostasis could provide novel opportunities to target lipid metabolism in AML and LSCs with the potential of improving outcomes for patients with AML.
Collapse
Affiliation(s)
- Cristiana O’Brien
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Courtney L. Jones
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
2
|
Zhang C, Lv Z, Liang H, Hu F, Bi H. Bioinformatics insights into ACSL1 and ACSL5: prognostic and immune roles in low-grade glioma. BMC Cancer 2025; 25:226. [PMID: 39924494 PMCID: PMC11809057 DOI: 10.1186/s12885-025-13651-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: 05/13/2024] [Accepted: 02/05/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Fatty acid metabolism disruptions affect low-grade gliomas (LGGs), with glioma cells depending on fatty acids for survival. Targeting fatty acid oxidation through the acyl-coenzyme A synthetase long-chain (ACSL) family could alleviate glioma growth and improve prognosis management. However, the integration of ACSLs for analyzing their relationship with LGGs remains unexplored. METHODS We collected RNA expression data of ACSLs for LGGs from TCGA, GTEx, CGGA, and GEO datasets and validated the prognostic significance of gene expression in 37 glioma samples. DNA methylation data from UCSC Xena and promoter methylation levels via MEXPRESS were analyzed. Functional enrichments of co-expressed ACSLs genes were conducted using Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene Set Enrichment Analysis. Protein-protein interaction networks were established via GeneMANIA, and cBioPortal assessed somatic mutations and copy number variations of ACSLs in LGGs. TIMER and TISIDB databases investigated the correlation between ACSLs expression and immune infiltration and checkpoint genes. Hazard ratios (HR) with 95% confidence intervals (95% CI) were computed, and net reclassification index and integrated discrimination improvement were estimated to evaluate the predictive capability of the prognosis model. RESULTS Independent prognostic factors for overall survival included age, gender, tumor grade, MGMT promoter status, ACSL1, ACSL3, ACSL5, and ACSL6 expression levels. High ACSL1 (HR = 2.352, 95%CI: 1.647-3.359, P = 9.00E-06), ACSL3 (HR = 2.367, 95%CI: 1.547-3.624, P = 2.92E-04) and ACSL5 (HR = 2.329, 95%CI: 1.611-3.367, P = 2.80E-05) expression correlated with poor prognosis, while increased ACSL6 (HR = 0.449, 95%CI: 0.290-0.696, P = 1.02E-03) expression related to better survival rates. Furthermore, these associations were also confirmed in the validation datasets and our external cohort. Negative correlation between ACSL1 and ACSL3 gene expression and methylation was found. Functional enrichment analyses highlighted the roles of ACSL1 and ACSL5 in glioma mechanisms and immune function, with significant associations between somatic CNVs and immune cell infiltration. CONCLUSIONS ACSL1 and ACSL5 exhibit prognostic significance in gliomas and influence tumor immunity and immune cell migration, providing valuable insights into LGG prognosis and therapeutic targets.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, Jiangsu, People's Republic of China
| | - Zhonghua Lv
- Department of Neurosurgery, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongsheng Liang
- Department of Neurosurgery, The First Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No. 1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong, China.
| | - Haoran Bi
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
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
|
4
|
Lin J, Lai Y, Lu F, Wang W. Targeting ACSLs to modulate ferroptosis and cancer immunity. Trends Endocrinol Metab 2024:S1043-2760(24)00255-8. [PMID: 39424456 DOI: 10.1016/j.tem.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/13/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024]
Abstract
Five acyl-CoA synthetase long-chain family members (ACSLs) are responsible for catalyzing diverse long-chain fatty acids (LCFAs) into LCFA-acyl-coenzyme A (CoA) for their subsequent metabolism, including fatty acid oxidation (FAO), lipid synthesis, and protein acylation. In this review, we focus on ACSLs and their LCFA substrates and introduce their involvement in regulation of cancer proliferation, metastasis, and therapeutic resistance. Along with the recognition of the decisive role of ACSL4 in ferroptosis - an immunogenic cell death (ICD) initiated by lipid peroxidation - we review the functions of ACSLs on regulating ferroptosis sensitivity. Last, we discuss the current understanding of ACSL on the antitumor immune response. We emphasize the necessity to explore the functions of immune cells expressing ACSLs for developing novel strategies to augment immunotherapy by targeting ACSL.
