1
|
Sentjens K, Pillai R, Joseph JW. The effects of free fatty acid-free bovine serum albumin and palmitate on pancreatic β-cell function. Islets 2025; 17:2479911. [PMID: 40091018 PMCID: PMC11917175 DOI: 10.1080/19382014.2025.2479911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Pancreatic β-cells release insulin in response to fluctuations in plasma glucose, amino acids, and free fatty acids (FFA). Clonal cell lines and isolated islets serve as essential early models for studying the impact of nutrients and evaluating potential therapies to address β-cell dysfunction. Acute and chronic changes in FFA levels have been shown to have positive and negative effects on β-cell function both in vivo and in vitro. A key problem in comparing islet lipid studies from different laboratories is that a wide variety of methods are used to isolate, culture, and assess islet function. The current study compares bovine serum albumin (BSA) types and lipid preparation methods in clonal 832/13 cells and human islets. Changing the percentage and culture conditions when using FFA-free BSA can negatively affect β-cell function compared to regular BSA. Preparing palmitate with FFA-free BSA can rescue insulin secretion compared to treating cells alone with FFA-free BSA. Different methods of preparing palmitate can have unique effects on insulin secretion. Overall, interpreting the effects of lipids on β-cell function is complicated by a number of variables that need to be controlled for in islet experiments.
Collapse
Affiliation(s)
| | - Renjitha Pillai
- School of Pharmacy, University of Waterloo, Kitchener, ON, Canada
| | - Jamie W. Joseph
- School of Pharmacy, University of Waterloo, Kitchener, ON, Canada
| |
Collapse
|
2
|
Das RK, Kuzmin AN, Pliss A, Mahajan SD, Shukla S, Prasad PN. Fentanyl-Induced Transformations in Composition of Lipid Droplets in Central Nervous System Cells revealed by Ramanomics. J Lipid Res 2025:100827. [PMID: 40398506 DOI: 10.1016/j.jlr.2025.100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/16/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025] Open
Abstract
Quantitative characterization of the transformations of subcellular molecular environment in response to fentanyl exposure in human microglia and astrocytes is warranted to provide insight into the regulation of neuroinflammatory responses and neural survival in the scenario of opiate drug addiction. Cytoplasmic lipid droplets (LD) act as depot for exogeneous hydrophobic molecules, such as fentanyl, which can lead to increased drug accumulation and alteration of their metabolism. In the present work, we have used an emerging Ramanomics technique that combines quantitative micro-Raman spectrometry with biomolecular component analysis to unravel fentanyl induced changes in concentrations of phospholipids, sterols, glycogen, sphingomyelin, phosphocholine as well as RNA and proteins, in the LDs of microglia and astrocytes. To our knowledge, this study represents, the first report on the effect of fentanyl overdose in the biomolecular composition of distinct organelles in single live cells, towards therapeutics advances to treat fentanyl addiction and development of diagnostic assays of illicit opiate abuse.
Collapse
Affiliation(s)
- Rahul K Das
- Institute for Lasers, Photonics and Biophotonics and Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Andrey N Kuzmin
- Institute for Lasers, Photonics and Biophotonics and Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States.
| | - Artem Pliss
- Institute for Lasers, Photonics and Biophotonics and Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States; School of Pharmacy, D'Youville University, Buffalo, New York 14201, United States
| | - Supriya D Mahajan
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, Buffalo, New York 14203, United States
| | - Shobha Shukla
- Nanostructures Engineering and Modeling Laboratory, Department of Metallurgical Engineering and Materials Science, Indian Institute of Technology Bombay, Mumbai 400076, MH, India
| | - Paras N Prasad
- Institute for Lasers, Photonics and Biophotonics and Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States.
| |
Collapse
|
3
|
Nakagawa H. Lipogenesis and MASLD: re-thinking the role of SREBPs. Arch Toxicol 2025:10.1007/s00204-025-04052-w. [PMID: 40327083 DOI: 10.1007/s00204-025-04052-w] [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: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 05/07/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide and a major risk factor for hepatocellular carcinoma (HCC). Lipid metabolism, particularly de novo lipogenesis (DNL) regulated by sterol regulatory element-binding proteins (SREBPs), plays a key role in MASLD progression. While excessive SREBP activation contributes to hepatic steatosis, our recent findings indicate that strong SREBP inhibition paradoxically exacerbates liver injury and accelerates carcinogenesis in murine MASLD models. Mechanistically, SREBP dysfunction disrupts phospholipid homeostasis, leading to impaired endoplasmic reticulum (ER) membrane fluidity, ER stress, and hepatocyte injury. Transcriptomic analysis of clinical samples revealed a dynamic shift in SREBP activity, with upregulation in early MASLD but significant downregulation in advanced, burned-out MASH. This suggests that SREBP dysfunction in advanced disease may contribute to fibrosis progression and increased HCC risk. Given these findings, therapeutic strategies targeting lipid metabolism in MASLD must be carefully tailored to disease stage. This review provides an updated perspective on the biphasic role of SREBP in MASLD, emphasizing the need to re-think lipid metabolism-targeted therapies and develop personalized interventions to mitigate disease progression and HCC development.
Collapse
Affiliation(s)
- Hayato Nakagawa
- Department of Gastroenterology and Hepatology, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
4
|
Engelfriet ML, Guo Y, Arnold A, Valen E, Ciosk R. Reprograming gene expression in 'hibernating' C. elegans involves the IRE-1/XBP-1 pathway. eLife 2025; 13:RP101186. [PMID: 40326887 PMCID: PMC12055002 DOI: 10.7554/elife.101186] [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] [Indexed: 05/07/2025] Open
Abstract
In nature, many animals respond to cold by entering hibernation, while in clinical settings, controlled cooling is used in transplantation and emergency medicine. However, the molecular mechanisms that enable cells to survive severe cold are still not fully understood. One key aspect of cold adaptation is the global downregulation of protein synthesis. Studying it in the nematode Caenorhabditis elegans, we find that the translation of most mRNAs continues in the cold, albeit at a slower rate, and propose that cold-specific gene expression is regulated primarily at the transcription level. Supporting this idea, we found that the transcription of certain cold-induced genes is linked to the activation of unfolded protein response (UPR) through the conserved IRE-1/XBP-1 signaling pathway. Our findings suggest that this pathway is triggered by cold-induced perturbations in proteins and lipids within the endoplasmic reticulum, and that its activation is beneficial for cold survival.
Collapse
Affiliation(s)
- Melanie Lianne Engelfriet
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of OsloOsloNorway
| | - Yanwu Guo
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of OsloOsloNorway
| | - Andreas Arnold
- Division of Molecular Neuroscience, Department of Biomedicine, University of BaselBaselSwitzerland
- University Psychiatric Clinics, University of BaselBaselSwitzerland
| | - Eivind Valen
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of OsloOsloNorway
| | - Rafal Ciosk
- Section for Biochemistry and Molecular Biology, Department of Biosciences, University of OsloOsloNorway
| |
Collapse
|
5
|
Fang X, Li J, Pang H, Zheng H, Shi X, Feng L, Hu K, Zhou T. Xingxiao pills suppresses lung adenocarcinoma progression by modulating lipid metabolism and inhibiting the PLA2G4A-GLI1-SOX2 Axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156826. [PMID: 40339555 DOI: 10.1016/j.phymed.2025.156826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/20/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) remains a leading cause of cancer mortality due to resistance, metastasis, and recurrence. Unlike conventional cytotoxic therapies, Xingxiao Pills (XXP), a classic traditional Chinese medicine formula, offers a complementary approach to treating LUAD, while its non-cytotoxic anti-cancer mechanisms remain unclear. PURPOSE To investigate the effect and mechanism of XXP on LUAD progression and stemness via lipid metabolism regulation. METHOD UHPLC-MS/MS was used to analyze the chemical constituents of XXP. The effects of XXP on LUAD cell proliferation, migration, invasion, and stemness were evaluated using CCK-8, Transwell, and tumor sphere assays. A LUAD xenograft model confirmed XXP's anti-tumor effects. Transcriptomics, metabolomics, ELISA, qRT-PCR, and Western blot were used to investigate the underlying mechanisms. Kaplan-Meier (KM) survival analysis and stemness index scores were performed for LUAD patients based on the TCGA dataset. Statistical analyses were performed using Student's t-test, ANOVA, and KM survival analysis (p< 0.05 considered significant). RESULTS XXP inhibits LUAD progression in mouse and cell models by targeting lipid metabolism reprogramming. It suppresses FA synthesis, elongation, oxidation, and glycerophospholipid (GPL) metabolism while upregulating arachidonic acid (AA) metabolism. Mechanistic studies revealed that XXP attenuates tumor stemness by inhibiting PLA2G4A (cPLA2), lowering AA release, and disrupting SMO/GLI1/SOX2 signaling, an effect also observed with the cPLA2 inhibitor AACOCF3. KM analysis showed that higher PLA2G4A expression correlated with a worse 5-year prognosis in LUAD (p = 0.0047). The low GPL/high AA group (consistent with XXP's metabolic pattern) had better survival (p = 0.0028) and a lower stemness index (p< 0.0001) than the high GPL/low AA unrelated group. CONCLUSION Xingxiao Pill modulates GPL and AA metabolism and downregulates the PLA2G4A (cPLA2)-AA/SMO/GLI1/SOX2 axis. Through this mechanism, XXP effectively inhibits tumor growth and stemness by targeting lipid metabolism.