Collapse
Affiliation(s)
- Junhong Lin
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yongfeng Lai
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Fujia Lu
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| | - Weimin Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China; Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Han H, Liu J, Zhu S, Zhao T. Identification of two key biomarkers CD93 and FGL2 associated with survival of acute myeloid leukaemia by weighted gene co-expression network analysis. J Cell Mol Med 2024; 28:e18552. [PMID: 39054581 PMCID: PMC11272607 DOI: 10.1111/jcmm.18552] [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: 03/18/2024] [Revised: 06/17/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a biologically heterogeneous haematological malignancy. This study was performed to identify the potential biomarkers for the prognosis and treatment of AML. We applied weighted gene co-expression network analysis to identify key modules and hub genes related to the prognosis of AML using data from The Cancer Genome Atlas (TCGA). In total, 1581 differentially expressed genes (1096 upregulated and 485 downregulated) were identified between AML patients and healthy controls, with the blue module being the most significant among 14 modules associated with AML morphology. Through functional enrichment analysis, we identified 217 genes in the blue module significantly enriched in 'neutrophil degranulation' and 'neutrophil activation involved in immune response' pathways. The survival analysis revealed six genes (S100A9, S100A8, HK3, CD93, CXCR2 and FGL2) located in the significantly enriched pathway that were notably related to AML survival. We validated the expression of these six genes at gene and single-cell levels and identified methylation loci of each gene, except for S100A8. Finally, in vitro experiments were performed to demonstrate whether the identified hub genes were associated with AML survival. After knockdown of CD93 and FGL2, cell proliferation was significantly reduced in U937 cell line over 5 days. In summary, we identified CD93 and FGL2 as key hub genes related to AML survival, with FGL2 being a novel biomarker for the prognosis and treatment of AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Gene Regulatory Networks
- Biomarkers, Tumor/genetics
- Prognosis
- Receptors, Complement/genetics
- Receptors, Complement/metabolism
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Expression Regulation, Leukemic
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Hepatitis A Virus Cellular Receptor 2/genetics
- Hepatitis A Virus Cellular Receptor 2/metabolism
- Gene Expression Profiling
- Cell Line, Tumor
- DNA Methylation/genetics
- Survival Analysis
- Fibrinogen
Collapse
Affiliation(s)
- Haijun Han
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of Medicine, Hangzhou City UniversityHangzhouChina
| | - Jie Liu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of Medicine, Hangzhou City UniversityHangzhouChina
- College of Life Sciences, Zhejiang Normal UniversityJinhuaChina
| | - Shengyu Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of Medicine, Hangzhou City UniversityHangzhouChina
| | - Tiejun Zhao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang ProvinceSchool of Medicine, Hangzhou City UniversityHangzhouChina
- College of Life Sciences, Zhejiang Normal UniversityJinhuaChina
| |
Collapse
|
6
|
Lai Y, Gao Y, Lin J, Liu F, Yang L, Zhou J, Xue Y, Li Y, Chang Z, Li J, Chao T, Chen J, Cheng X, Gao X, Li X, Lu F, Chu Q, Wang W. Dietary elaidic acid boosts tumoral antigen presentation and cancer immunity via ACSL5. Cell Metab 2024; 36:822-838.e8. [PMID: 38350448 DOI: 10.1016/j.cmet.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/12/2023] [Accepted: 01/20/2024] [Indexed: 02/15/2024]
Abstract
Immunomodulatory effects of long-chain fatty acids (LCFAs) and their activating enzyme, acyl-coenzyme A (CoA) synthetase long-chain family (ACSL), in the tumor microenvironment remain largely unknown. Here, we find that ACSL5 functions as an immune-dependent tumor suppressor. ACSL5 expression sensitizes tumors to PD-1 blockade therapy in vivo and the cytotoxicity mediated by CD8+ T cells in vitro via regulation of major histocompatibility complex class I (MHC-I)-mediated antigen presentation. Through screening potential substrates for ACSL5, we further identify that elaidic acid (EA), a trans LCFA that has long been considered harmful to human health, phenocopies to enhance MHC-I expression. EA supplementation can suppress tumor growth and sensitize PD-1 blockade therapy. Clinically, ACSL5 expression is positively associated with improved survival in patients with lung cancer, and plasma EA level is also predictive for immunotherapy efficiency. Our findings provide a foundation for enhancing immunotherapy through either targeting ACSL5 or metabolic reprogramming of antigen presentation via dietary EA supplementation.
Collapse
Affiliation(s)
- Yongfeng Lai
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junhong Lin
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Fangfang Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liguo Yang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Xue
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenzhen Chang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianfu Gao
- Shanghai ProfLeader Biotech Co., Ltd, Shanghai, China
| | - Xiong Li
- Department of Gynecology & Obstetrics, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fujia Lu
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Weimin Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China; Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
7
|
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
|
8
|
Zhang C, Zhang Y, Dong Y, Zi R, Wang Y, Chen Y, Liu C, Wang J, Wang X, Li J, Liang H, Ou J. Non-alcoholic fatty liver disease promotes liver metastasis of colorectal cancer via fatty acid synthase dependent EGFR palmitoylation. Cell Death Discov 2024; 10:41. [PMID: 38263401 PMCID: PMC10805926 DOI: 10.1038/s41420-023-01770-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024] Open
Abstract
Liver metastasis is the major reason for most of colorectal cancer (CRC) related deaths. Accumulating evidence indicates that CRC patients with non-alcoholic fatty liver disease (NAFLD) are at a greater risk of developing liver metastasis. With the growing prevalence of NAFLD, a better understanding of the molecular mechanism in NAFLD-driven CRC liver metastasis is needed. In this study, we demonstrated that NAFLD facilitated CRC liver metastasis as a metabolic disorder and promoted the stemness of metastatic CRC cells for their colonization and outgrowth in hepatic niches. Metabolically, the lipid-rich microenvironment in NAFLD activated de novo palmitate biosynthesis in metastatic CRC cells via upregulating fatty acid synthase (FASN). Moreover, increased intracellular palmitate bioavailability promoted EGFR palmitoylation to enhance its protein stability and plasma membrane localization. Furthermore, we demonstrated that the FDA-approved FASN inhibitor orlistat could reduce NAFLD-activated endogenous palmitate production, thus inhibiting palmitoylation of EGFR to suppress CRC cell stemness and restrict liver metastasis in synergy with conventional chemotherapy. These findings reveal that the NAFLD metabolic microenvironment boosts endogenous palmitate biosynthesis in metastatic CRC cells and promotes cell stemness via EGFR palmitoylation, and FASN inhibitor orlistat could be a candidate adjuvant drug to suppress liver metastasis in CRC patients with NAFLD.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yue Zhang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yan Dong
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ruiyang Zi
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yijie Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yanrong Chen
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chengxiang Liu
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Junyi Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Xuesong Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| | - Juanjuan Ou
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
- Jinfeng Laboratory, 401329, Chongqing, China.
| |
Collapse
|