Collapse
Affiliation(s)
- Xueni Fang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - JingHua Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - HaoYue Pang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Zheng
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiang Shi
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Kaiwen Hu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Tian Zhou
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
6
|
Zarrella S, Miranda MR, Covelli V, Restivo I, Novi S, Pepe G, Tesoriere L, Rodriquez M, Bertamino A, Campiglia P, Tecce MF, Vestuto V. Endoplasmic Reticulum Stress and Its Role in Metabolic Reprogramming of Cancer. Metabolites 2025; 15:221. [PMID: 40278350 PMCID: PMC12029571 DOI: 10.3390/metabo15040221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Endoplasmic reticulum (ER) stress occurs when ER homeostasis is disrupted, leading to the accumulation of misfolded or unfolded proteins. This condition activates the unfolded protein response (UPR), which aims to restore balance or trigger cell death if homeostasis cannot be achieved. In cancer, ER stress plays a key role due to the heightened metabolic demands of tumor cells. This review explores how metabolomics can provide insights into ER stress-related metabolic alterations and their implications for cancer therapy. Methods: A comprehensive literature review was conducted to analyze recent findings on ER stress, metabolomics, and cancer metabolism. Studies examining metabolic profiling of cancer cells under ER stress conditions were selected, with a focus on identifying potential biomarkers and therapeutic targets. Results: Metabolomic studies highlight significant shifts in lipid metabolism, protein synthesis, and oxidative stress management in response to ER stress. These metabolic alterations are crucial for tumor adaptation and survival. Additionally, targeting ER stress-related metabolic pathways has shown potential in preclinical models, suggesting new therapeutic strategies. Conclusions: Understanding the metabolic impact of ER stress in cancer provides valuable opportunities for drug development. Metabolomics-based approaches may help identify novel biomarkers and therapeutic targets, enhancing the effectiveness of antitumor therapies.
Collapse
Affiliation(s)
- Salvatore Zarrella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Verdiana Covelli
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Manuela Rodriquez
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| |
Collapse
|
7
|
Ohba Y, Motohashi M, Arita M. Characterization of UGT8 as a monogalactosyl diacylglycerol synthase in mammals. J Biochem 2025; 177:141-152. [PMID: 39658193 PMCID: PMC11795506 DOI: 10.1093/jb/mvae084] [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: 09/29/2024] [Revised: 11/18/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024] Open
Abstract
Monogalactosyl diacylglycerol (MGDG) is a major membrane lipid component in plants and is crucial for proper thylakoid functioning. However, MGDG in mammals has not received much attention, partly because of its relative scarcity in mammalian tissues. In addition, the biosynthetic pathway of MGDG in mammals has not been thoroughly analysed, although some reports have suggested that UGT8, a ceramide galactosyltransferase, has the potential to catalyse MGDG biosynthesis. Here, we successfully captured the endogenous levels of MGDG in HeLa cells using liquid chromatography quadrupole time-of-flight mass spectrometry (LC-QTOF-MS)-based lipidomics. Cellular MGDG was completely depleted in CRISPR/Cas9-mediated UGT8 knockout (KO) HeLa cells. Transient overexpression of UGT8 enhanced MGDG production in HeLa cells, and the corresponding cell lysates displayed MGDG biosynthetic activity in vitro. Site-directed mutagenesis revealed that His358 within the UGT signature sequence was important for its activity. UGT8 was localized in the endoplasmic reticulum and activation of the unfolded protein response by membrane lipid saturation was impaired in UGT8 KO cells. These results demonstrate that UGT8 is an MGDG synthase in mammals and that UGT8 regulates membrane lipid saturation signals in cells.
Collapse
Affiliation(s)
- Yohsuke Ohba
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Mizuki Motohashi
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Makoto Arita
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
8
|
Bispo P, Rodrigues PO, Bandarra NM. Dietary Oleic Acid and SCD16 and ELOVL6 Estimated Activities Can Modify Erythrocyte Membrane n-3 and n-6 HUFA Partition: A Pilot Study. Curr Issues Mol Biol 2025; 47:81. [PMID: 39996802 PMCID: PMC11854775 DOI: 10.3390/cimb47020081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
In this work, we studied the relationships between the most representative fatty acids (FAs) and their ratios in red blood cell (RBC) membranes and dietary fatty acids alongside several cardiometabolic risk factors. Twenty-six individuals were enrolled with a mean age of 50.4 ± 12.7 years (16 males and 10 females). By bivariate analysis, dietary oleic acid (OA) correlated negatively with C20:4n-6 (AA) (p = 0.031) in RBCs. With multivariate regression analysis, dietary OA (p < 0.001) is an independent predictor and negatively associated with AA levels in RBCs, while the elongation of very-long-chain fatty acids 6 (ELOVL6) and stearoyl-CoA desaturase 16 (SCD16) activities (p < 0.05) was positively associated with AA levels in RBCs. The multivariate regression models also showed that dietary OA was an independent predictor and positively associated with C22:5n-3 (DPA) in RBCs. Furthermore, BMI positively correlated with SCD16, and both SCD16 and SCD18 were positively associated with triacylglycerols levels. In addition, SCD16 positively and significantly correlated with LDL-c and the LDL-c/HDL-c ratio and negatively correlated with the ApoA1/ApoB ratio, and SCD16 and ELOVL6 were significantly associated with HDL molecular subfractions. Therefore, our data underline that OA, SCD16 and ELOVL6 can interfere with n-3 and n-6 partition in biomembranes such as RBCs, suggesting an important molecular (patho)physiological regulatory mechanism role in controlling bioactive molecules' availability such as those involved in the immune-inflammatory response.
Collapse
Affiliation(s)
- Paulo Bispo
- Department of Food Tecnhology, Biotecnology and Nutrition, Escola Superior Agrária, Instituto Politécnico de Santarém, 2001-904 Santarém, Portugal
| | - Pedro O. Rodrigues
- Department of Biochemistry and CEDOC, Faculty of Medical Sciences/NOVA Medical School, Universidade Nova de Lisboa (UNL), 1169-056 Lisboa, Portugal;
| | - Narcisa M. Bandarra
- IPMA, IP, Departamento do Mar e dos Recursos Marinhos, Divisão de Aquacultura, Valorização e Biosprospeção, 1495-165 Lisboa, Portugal;
- CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros de Leixões, 4450-208 Matosinhos, Portugal
| |
Collapse
|
9
|
Białek W, Hryniewicz-Jankowska A, Czechowicz P, Sławski J, Collawn JF, Czogalla A, Bartoszewski R. The lipid side of unfolded protein response. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159515. [PMID: 38844203 DOI: 10.1016/j.bbalip.2024.159515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/16/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Although our current knowledge of the molecular crosstalk between the ER stress, the unfolded protein response (UPR), and lipid homeostasis remains limited, there is increasing evidence that dysregulation of either protein or lipid homeostasis profoundly affects the other. Most research regarding UPR signaling in human diseases has focused on the causes and consequences of disrupted protein folding. The UPR itself consists of very complex pathways that function to not only maintain protein homeostasis, but just as importantly, modulate lipid biogenesis to allow the ER to adjust and promote cell survival. Lipid dysregulation is known to activate many aspects of the UPR, but the complexity of this crosstalk remains a major research barrier. ER lipid disequilibrium and lipotoxicity are known to be important contributors to numerous human pathologies, including insulin resistance, liver disease, cardiovascular diseases, neurodegenerative diseases, and cancer. Despite their medical significance and continuous research, however, the molecular mechanisms that modulate lipid synthesis during ER stress conditions, and their impact on cell fate decisions, remain poorly understood. Here we summarize the current view on crosstalk and connections between altered lipid metabolism, ER stress, and the UPR.
Collapse
Affiliation(s)
- Wojciech Białek
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | - Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Rafał Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.
| |
Collapse
|
10
|
Igal RA. Death and the desaturase: Implication of Stearoyl-CoA desaturase-1 in the mechanisms of cell stress, apoptosis, and ferroptosis. Biochimie 2024; 225:156-167. [PMID: 38823621 DOI: 10.1016/j.biochi.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/05/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Growth and proliferation of normal and cancerous cells necessitate a finely-tuned regulation of lipid metabolic pathways to ensure the timely supply of structural, energetic, and signaling lipid molecules. The synthesis and remodeling of lipids containing fatty acids with an appropriate carbon length and insaturation level are required for supporting each phase of the mechanisms of cell replication and survival. Mammalian Stearoyl-CoA desaturases (SCD), particularly SCD1, play a crucial role in modulating the fatty acid composition of cellular lipids, converting saturated fatty acids (SFA) into monounsaturated fatty acids (MUFA) in the endoplasmic reticulum (ER). Extensive research has elucidated in great detail the participation of SCD1 in the molecular mechanisms that govern cell replication in normal and cancer cells. More recently, investigations have shed new light on the functional and regulatory role of the Δ9-desaturase in the processes of cell stress and cell death. This review will examine the latest findings on the involvement of SCD1 in the molecular pathways of cell survival, particularly on the mechanisms of ER stress and autophagy, as well in apoptotic and non-apoptotic cell death.
Collapse
Affiliation(s)
- R Ariel Igal
- Institute of Human Nutrition and Department of Pediatrics, Columbia University Irving Medical Center, New York City, New York, USA.
| |
Collapse
|
11
|
Rajakumar T, Hossain MA, Stopka SA, Micoogullari Y, Ang J, Agar NYR, Hanna J. Dysregulation of ceramide metabolism causes phytoceramide-dependent induction of the unfolded protein response. Mol Biol Cell 2024; 35:ar117. [PMID: 39024283 PMCID: PMC11449394 DOI: 10.1091/mbc.e24-03-0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/25/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
The unfolded protein response (UPR) detects and mitigates the harmful effects of dysregulated endoplasmic reticulum (ER) function. The UPR has been best characterized as a protein quality control response, and the sole UPR sensor in yeast, Ire1, is known to detect misfolded ER proteins. However, recent work suggests the UPR can also sense diverse defects within the ER membrane, including increased fatty acid saturation and altered phospholipid abundance. These and other lipid-related stimuli have been referred to as lipid bilayer stress and may be sensed independently through Ire1's transmembrane domain. Here, we show that the loss of Isc1, a phospholipase that catabolizes complex ceramides, causes UPR induction, even in the absence of exogenous stress. A series of chemical and genetic approaches identified a requirement for very long-chain fatty acid (VLCFA)-containing phytoceramides for UPR induction. In parallel, comprehensive lipidomics analyses identified large increases in the abundance of specific VLCFA-containing phytoceramides in the isc1Δ mutant. We failed to identify evidence of an accompanying defect in protein quality control or ER-associated protein degradation. These results extend our understanding of lipid bilayer stress in the UPR and provide a foundation for mechanistic investigation of this fascinating intersection between ceramide metabolism, membrane homeostasis, and the UPR.
Collapse
Affiliation(s)
- Tamayanthi Rajakumar
- Department of Pathology, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115
| | - Md Amin Hossain
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115
| | - Sylwia A. Stopka
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115
| | - Yagmur Micoogullari
- Department of Pathology, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115
| | - Jessie Ang
- Department of Pathology, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115
| | - Nathalie Y. R. Agar
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115
| | - John Hanna
- Department of Pathology, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115
| |
Collapse
|
12
|
Ernst R, Renne MF, Jain A, von der Malsburg A. Endoplasmic Reticulum Membrane Homeostasis and the Unfolded Protein Response. Cold Spring Harb Perspect Biol 2024; 16:a041400. [PMID: 38253414 PMCID: PMC11293554 DOI: 10.1101/cshperspect.a041400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The endoplasmic reticulum (ER) is the key organelle for membrane biogenesis. Most lipids are synthesized in the ER, and most membrane proteins are first inserted into the ER membrane before they are transported to their target organelle. The composition and properties of the ER membrane must be carefully controlled to provide a suitable environment for the insertion and folding of membrane proteins. The unfolded protein response (UPR) is a powerful signaling pathway that balances protein and lipid production in the ER. Here, we summarize our current knowledge of how aberrant compositions of the ER membrane, referred to as lipid bilayer stress, trigger the UPR.
Collapse
Affiliation(s)
- Robert Ernst
- Medical Biochemistry and Molecular Biology, Medical Faculty, Saarland University, 66421 Homburg, Germany
- Preclinical Center for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Mike F Renne
- Medical Biochemistry and Molecular Biology, Medical Faculty, Saarland University, 66421 Homburg, Germany
- Preclinical Center for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Aamna Jain
- Medical Biochemistry and Molecular Biology, Medical Faculty, Saarland University, 66421 Homburg, Germany
- Preclinical Center for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Alexander von der Malsburg
- Medical Biochemistry and Molecular Biology, Medical Faculty, Saarland University, 66421 Homburg, Germany
- Preclinical Center for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
13
|
Pelletier AN, Sanchez GP, Izmirly A, Watson M, Di Pucchio T, Carvalho KI, Filali-Mouhim A, Paramithiotis E, Timenetsky MDCST, Precioso AR, Kalil J, Diamond MS, Haddad EK, Kallas EG, Sekaly RP. A pre-vaccination immune metabolic interplay determines the protective antibody response to a dengue virus vaccine. Cell Rep 2024; 43:114370. [PMID: 38900640 PMCID: PMC11404042 DOI: 10.1016/j.celrep.2024.114370] [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: 07/14/2023] [Revised: 02/05/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024] Open
Abstract
Protective immunity to dengue virus (DENV) requires antibody response to all four serotypes. Systems vaccinology identifies a multi-OMICs pre-vaccination signature and mechanisms predictive of broad antibody responses after immunization with a tetravalent live attenuated DENV vaccine candidate (Butantan-DV/TV003). Anti-inflammatory pathways, including TGF-β signaling expressed by CD68low monocytes, and the metabolites phosphatidylcholine (PC) and phosphatidylethanolamine (PE) positively correlate with broadly neutralizing antibody responses against DENV. In contrast, expression of pro-inflammatory pathways and cytokines (IFN and IL-1) in CD68hi monocytes and primary and secondary bile acids negatively correlates with broad DENV-specific antibody responses. Induction of TGF-β and IFNs is done respectively by PC/PE and bile acids in CD68low and CD68hi monocytes. The inhibition of viral sensing by PC/PE-induced TGF-β is confirmed in vitro. Our studies show that the balance between metabolites and the pro- or anti-inflammatory state of innate immune cells drives broad and protective B cell response to a live attenuated dengue vaccine.
Collapse
Affiliation(s)
- Adam-Nicolas Pelletier
- RPM Bioinfo Solutions, Sainte-Thérèse, QC, Canada; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Gabriela Pacheco Sanchez
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Abdullah Izmirly
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Tiziana Di Pucchio
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Karina Inacio Carvalho
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Abdelali Filali-Mouhim
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | | | | | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Institute for Investigation in Immunology-Instituto Nacional de Ciência e Tecnologia-iii-INCT, São Paulo, SP, Brazil
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elias K Haddad
- Department of Medicine and Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Esper G Kallas
- Instituto Butantan, São Paulo, Brazil; Department of Infectious and Parasitic Diseases, Hospital das Clínicas, School of Medicine, University of Sao Paulo, São Paulo 01246-903, Brazil
| | - Rafick Pierre Sekaly
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
Kettel P, Karagöz GE. Endoplasmic reticulum: Monitoring and maintaining protein and membrane homeostasis in the endoplasmic reticulum by the unfolded protein response. Int J Biochem Cell Biol 2024; 172:106598. [PMID: 38768891 DOI: 10.1016/j.biocel.2024.106598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/01/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
The endoplasmic reticulum (ER) regulates essential cellular processes, including protein folding, lipid synthesis, and calcium homeostasis. The ER homeostasis is maintained by a conserved set of signaling cascades called the Unfolded Protein Response (UPR). How the UPR senses perturbations in ER homeostasis has been the subject of active research for decades. In metazoans, the UPR consists of three ER-membrane embedded sensors: IRE1, PERK and ATF6. These sensors detect the accumulation of misfolded proteins in the ER lumen and adjust protein folding capacity according to cellular needs. Early work revealed that the ER-resident chaperone BiP binds to all three UPR sensors in higher eukaryotes and BiP binding was suggested to regulate their activity. More recent data have shown that in higher eukaryotes the interaction of the UPR sensors with a complex network of chaperones and misfolded proteins modulates their activation and deactivation dynamics. Furthermore, emerging evidence suggests that the UPR monitors ER membrane integrity beyond protein folding defects. However, the mechanistic and structural basis of UPR activation by proteotoxic and lipid bilayer stress in higher eukaryotes remains only partially understood. Here, we review the current understanding of novel protein interaction networks and the contribution of the lipid membrane environment to UPR activation.
Collapse
Affiliation(s)
- Paulina Kettel
- Max Perutz Laboratories Vienna, Vienna BioCenter, Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - G Elif Karagöz
- Max Perutz Laboratories Vienna, Vienna BioCenter, Vienna, Austria; Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
15
|
Ikeda T, Katoh Y, Hino H, Seta D, Ogawa T, Iwata T, Nishio H, Sugawara M, Hirai S. FADS2 confers SCD1 inhibition resistance to cancer cells by modulating the ER stress response. Sci Rep 2024; 14:13116. [PMID: 38849435 PMCID: PMC11161504 DOI: 10.1038/s41598-024-64043-2] [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: 04/05/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Stearoyl-CoA desaturase 1 (SCD1) is an attractive target for cancer therapy. However, the clinical efficacy of SCD1 inhibitor monotherapy is limited. There is thus a need to elucidate the mechanisms of resistance to SCD1 inhibition and develop new therapeutic strategies for combination therapy. In this study, we investigated the molecular mechanisms by which cancer cells acquire resistance to endoplasmic reticulum (ER) stress-dependent cancer cell death induced by SCD1 inhibition. SCD1 inhibitor-sensitive and -resistant cancer cells were treated with SCD1 inhibitors in vitro, and SCD1 inhibitor-sensitive cancer cells accumulated palmitic acid and underwent ER stress response-induced cell death. Conversely, SCD1-resistant cancer cells did not undergo ER stress response-induced cell death because fatty acid desaturase 2 (FADS2) eliminated the accumulation of palmitic acid. Furthermore, genetic depletion using siRNA showed that FADS2 is a key determinant of sensitivity/resistance of cancer cells to SCD1 inhibitor. A549 cells, an SCD1 inhibitor-resistant cancer cell line, underwent ER stress-dependent cancer cell death upon dual inhibition of SCD1 and FADS2. Thus, combination therapy with SCD1 inhibition and FADS2 inhibition is potentially a new cancer therapeutic strategy targeting fatty acid metabolism.
Collapse
Affiliation(s)
- Toshikatsu Ikeda
- Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, 30-1 Ohyaguchi-Kami-Cho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Yuki Katoh
- Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, 30-1 Ohyaguchi-Kami-Cho, Itabashi-ku, Tokyo, 173-8610, Japan.
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Hirotsugu Hino
- Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, 30-1 Ohyaguchi-Kami-Cho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Daichi Seta
- Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, 30-1 Ohyaguchi-Kami-Cho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Tadashi Ogawa
- Department of Legal Medicine, Aichi Medical University School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Takashi Iwata
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroshi Nishio
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masaki Sugawara
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shuichi Hirai
- Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, 30-1 Ohyaguchi-Kami-Cho, Itabashi-ku, Tokyo, 173-8610, Japan
| |
Collapse
|
16
|
Cavaleri D, Riboldi I, Crocamo C, Paglia G, Carrà G, Bartoli F. Evidence from preclinical and clinical metabolomics studies on the antidepressant effects of ketamine and esketamine. Neurosci Lett 2024; 831:137791. [PMID: 38670523 DOI: 10.1016/j.neulet.2024.137791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
The antidepressant effects of ketamine and esketamine are well-documented. Nonetheless, most of the underlying molecular mechanisms have to be uncovered yet. In the last decade, metabolomics has emerged as a useful means to investigate the metabolic phenotype associated with depression as well as changes induced by antidepressant treatments. This mini-review aims at summarizing the main findings from preclinical and clinical studies that used metabolomics to investigate the metabolic effects of subanesthetic, antidepressant doses of ketamine and esketamine and their relationship with clinical response. Both animal and human studies report alterations in several metabolic pathways - including the tricarboxylic acid cycle, glycolysis, the pentose phosphate pathway, lipid metabolism, amino acid metabolism, the kynurenine pathway, and the urea cycle - following the administration of ketamine or its enantiomers. Although more research is needed to clarify commonalities and differences in molecular mechanisms of action between the racemic compound and its enantiomers, these findings comprehensively support an influence of ketamine and esketamine on mitochondrial and cellular energy production, membrane homeostasis, neurotransmission, and signaling. Metabolomics may thus represent a promising strategy to clarify molecular mechanisms underlying treatment-resistant depression and related markers of clinical response to ketamine and esketamine. This body of preclinical and clinical evidence, if further substantiated, has the potential to guide clinicians towards personalized approaches, contributing to new paradigms in the clinical management of depression.
Collapse
Affiliation(s)
- Daniele Cavaleri
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Ilaria Riboldi
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Cristina Crocamo
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Giuseppe Paglia
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Giuseppe Carrà
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy; Division of Psychiatry, University College London, 149 Tottenham Ct Rd, London W1T 7NF, United Kingdom
| | - Francesco Bartoli
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy.
| |
Collapse
|
17
|
Yuan Y, Xu J, Jiang Q, Yang C, Wang N, Liu X, Piao HL, Lu S, Zhang X, Han L, Liu Z, Cai J, Liu F, Chen S, Liu J. Ficolin 3 promotes ferroptosis in HCC by downregulating IR/SREBP axis-mediated MUFA synthesis. J Exp Clin Cancer Res 2024; 43:133. [PMID: 38698462 PMCID: PMC11067213 DOI: 10.1186/s13046-024-03047-2] [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: 11/30/2023] [Accepted: 04/15/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Targeting ferroptosis has been identified as a promising approach for the development of cancer therapies. Monounsaturated fatty acid (MUFA) is a type of lipid that plays a crucial role in inhibiting ferroptosis. Ficolin 3 (FCN3) is a component of the complement system, serving as a recognition molecule against pathogens in the lectin pathway. Recent studies have reported that FCN3 demonstrates inhibitory effects on the progression of certain tumors. However, whether FCN3 can modulate lipid metabolism and ferroptosis remains largely unknown. METHODS Cell viability, BODIPY-C11 staining, and MDA assay were carried out to detect ferroptosis. Primary hepatocellular carcinoma (HCC) and xenograft models were utilized to investigate the effect of FCN3 on the development of HCC in vivo. A metabonomic analysis was conducted to assess alterations in intracellular and HCC intrahepatic lipid levels. RESULTS Our study elucidates a substantial decrease in the expression of FCN3, a component of the complement system, leads to MUFA accumulation in human HCC specimens and thereby significantly promotes ferroptosis resistance. Overexpression of FCN3 efficiently sensitizes HCC cells to ferroptosis, resulting in the inhibition of the oncogenesis and progression of both primary HCC and subcutaneous HCC xenograft. Mechanistically, FCN3 directly binds to the insulin receptor β (IR-β) and its pro-form (pro-IR), inhibiting pro-IR cleavage and IR-β phosphorylation, ultimately resulting in IR-β inactivation. This inactivation of IR-β suppresses the expression of sterol regulatory element binding protein-1c (SREBP1c), which subsequently suppresses the transcription of genes related to de novo lipogenesis (DNL) and lipid desaturation, and consequently downregulates intracellular MUFA levels. CONCLUSIONS These findings uncover a novel regulatory mechanism by which FCN3 enhances the sensitivity of HCC cells to ferroptosis, indicating that targeting FCN3-induced ferroptosis is a promising strategy for HCC treatment.
Collapse
Affiliation(s)
- Yanmei Yuan
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Junting Xu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Quanxin Jiang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chuanxin Yang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ning Wang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaolong Liu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Hai-Long Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Sijia Lu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xianjing Zhang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Liu Han
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhiyan Liu
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jiabin Cai
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Zhongshan Hospital, Shanghai, 200032, China.
| | - Fang Liu
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Suzhen Chen
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Junli Liu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
18
|
Lee J, Jang S, Im J, Han Y, Kim S, Jo H, Wang W, Cho U, Kim SI, Seol A, Kim B, Song YS. Stearoyl-CoA desaturase 1 inhibition induces ER stress-mediated apoptosis in ovarian cancer cells. J Ovarian Res 2024; 17:73. [PMID: 38566208 PMCID: PMC10988872 DOI: 10.1186/s13048-024-01389-1] [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: 11/30/2023] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Ovarian cancer is a leading cause of death among gynecologic tumors, often detected at advanced stages. Metabolic reprogramming and increased lipid biosynthesis are key factors driving cancer cell growth. Stearoyl-CoA desaturase 1 (SCD1) is a crucial enzyme involved in de novo lipid synthesis, producing mono-unsaturated fatty acids (MUFAs). Here, we aimed to investigate the expression and significance of SCD1 in epithelial ovarian cancer (EOC). Comparative analysis of normal ovarian surface epithelial (NOSE) tissues and cell lines revealed elevated SCD1 expression in EOC tissues and cells. Inhibition of SCD1 significantly reduced the proliferation of EOC cells and patient-derived organoids and induced apoptotic cell death. Interestingly, SCD1 inhibition did not affect the viability of non-cancer cells, indicating selective cytotoxicity against EOC cells. SCD1 inhibition on EOC cells induced endoplasmic reticulum (ER) stress by activating the unfolded protein response (UPR) sensors and resulted in apoptosis. The addition of exogenous oleic acid, a product of SCD1, rescued EOC cells from ER stress-mediated apoptosis induced by SCD1 inhibition, underscoring the importance of lipid desaturation for cancer cell survival. Taken together, our findings suggest that the inhibition of SCD1 is a promising biomarker as well as a novel therapeutic target for ovarian cancer by regulating ER stress and inducing cancer cell apoptosis.
Collapse
Affiliation(s)
- Juwon Lee
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Suin Jang
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jihye Im
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Youngjin Han
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Soochi Kim
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - HyunA Jo
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Wenyu Wang
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Untack Cho
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Aeran Seol
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Boyun Kim
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan, Republic of Korea
| | - Yong Sang Song
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea.
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, Republic of Korea.
- Department of Obstetrics and Gynecology, Myongji Hospital, Hanyang University College of Medicine, Goyang, Republic of Korea.
| |
Collapse
|
19
|
Wang J, Li J, Fu Y, Zhu Y, Lin L, Li Y. Research progress, challenges and perspectives of phospholipids metabolism in the LXR‑LPCAT3 signaling pathway and its relation to NAFLD (Review). Int J Mol Med 2024; 53:32. [PMID: 38362962 PMCID: PMC10903931 DOI: 10.3892/ijmm.2024.5356] [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: 09/22/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Phospholipids (PLs) are principle constituents of biofilms, with their fatty acyl chain composition significantly impacting the biophysical properties of membranes, thereby influencing biological processes. Recent studies have elucidated that fatty acyl chains, under the enzymatic action of lyso‑phosphatidyl‑choline acyltransferases (LPCATs), expedite incorporation into the sn‑2 site of phosphatidyl‑choline (PC), profoundly affecting pathophysiology. Accumulating evidence suggests that alterations in LPCAT activity are implicated in various diseases, including non‑alcoholic fatty liver disease (NAFLD), hepatitis C, atherosclerosis and cancer. Specifically, LPCAT3 is instrumental in maintaining systemic lipid homeostasis through its roles in hepatic lipogenesis, intestinal lipid absorption and lipoprotein secretion. The liver X receptor (LXR), pivotal in lipid homeostasis, modulates cholesterol, fatty acid (FA) and PL metabolism. LXR's capacity to modify PL composition in response to cellular sterol fluctuations is a vital mechanism for protecting biofilms against lipid stress. Concurrently, LXR activation enhances LPCAT3 expression on cell membranes and elevates polyunsaturated PL levels. This activation can ameliorate saturated free FA effects in vitro or endoplasmic reticulum stress in vivo due to lipid accumulation in hepatic cells. Pharmacological interventions targeting LXR, LPCAT and membrane PL components could offer novel therapeutic directions for NAFLD management. The present review primarily focused on recent advancements in understanding the LPCAT3 signaling pathway's role in lipid metabolism related to NAFLD, aiming to identify new treatment targets for the disease.
Collapse
Affiliation(s)
- Junmin Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Jiacheng Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yugang Fu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yingying Zhu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Liubing Lin
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yong Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
20
|
Heravi G, Liu Z, Herroon M, Wilson A, Fan YY, Jiang Y, Vakeesan N, Tao L, Peng Z, Zhang K, Li J, Chapkin RS, Podgorski I, Liu W. Targeting Fatty Acid Desaturase I Inhibits Renal Cancer Growth Via ATF3-mediated ER Stress Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.23.586426. [PMID: 38586033 PMCID: PMC10996531 DOI: 10.1101/2024.03.23.586426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Monounsaturated fatty acids (MUFAs) play a pivotal role in maintaining endoplasmic reticulum (ER) homeostasis, an emerging hallmark of cancer. However, the role of polyunsaturated fatty acid (PUFAs) desaturation in persistent ER stress driven by oncogenic abnormalities remains elusive. Fatty Acid Desaturase 1 (FADS1) is a rate-limiting enzyme controlling the bioproduction of long-chain PUFAs. Our previous research has demonstrated the significant role of FADS1 in cancer survival, especially in kidney cancers. We explored the underlying mechanism in this study. We found that pharmacological inhibition or knockdown of the expression of FADS1 effectively inhibits renal cancer cell proliferation and induces cell cycle arrest. The stable knockdown of FADS1 also significantly inhibits tumor formation in vivo. Mechanistically, we show that while FADS1 inhibition induces ER stress, its expression is also augmented by ER-stress inducers. Notably, FADS1-inhibition sensitized cellular response to ER stress inducers, providing evidence of FADS1's role in modulating the ER stress response in cancer cells. We show that, while FADS1 inhibition-induced ER stress leads to activation of ATF3, ATF3-knockdown rescues the FADS1 inhibition-induced ER stress and cell growth suppression. In addition, FADS1 inhibition results in the impaired biosynthesis of nucleotides and decreases the level of UPD-N-Acetylglucosamine, a critical mediator of the unfolded protein response. Our findings suggest that PUFA desaturation is crucial for rescuing cancer cells from persistent ER stress, supporting FADS1 as a new therapeutic target.
Collapse
Affiliation(s)
- Gioia Heravi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Zhenjie Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Mackenzie Herroon
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Alexis Wilson
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Yang-Yi Fan
- Department of Nutrition, Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
| | - Yang Jiang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Nivisa Vakeesan
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Li Tao
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zheyun Peng
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Jing Li
- Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Robert S. Chapkin
- Department of Nutrition, Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX, 77843, USA
| | - Izabela Podgorski
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| |
Collapse
|
21
|
Zhao Z, Li B, Chen Q, Xiang X, Xu X, Han S, Lai W, Li Y, Xu W, Mai K, Ai Q. Dietary palm oil enhances Sterol regulatory element-binding protein 2-mediated cholesterol biosynthesis through inducing endoplasmic reticulum stress in muscle of large yellow croaker ( Larimichthys crocea). Br J Nutr 2024; 131:553-566. [PMID: 37699661 DOI: 10.1017/s0007114523001344] [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: 09/14/2023]
Abstract
Sterol regulatory element-binding protein 2 (SREBP2) is considered to be a major regulator to control cholesterol homoeostasis in mammals. However, the role of SREBP2 in teleost remains poorly understand. Here, we explored the molecular characterisation of SREBP2 and identified SREBP2 as a key modulator for 3-hydroxy-3-methylglutaryl-coenzyme A reductase and 7-dehydrocholesterol reductase, which were rate-limiting enzymes of cholesterol biosynthesis. Moreover, dietary palm oil in vivo or palmitic acid (PA) treatment in vitro elevated cholesterol content through triggering SREBP2-mediated cholesterol biosynthesis in large yellow croaker. Furthermore, our results also found that PA-induced activation of SREBP2 was dependent on the stimulating of endoplasmic reticulum stress (ERS) in croaker myocytes and inhibition of ERS by 4-Phenylbutyric acid alleviated PA-induced SREBP2 activation and cholesterol biosynthesis. In summary, our findings reveal a novel insight for understanding the role of SREBP2 in the regulation of cholesterol metabolism in fish and may deepen the link between dietary fatty acid and cholesterol biosynthesis.
Collapse
Affiliation(s)
- Zengqi Zhao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Baolin Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Qiang Chen
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Xiaojun Xiang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Xiang Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Shangzhe Han
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Wencong Lai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Yueru Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Wei Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong266237, People's Republic of China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Ocean University of China, 5 Yushan Road, Qingdao, Shandong266003, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong266237, People's Republic of China
| |
Collapse
|
22
|
Ning D, Jin J, Fang Y, Du P, Yuan C, Chen J, Huang Q, Cheng K, Mo J, Xu L, Guo H, Yang MJ, Chen X, Liang H, Zhang B, Zhang W. DEAD-Box Helicase 17 exacerbates non-alcoholic steatohepatitis via transcriptional repression of cyp2c29, inducing hepatic lipid metabolism disorder and eliciting the activation of M1 macrophages. Clin Transl Med 2024; 14:e1529. [PMID: 38303609 PMCID: PMC10835191 DOI: 10.1002/ctm2.1529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 02/03/2024] Open
Abstract
OBJECTIVE Our study was to elucidate the role of RNA helicase DEAD-Box Helicase 17 (DDX17) in NAFLD and to explore its underlying mechanisms. METHODS We created hepatocyte-specific Ddx17-deficient mice aim to investigate the impact of Ddx17 on NAFLD induced by a high-fat diet (HFD) as well as methionine and choline-deficient l-amino acid diet (MCD) in adult male mice. RNA-seq and lipidomic analyses were conducted to depict the metabolic landscape, and CUT&Tag combined with chromatin immunoprecipitation (ChIP) and luciferase reporter assays were conducted. RESULTS In this work, we observed a notable increase in DDX17 expression in the livers of patients with NASH and in murine models of NASH induced by HFD or MCD. After introducing lentiviruses into hepatocyte L02 for DDX17 knockdown or overexpression, we found that lipid accumulation induced by palmitic acid/oleic acid (PAOA) in L02 cells was noticeably weakened by DDX17 knockdown but augmented by DDX17 overexpression. Furthermore, hepatocyte-specific DDX17 knockout significantly alleviated hepatic steatosis, inflammatory response and fibrosis in mice after the administration of MCD and HFD. Mechanistically, our analysis of RNA-seq and CUT&Tag results combined with ChIP and luciferase reporter assays indicated that DDX17 transcriptionally represses Cyp2c29 gene expression by cooperating with CCCTC binding factor (CTCF) and DEAD-Box Helicase 5 (DDX5). Using absolute quantitative lipidomics analysis, we identified a hepatocyte-specific DDX17 deficiency that decreased lipid accumulation and altered lipid composition in the livers of mice after MCD administration. Based on the RNA-seq analysis, our findings suggest that DDX17 could potentially have an impact on the modulation of lipid metabolism and the activation of M1 macrophages in murine NASH models. CONCLUSION These results imply that DDX17 is involved in NASH development by promoting lipid accumulation in hepatocytes, inducing the activation of M1 macrophages, subsequent inflammatory responses and fibrosis through the transcriptional repression of Cyp2c29 in mice. Therefore, DDX17 holds promise as a potential drug target for the treatment of NASH.
Collapse
Affiliation(s)
- Deng Ning
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
- Department of Hepatobiliary SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jie Jin
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
| | - Yuanyuan Fang
- Department of NeurologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Pengcheng Du
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
| | - Chaoyi Yuan
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
| | - Jin Chen
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
| | - Qibo Huang
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
| | - Kun Cheng
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
| | - Jie Mo
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
| | - Lei Xu
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
| | - Hui Guo
- Institute of Organ TransplantationTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Mia Jiming Yang
- Institute for Management in Medicine and Health SciencesUniversity of BayreuthBayreuthGermany
| | - Xiaoping Chen
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of HealthWuhanChina
| | - Huifang Liang
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of HealthWuhanChina
| | - Bixiang Zhang
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of HealthWuhanChina
| | - Wanguang Zhang
- Department of Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanChina
- Key Laboratory of Organ TransplantationMinistry of Education and Ministry of HealthWuhanChina
| |
Collapse
|
23
|
Wu J, Yang OJ, Soderblom EJ, Yan D. Heat Shock Proteins Function as Signaling Molecules to Mediate Neuron-Glia Communication During Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576052. [PMID: 38293019 PMCID: PMC10827141 DOI: 10.1101/2024.01.18.576052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The nervous system is primarily composed of neurons and glia, and the communication between them plays profound roles in regulating the development and function of the brain. Neuron-glia signal transduction is known to be mediated by secreted or juxtacrine signals through ligand-receptor interactions on the cell membrane. Here, we report a novel mechanism for neuron-glia signal transduction, wherein neurons transmit proteins to glia through extracellular vesicles, activating glial signaling pathways. We find that in the amphid sensory organ of Caenorhabditis elegans, different sensory neurons exhibit varying aging rates. This discrepancy in aging is governed by the crosstalk between neurons and glia. We demonstrate that early-aged neurons can transmit heat shock proteins (HSP) to glia via extracellular vesicles. These neuronal HSPs activate the IRE1-XBP1 pathway, further increasing their expression in glia, forming a positive feedback loop. Ultimately, the activation of the IRE1-XBP-1 pathway leads to the transcriptional regulation of chondroitin synthases to protect glia-embedded neurons from aging-associated functional decline. Therefore, our studies unveil a novel mechanism for neuron-glia communication in the nervous system and provide new insights into our understanding of brain aging.
Collapse
Affiliation(s)
- Jieyu Wu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Olivia Jiaming Yang
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
- East Chapel Hill High School, Chapel Hill, NC 27514, USA
| | - Erik J. Soderblom
- Proteomics and Metabolomics Core Facility, Duke University Medical School, Durham, NC 27710, USA
| | - Dong Yan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell biology, Department of Neurobiology, Regeneration next, and Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
24
|
Yilmaz E. Endoplasmic Reticulum Stress and Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:373-390. [PMID: 39287859 DOI: 10.1007/978-3-031-63657-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
In recent years, the world has seen an alarming increase in obesity and is closely associated with insulin resistance, which is a state of low-grade inflammation, the latter characterized by elevated levels of proinflammatory cytokines in blood and tissues. A shift in energy balance alters systemic metabolic regulation and the important role that chronic inflammation, endoplasmic reticulum (ER) dysfunction, and activation of the unfolded protein response (UPR) plays in this process.Why obesity is so closely associated with insulin resistance and inflammation is not understood well. This suggests that there are probably many causes for obesity-related insulin resistance and inflammation. One of the faulty mechanisms is protein homeostasis, protein quality control system included protein folding, chaperone activity, and ER-associated degradation leading to endoplasmic reticulum (ER) stress.The ER is a vast membranous network responsible for the trafficking of a wide range of proteins and plays a central role in integrating multiple metabolic signals critical in cellular homeostasis. Conditions that may trigger unfolded protein response activation include increased protein synthesis, the presence of mutant or misfolded proteins, inhibition of protein glycosylation, imbalance of ER calcium levels, glucose and energy deprivation, hypoxia, pathogens, or pathogen-associated components and toxins. Thus, characterizing the mechanisms contributing to obesity and identifying potential targets for its prevention and treatment will have a great impact on the control of associated conditions, particularly T2D.
Collapse
Affiliation(s)
- Erkan Yilmaz
- Biotechnology Institute, Ankara University, Kecioren, Ankara, Turkey.
| |
Collapse
|
25
|
Kohler A, Kohler V. Better Together: Interorganellar Communication in the Regulation of Proteostasis. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241272245. [PMID: 39385949 PMCID: PMC11462569 DOI: 10.1177/25152564241272245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 10/12/2024]
Abstract
An extensive network of chaperones and folding factors is responsible for maintaining a functional proteome, which is the basis for cellular life. The underlying proteostatic mechanisms are not isolated within organelles, rather they are connected over organellar borders via signalling processes or direct association via contact sites. This review aims to provide a conceptual understanding of proteostatic mechanisms across organelle borders, not focussing on individual organelles. This discussion highlights the precision of these finely tuned systems, emphasising the complicated balance between cellular protection and adaptation to stress. In this review, we discuss widely accepted aspects while shedding light on newly discovered perspectives.
Collapse
Affiliation(s)
- Andreas Kohler
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87 Umeå, Sweden
| | - Verena Kohler
- Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
26
|
Morito K, Ali H, Kishino S, Tanaka T. Fatty Acid Metabolism in Peroxisomes and Related Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1470:31-55. [PMID: 38811487 DOI: 10.1007/5584_2024_802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
One of the functions of peroxisomes is the oxidation of fatty acids (FAs). The importance of this function in our lives is evidenced by the presence of peroxisomal disorders caused by the genetic deletion of proteins involved in these processes. Unlike mitochondrial oxidation, peroxisomal oxidation is not directly linked to ATP production. What is the role of FA oxidation in peroxisomes? Recent studies have revealed that peroxisomes supply the building blocks for lipid synthesis in the endoplasmic reticulum and facilitate intracellular carbon recycling for membrane quality control. Accumulation of very long-chain fatty acids (VLCFAs), which are peroxisomal substrates, is a diagnostic marker in many types of peroxisomal disorders. However, the relationship between VLCFA accumulation and various symptoms of these disorders remains unclear. Recently, we developed a method for solubilizing VLCFAs in aqueous media and found that VLCFA toxicity could be mitigated by oleic acid replenishment. In this chapter, we present the physiological role of peroxisomal FA oxidation and the knowledge obtained from VLCFA-accumulating peroxisome-deficient cells.
Collapse
Affiliation(s)
- Katsuya Morito
- Laboratory of Environmental Biochemistry, Division of Biological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hanif Ali
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | | | - Tamotsu Tanaka
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan.
| |
Collapse
|
27
|
Raza S, Rajak S, Singh R, Zhou J, Sinha RA, Goel A. Cell-type specific role of autophagy in the liver and its implications in non-alcoholic fatty liver disease. World J Hepatol 2023; 15:1272-1283. [PMID: 38192406 PMCID: PMC7615497 DOI: 10.4254/wjh.v15.i12.1272] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/07/2023] [Accepted: 12/08/2023] [Indexed: 12/25/2023] Open
Abstract
Autophagy, a cellular degradative process, has emerged as a key regulator of cellular energy production and stress mitigation. Dysregulated autophagy is a common phenomenon observed in several human diseases, and its restoration offers curative advantage. Non-alcoholic fatty liver disease (NAFLD), more recently renamed metabolic dysfunction-associated steatotic liver disease, is a major metabolic liver disease affecting almost 30% of the world population. Unfortunately, NAFLD has no pharmacological therapies available to date. Autophagy regulates several hepatic processes including lipid metabolism, inflammation, cellular integrity and cellular plasticity in both parenchymal (hepatocytes) and non-parenchymal cells (Kupffer cells, hepatic stellate cells and sinusoidal endothelial cells) with a profound impact on NAFLD progression. Understanding cell type-specific autophagy in the liver is essential in order to develop targeted treatments for liver diseases such as NAFLD. Modulating autophagy in specific cell types can have varying effects on liver function and pathology, making it a promising area of research for liver-related disorders. This review aims to summarize our present understanding of cell-type specific effects of autophagy and their implications in developing autophagy centric therapies for NAFLD.
Collapse
Affiliation(s)
- Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India
| | - Rajani Singh
- Department of Hepatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India
| | - Jin Zhou
- CVMD, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India
| | - Amit Goel
- Department of Hepatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India.
| |
Collapse
|
28
|
Liu M, Zhang Z, Chen Y, Feng T, Zhou Q, Tian X. Circadian clock and lipid metabolism disorders: a potential therapeutic strategy for cancer. Front Endocrinol (Lausanne) 2023; 14:1292011. [PMID: 38189049 PMCID: PMC10770836 DOI: 10.3389/fendo.2023.1292011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Recent research has emphasized the interaction between the circadian clock and lipid metabolism, particularly in relation to tumors. This review aims to explore how the circadian clock regulates lipid metabolism and its impact on carcinogenesis. Specifically, targeting key enzymes involved in fatty acid synthesis (SREBP, ACLY, ACC, FASN, and SCD) has been identified as a potential strategy for cancer therapy. By disrupting these enzymes, it may be possible to inhibit tumor growth by interfering with lipid metabolism. Transcription factors, like SREBP play a significant role in regulating fatty acid synthesis which is influenced by circadian clock genes such as BMAL1, REV-ERB and DEC. This suggests a strong connection between fatty acid synthesis and the circadian clock. Therefore, successful combination therapy should target fatty acid synthesis in addition to considering the timing and duration of drug use. Ultimately, personalized chronotherapy can enhance drug efficacy in cancer treatment and achieve treatment goals.
Collapse
Affiliation(s)
- Mengsi Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention and Treatment, Hunan University of Chinese Medicine, Changsha, China
| | - Zhen Zhang
- Department of Oncology, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China
| | - Yating Chen
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention and Treatment, Hunan University of Chinese Medicine, Changsha, China
| | - Ting Feng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention and Treatment, Hunan University of Chinese Medicine, Changsha, China
| | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xuefei Tian
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention and Treatment, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
29
|
Shao F, Hu X, Li J, Bai B, Tian L. Lipidomics analysis of impaired glucose tolerance and type 2 diabetes mellitus in overweight or obese elderly adults. Endocr Connect 2023; 12:e230212. [PMID: 37878774 PMCID: PMC10692693 DOI: 10.1530/ec-23-0212] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/25/2023] [Indexed: 10/27/2023]
Abstract
Aims Aging, obesity, and type 2 diabetes mellitus (T2DM) form a metabolic disease continuum that has a continuously increasing prevalence. Lipidomics explains the complex interactions between lipid metabolism and metabolic diseases. We aimed to systematically investigate the plasma lipidome changes induced by newly diagnosed impaired glucose tolerance (IGT) and T2DM in overweight/obese elderly individuals and to identify potential biomarkers to differentiate between the IGT, T2DM, and control groups. Methods Plasma samples from 148 overweight/obese elderly individuals, including 52 patients with IGT, 47 patients with T2DM, and 49 euglycemic controls, were analyzed using a high-coverage nontargeted absolute quantitative lipidomics approach. Results We quantified 1840 lipids from thirty-eight classes and seven lipid categories. Among overweight/obese elderly individuals, the lipidomic profiles of IGT and T2DM patients were significantly different from those of controls, while they were similar in the IGT and T2DM groups. The concentrations of diglycerides, triglycerides, phosphatidylcholines, and ceramides were obviously altered in the IGT and T2DM groups. Particularly, IGT and T2DM induced the accumulation of triglycerides with longer carbon atom numbers (C44-50) and saturated or lower double bond numbers (n (C=C) = 0-2). Furthermore, a total of 17 potential lipidic biomarkers were identified to successfully differentiate between the IGT, T2DM, and control groups. Conclusions In overweight/obese elderly patients, IGT and T2DM induced apparent lipidome-wide changes. This study's results may contribute to explaining the complex dysfunctional lipid metabolism in aging, obesity, and diabetes.
Collapse
Affiliation(s)
- Feifei Shao
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu, China
- Clinical Research Center for Metabolic Disease, Gansu Province, Lanzhou, Gansu, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Xinxin Hu
- Clinical Research Center for Metabolic Disease, Gansu Province, Lanzhou, Gansu, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Jiayu Li
- Clinical Research Center for Metabolic Disease, Gansu Province, Lanzhou, Gansu, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Bona Bai
- Clinical Research Center for Metabolic Disease, Gansu Province, Lanzhou, Gansu, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Limin Tian
- Department of Endocrinology, Gansu Provincial Hospital, Lanzhou, Gansu, China
- Clinical Research Center for Metabolic Disease, Gansu Province, Lanzhou, Gansu, China
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
30
|
Gebert M, Sławski J, Kalinowski L, Collawn JF, Bartoszewski R. The Unfolded Protein Response: A Double-Edged Sword for Brain Health. Antioxidants (Basel) 2023; 12:1648. [PMID: 37627643 PMCID: PMC10451475 DOI: 10.3390/antiox12081648] [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: 07/26/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Efficient brain function requires as much as 20% of the total oxygen intake to support normal neuronal cell function. This level of oxygen usage, however, leads to the generation of free radicals, and thus can lead to oxidative stress and potentially to age-related cognitive decay and even neurodegenerative diseases. The regulation of this system requires a complex monitoring network to maintain proper oxygen homeostasis. Furthermore, the high content of mitochondria in the brain has elevated glucose demands, and thus requires a normal redox balance. Maintaining this is mediated by adaptive stress response pathways that permit cells to survive oxidative stress and to minimize cellular damage. These stress pathways rely on the proper function of the endoplasmic reticulum (ER) and the activation of the unfolded protein response (UPR), a cellular pathway responsible for normal ER function and cell survival. Interestingly, the UPR has two opposing signaling pathways, one that promotes cell survival and one that induces apoptosis. In this narrative review, we discuss the opposing roles of the UPR signaling pathways and how a better understanding of these stress pathways could potentially allow for the development of effective strategies to prevent age-related cognitive decay as well as treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Magdalena Gebert
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-134 Gdansk, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383 Wroclaw, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-134 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland
| | - James F. Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383 Wroclaw, Poland
| |
Collapse
|
31
|
Waidmann S, Béziat C, Ferreira Da Silva Santos J, Feraru E, Feraru MI, Sun L, Noura S, Boutté Y, Kleine-Vehn J. Endoplasmic reticulum stress controls PIN-LIKES abundance and thereby growth adaptation. Proc Natl Acad Sci U S A 2023; 120:e2218865120. [PMID: 37487064 PMCID: PMC10400986 DOI: 10.1073/pnas.2218865120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 06/21/2023] [Indexed: 07/26/2023] Open
Abstract
Extreme environmental conditions eventually limit plant growth [J. R. Dinneny, Annu. Rev. Cell Dev. Biol. 35, 1-19 (2019), N. Gigli-Bisceglia, C. Testerink, Curr. Opin. Plant Biol. 64, 102120 (2021)]. Here, we reveal a mechanism that enables multiple external cues to get integrated into auxin-dependent growth programs in Arabidopsis thaliana. Our forward genetics approach on dark-grown hypocotyls uncovered that an imbalance in membrane lipids enhances the protein abundance of PIN-LIKES (PILS) [E. Barbez et al., Nature 485, 119 (2012)] auxin transport facilitators at the endoplasmic reticulum (ER), which thereby limits nuclear auxin signaling and growth rates. We show that this subcellular response relates to ER stress signaling, which directly impacts PILS protein turnover in a tissue-dependent manner. This mechanism allows PILS proteins to integrate environmental input with phytohormone auxin signaling, contributing to stress-induced growth adaptation in plants.
Collapse
Affiliation(s)
- Sascha Waidmann
- Institute of Biology II, Chair of Molecular Plant Physiology, University of Freiburg, 79104Freiburg, Germany
- Center for Integrative Biological Signalling Studies, University of Freiburg, 79104Freiburg, Germany
- Department of Applied Genetics and Cell Biology, Institute of Molecular Plant Biology, University of Natural Resources and Life Sciences,1190Vienna, Austria
| | - Chloé Béziat
- Department of Applied Genetics and Cell Biology, Institute of Molecular Plant Biology, University of Natural Resources and Life Sciences,1190Vienna, Austria
| | - Jonathan Ferreira Da Silva Santos
- Institute of Biology II, Chair of Molecular Plant Physiology, University of Freiburg, 79104Freiburg, Germany
- Department of Applied Genetics and Cell Biology, Institute of Molecular Plant Biology, University of Natural Resources and Life Sciences,1190Vienna, Austria
| | - Elena Feraru
- Department of Applied Genetics and Cell Biology, Institute of Molecular Plant Biology, University of Natural Resources and Life Sciences,1190Vienna, Austria
| | - Mugurel I. Feraru
- Department of Applied Genetics and Cell Biology, Institute of Molecular Plant Biology, University of Natural Resources and Life Sciences,1190Vienna, Austria
| | - Lin Sun
- Department of Applied Genetics and Cell Biology, Institute of Molecular Plant Biology, University of Natural Resources and Life Sciences,1190Vienna, Austria
| | - Seinab Noura
- Institute of Biology II, Chair of Molecular Plant Physiology, University of Freiburg, 79104Freiburg, Germany
- Center for Integrative Biological Signalling Studies, University of Freiburg, 79104Freiburg, Germany
| | - Yohann Boutté
- CNRS-University of Bordeaux, UMR 5200 Membrane Biogenesis Laboratory, National Research Institute for Agriculture, Food and the Environment Bordeaux Aquitaine, 33140 Bordeaux, France
| | - Jürgen Kleine-Vehn
- Institute of Biology II, Chair of Molecular Plant Physiology, University of Freiburg, 79104Freiburg, Germany
- Center for Integrative Biological Signalling Studies, University of Freiburg, 79104Freiburg, Germany
- Department of Applied Genetics and Cell Biology, Institute of Molecular Plant Biology, University of Natural Resources and Life Sciences,1190Vienna, Austria
| |
Collapse
|
32
|
Pang MD, Bastings JJAJ, Op den Kamp-Bruls YMH, Harrold JA, Kjølbaek L, Halford JCG, Adam TCM, Raben A, Schrauwen-Hinderling VB, Goossens GH, Blaak EE. The effect of weight loss on whole-body and tissue-specific insulin sensitivity and hepatic lipid content and composition: SWEET substudy. Obesity (Silver Spring) 2023; 31:1745-1754. [PMID: 37368517 DOI: 10.1002/oby.23773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/22/2023] [Accepted: 03/13/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE This study (1) investigated the effect of weight loss on whole-body and tissue-specific insulin sensitivity and on intrahepatic lipid (IHL) content and composition and (2) investigated the association between weight-loss-induced changes in insulin sensitivity and IHL content in individuals with overweight or obesity. METHODS In this secondary analysis of the European SWEET project, 50 adults (age 18-65 years) with overweight or obesity (BMI ≥ 25 kg/m2 ) followed a low-energy diet (LED) for 2 months. At baseline and after the LED, body composition (dual-energy x-ray absorptiometry), IHL content and composition (proton magnetic resonance spectroscopy), whole-body insulin sensitivity (Matsuda index), muscle insulin sensitivity index (MISI), and hepatic insulin resistance index (HIRI) were determined (7-point oral glucose tolerance test). RESULTS The LED reduced body weight (p < 0.001). This was accompanied by increased Matsuda index and reduced HIRI (both p < 0.001) but no change in MISI (p = 0.260). Weight loss decreased IHL content (mean [SEM], 3.9% [0.7%] vs. 1.6% [0.5%], p < 0.001) and the hepatic saturated fatty acid fraction (41.0% [1.5%] vs. 36.6% [1.9%], p = 0.039). The reduced IHL content was associated with an improvement in HIRI (r = 0.402, p = 0.025). CONCLUSIONS Weight loss decreased IHL content and the hepatic saturated fatty acid fraction. The decrease in IHL content was associated with weight-loss-induced improvement in hepatic insulin sensitivity in individuals with overweight or obesity.
Collapse
Affiliation(s)
- Michelle D Pang
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jacco J A J Bastings
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Yvonne M H Op den Kamp-Bruls
- Department of Radiology and Nuclear Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Joanne A Harrold
- Department of Psychology, Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Louise Kjølbaek
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Jason C G Halford
- Department of Psychology, Institute of Population Health, University of Liverpool, Liverpool, UK
- School of Psychology, University of Leeds, Leeds, UK
| | - Tanja C M Adam
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Anne Raben
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
- Clinical Research, Copenhagen University Hospital-Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Vera B Schrauwen-Hinderling
- Department of Radiology and Nuclear Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Gijs H Goossens
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
33
|
Sen U, Coleman C, Sen T. Stearoyl coenzyme A desaturase-1: multitasker in cancer, metabolism, and ferroptosis. Trends Cancer 2023; 9:480-489. [PMID: 37029018 DOI: 10.1016/j.trecan.2023.03.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 04/09/2023]
Abstract
Cancer progression is a highly balanced process and is maintained by a sequence of finely tuned metabolic pathways. Stearoyl coenzyme A desaturase-1 (SCD1), the fatty enzyme that converts saturated fatty acids into monounsaturated fatty acids, is a critical modulator of the fatty acid metabolic pathway. SCD1 expression is associated with poor prognosis in several cancer types. SCD1 triggers an iron-dependent cell death called ferroptosis and elevated levels of SCD1 protect cancer cells against ferroptosis. Pharmacological inhibition of SCD1 as monotherapy and in combination with chemotherapeutic agents shows promising antitumor potential in preclinical models. In this review, we summarize the role of SCD in cancer cell progression, survival, and ferroptosis and discuss potential strategies to exploit SCD1 inhibition in future clinical trials.
Collapse
Affiliation(s)
- Utsav Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Charles Coleman
- The Bioinformatics for Next Generation Sequencing (BiNGS) Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA
| | - Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
34
|
Dianat-Moghadam H, Abbasspour-Ravasjani S, Hamishehkar H, Rahbarghazi R, Nouri M. LXR inhibitor SR9243-loaded immunoliposomes modulate lipid metabolism and stemness in colorectal cancer cells. Med Oncol 2023; 40:156. [PMID: 37093287 DOI: 10.1007/s12032-023-02027-4] [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: 02/28/2023] [Accepted: 04/13/2023] [Indexed: 04/25/2023]
Abstract
Reprogrammed metabolism and active stemness contribute to cancer stem cells' (CSCs) survival and tumorigenesis. LXR signaling regulates the metabolism of different cancers. A selective LXR inhibitor, SR9243 (SR), can target and eradicate non-CSC tumor cells. CD133 is a stem marker in solid tumors-associated CSCs within the active lipogenesis, and anti-CD133 mAb targeting liposomal drug delivery systems expected to increase drug internalization and improve the therapeutic efficacy of poor-in water solubility drugs, e, g., SR. In this study, anti-CD133 mAbs-targeted Immunoliposomes (ILipo) were developed for specific delivery of SR into MACS-enriched CD133 + CSCs and induce their functional effects. Results have shown that ILipo having an average size of 64.79 nm can encapsulate SR in maximum proportion, and cell association studies have shown cationic ILipo and targeting CD133 provide the CSCs binding. Also, FCM analysis of RhoB has demonstrated that the ILipo uptake was higher in CD133 + CSCs than in the non-targeted liposomes. ILipo-SR was significantly more toxic in CD133 + CSCs compared to the free SR and non-targeted ones. More efficient than Lipo-SR, ILipo-SR improved the reduction of clonogenicity, stemness, and lipogenesis in CD133 + CSCs in vitro, boosted ROS generation, and induced apoptosis. Our study revealed the dual targeting of CD133 and LXR appears to be a promising strategy for targeting CD133 + CSCs-presenting dynamic metabolism and self-renewal potentials.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahammad Nouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
35
|
Janikiewicz J, Dobosz AM, Majzner K, Bernas T, Dobrzyn A. Stearoyl-CoA desaturase 1 deficiency exacerbates palmitate-induced lipotoxicity by the formation of small lipid droplets in pancreatic β-cells. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166711. [PMID: 37054998 DOI: 10.1016/j.bbadis.2023.166711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
The accelerating accumulation of surplus lipids in the pancreas triggers structural and functional changes in type 2 diabetes-affected islets. Pancreatic β-cells exhibit a restricted capacity to store fat reservoirs in lipid droplets (LDs), which act as transient buffers to prevent lipotoxic stress. With the increasing incidence of obesity, growing interest has been seen in the intracellular regulation of LD metabolism for β-cell function. Stearoyl-CoA desaturase 1 (SCD1) is critical for producing unsaturated fatty acyl moieties for fluent storage into and out of LDs, likely affecting the overall rate of β-cell survival. We explored LD-associated composition and remodeling in SCD1-deprived INS-1E cells and in pancreatic islets in wildtype and SCD1-/- mice in the lipotoxic milieu. Deficiency in the enzymatic activity of SCD1 led to decrease in the size and number of LDs and the lower accumulation of neutral lipids. This occurred in parallel with a higher compactness and lipid order inside LDs, followed by changes in the saturation status and composition of fatty acids within core lipids and the phospholipid coat. The lipidome of LDs was enriched in 18:2n-6 and 20:4n-6 in β-cells and pancreatic islets. These rearrangements markedly contributed to differences in protein association with the LD surface. Our findings highlight an unexpected molecular mechanism by which SCD1 activity affects the morphology, composition and metabolism of LDs. We demonstrate that SCD1-dependent disturbances in LD enrichment can impact proper pancreatic β-cells and islet functioning, which may have considerable therapeutic value for the management of type 2 diabetes.
Collapse
Affiliation(s)
- Justyna Janikiewicz
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| | - Aneta M Dobosz
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Majzner
- Faculty of Chemistry, Jagiellonian University, Cracow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Cracow, Poland
| | - Tytus Bernas
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Agnieszka Dobrzyn
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
36
|
Sun J, Mai K, Ai Q. Effects of GRP78 on Endoplasmic Reticulum Stress and Inflammatory Response in Macrophages of Large Yellow Croaker ( Larimichthys crocea). Int J Mol Sci 2023; 24:ijms24065855. [PMID: 36982929 PMCID: PMC10054070 DOI: 10.3390/ijms24065855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Endoplasmic reticulum (ER) homeostasis plays a vital role in cell physiological functions. Various factors can destroy the homeostasis of the ER and cause ER stress. Moreover, ER stress is often related to inflammation. Glucose-regulated protein 78 (GRP78) is an ER chaperone, which plays a vital role in maintaining cellular homeostasis. Nevertheless, the potential effects of GRP78 on ER stress and inflammation is still not fully elucidated in fish. In the present study, ER stress and inflammation was induced by tunicamycin (TM) or palmitic acid (PA) in the macrophages of large yellow croakers. GRP78 was treated with an agonist/inhibitor before or after the TM/PA treatment. The results showed that the TM/PA treatment could significantly induce ER stress and an inflammatory response in the macrophages of large yellow croakers whereas the incubation of the GRP78 agonist could reduce TM/PA-induced ER stress and an inflammatory response. Moreover, the incubation of the GRP78 inhibitor could further induce TM/PA-induced ER stress and an inflammatory response. These results provide an innovative idea to explain the relationship between GRP78 and TM/PA-induced ER stress or inflammation in large yellow croakers.
Collapse
Affiliation(s)
- Jie Sun
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
37
|
Sinha RA. Autophagy: A Cellular Guardian against Hepatic Lipotoxicity. Genes (Basel) 2023; 14:553. [PMID: 36874473 PMCID: PMC7614268 DOI: 10.3390/genes14030553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Lipotoxicity is a phenomenon of lipid-induced cellular injury in nonadipose tissue. Excess of free saturated fatty acids (SFAs) contributes to hepatic injury in nonalcoholic fatty liver disease (NAFLD), which has been growing at an unprecedented rate in recent years. SFAs and their derivatives such as ceramides and membrane phospholipids have been shown to induce intrahepatic oxidative damage and ER stress. Autophagy represents a cellular housekeeping mechanism to counter the perturbation in organelle function and activation of stress signals within the cell. Several aspects of autophagy, including lipid droplet assembly, lipophagy, mitophagy, redox signaling and ER-phagy, play a critical role in mounting a strong defense against lipotoxic lipid species within the hepatic cells. This review provides a succinct overview of our current understanding of autophagy-lipotoxicity interaction and its pharmacological and nonpharmacological modulation in treating NAFLD.
Collapse
Affiliation(s)
- Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
38
|
Erythrocyte Plasma Membrane Lipid Composition Mirrors That of Neurons and Glial Cells in Murine Experimental In Vitro and In Vivo Inflammation. Cells 2023; 12:cells12040561. [PMID: 36831228 PMCID: PMC9953778 DOI: 10.3390/cells12040561] [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: 01/05/2023] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Lipid membrane turnover and myelin repair play a central role in diseases and lesions of the central nervous system (CNS). The aim of the present study was to analyze lipid composition changes due to inflammatory conditions. We measured the fatty acid (FA) composition in erythrocytes (RBCs) and spinal cord tissue (gas chromatography) derived from mice affected by experimental allergic encephalomyelitis (EAE) in acute and remission phases; cholesterol membrane content (Filipin) and GM1 membrane assembly (CT-B) in EAE mouse RBCs, and in cultured neurons, oligodendroglial cells and macrophages exposed to inflammatory challenges. During the EAE acute phase, the RBC membrane showed a reduction in polyunsaturated FAs (PUFAs) and an increase in saturated FAs (SFAs) and the omega-6/omega-3 ratios, followed by a restoration to control levels in the remission phase in parallel with an increase in monounsaturated fatty acid residues. A decrease in PUFAs was also shown in the spinal cord. CT-B staining decreased and Filipin staining increased in RBCs during acute EAE, as well as in cultured macrophages, neurons and oligodendrocyte precursor cells exposed to inflammatory challenges. This regulation in lipid content suggests an increased cell membrane rigidity during the inflammatory phase of EAE and supports the investigation of peripheral cell membrane lipids as possible biomarkers for CNS lipid membrane concentration and assembly.
Collapse
|
39
|
Raj RR, Lofquist S, Lee MJ. Remodeling of Adipose Tissues by Fatty Acids: Mechanistic Update on Browning and Thermogenesis by n-3 Polyunsaturated Fatty Acids. Pharm Res 2023; 40:467-480. [PMID: 36050546 DOI: 10.1007/s11095-022-03377-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
Enhancing thermogenesis by increasing the amount and activity of brown and brite adipocytes is a potential therapeutic target for obesity and its associated diseases. Diet plays important roles in energy metabolism and a myriad of dietary components including lipids are known to regulate thermogenesis through recruitment and activation of brown and brite adipocytes. Depending on types of fatty acids (FAs), the major constituent in lipids, their health benefits differ. Long-chain polyunsaturated FAs (PUFAs), especially n-3 PUFAs remodel adipose tissues in a healthier manner with reduced inflammation and enhanced thermogenesis, while saturated FAs exhibit contrasting effects. Lipid mediators derived from FAs act as autocrine/paracrine as well as endocrine factors to regulate thermogenesis. We discuss lipid mediators that may contribute to the differential effects of FAs on adipose tissue remodeling and hence, cardiometabolic diseases. We also discuss current understanding of molecular and cellular mechanisms through which n-3 PUFAs enhance thermogenesis. Elucidating molecular details of beneficial effects of n-3 PUFAs on thermogenesis is expected to provide information that can be used for development of novel therapeutics for obesity and its associated diseases.
Collapse
Affiliation(s)
- Radha Raman Raj
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, 1955 East West Road, Honolulu, HI, 98622, USA
| | - Sydney Lofquist
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, 1955 East West Road, Honolulu, HI, 98622, USA
| | - Mi-Jeong Lee
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, 1955 East West Road, Honolulu, HI, 98622, USA.
| |
Collapse
|
40
|
Jia H, Liu J, Fang T, Zhou Z, Li R, Yin W, Qian Y, Wang Q, Zhou W, Liu C, Sun D, Chen X, Ouyang Z, Dong J, Wang Y, Yue S. The role of altered lipid composition and distribution in liver fibrosis revealed by multimodal nonlinear optical microscopy. SCIENCE ADVANCES 2023; 9:eabq2937. [PMID: 36638165 PMCID: PMC9839333 DOI: 10.1126/sciadv.abq2937] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Intracellular lipid accumulation is commonly seen in fibrotic livers, but its exact role in liver fibrosis remains elusive. Here, we established a multimodal nonlinear optical microscopy to quantitatively map distribution of biomolecules in fibrotic livers. Our data revealed that unsaturated triglycerides were predominantly accumulated in central vein area during liver fibrosis but not in portal vein area. Moreover, the lipid homeostasis was remarkably dysregulated in the late-stage compared to the early-stage fibrosis, including increased unsaturated triglycerides with decreased lipid unsaturation degree and decreased membrane fluidity. Such alterations were likely due to up-regulated lipogenesis, desaturation, and peroxidation, which consequently led to endoplasmic reticulum stress and cell death. Inspiringly, injured hepatocyte could be rescued by remodeling lipid homeostasis via either supply of unsaturated fatty acids or enhancement of membrane fluidity. Collectively, our study improves current understanding of the role of lipid homeostasis in fibrosis and open opportunities for treatment.
Collapse
Affiliation(s)
- Hao Jia
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Juan Liu
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, 102218, China
| | - Tinghe Fang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Zhen Zhou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Ruihong Li
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, 102218, China
| | - Wenzhen Yin
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Yao Qian
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing 100084, China
| | - Qi Wang
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, 102218, China
| | - Wanhui Zhou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Chang Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Dingcheng Sun
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xun Chen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Zheng Ouyang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing 100084, China
| | - Jiahong Dong
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, 102218, China
| | - Yunfang Wang
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, 102218, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Shuhua Yue
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
41
|
Celik C, Lee SYT, Yap WS, Thibault G. Endoplasmic reticulum stress and lipids in health and diseases. Prog Lipid Res 2023; 89:101198. [PMID: 36379317 DOI: 10.1016/j.plipres.2022.101198] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022]
Abstract
The endoplasmic reticulum (ER) is a complex and dynamic organelle that regulates many cellular pathways, including protein synthesis, protein quality control, and lipid synthesis. When one or multiple ER roles are dysregulated and saturated, the ER enters a stress state, which, in turn, activates the highly conserved unfolded protein response (UPR). By sensing the accumulation of unfolded proteins or lipid bilayer stress (LBS) at the ER, the UPR triggers pathways to restore ER homeostasis and eventually induces apoptosis if the stress remains unresolved. In recent years, it has emerged that the UPR works intimately with other cellular pathways to maintain lipid homeostasis at the ER, and so does at cellular levels. Lipid distribution, along with lipid anabolism and catabolism, are tightly regulated, in part, by the ER. Dysfunctional and overwhelmed lipid-related pathways, independently or in combination with ER stress, can have reciprocal effects on other cellular functions, contributing to the development of diseases. In this review, we summarize the current understanding of the UPR in response to proteotoxic stress and LBS and the breadth of the functions mitigated by the UPR in different tissues and in the context of diseases.
Collapse
Affiliation(s)
- Cenk Celik
- School of Biological Sciences, Nanyang Technological University, Singapore
| | | | - Wei Sheng Yap
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Guillaume Thibault
- School of Biological Sciences, Nanyang Technological University, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore.
| |
Collapse
|
42
|
William S, Duncan T, Redmond TM. Pretreatment of human retinal pigment epithelial cells with sterculic acid forestalls fenretinide-induced apoptosis. Sci Rep 2022; 12:22442. [PMID: 36575190 PMCID: PMC9794835 DOI: 10.1038/s41598-022-26383-9] [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: 08/17/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
The ratio of saturated to monounsaturated fatty acids, thought to play a critical role in many cellular functions, is regulated by stearoyl-CoA desaturase (SCD), a rate-limiting enzyme in the biosynthesis of monounsaturated fatty acids. Previously, we observed a decrease in both SCD protein and enzymatic activity in apoptosis induced by fenretinide, a synthetic analog of retinoic acid, in the human retinal pigment epithelial (RPE) cell line ARPE-19. Here, we investigated the effect of pretreating ARPE-19 with sterculic acid, a cyclopropenoic fatty acid inhibitor of SCD, on preventing fenretinide-induced apoptosis, given the role of SCD in cell proliferation and apoptosis. We show that sterculic acid pretreatment prevents the effects of fenretinide-induced apoptosis shown by changes in cell morphology, viability, and caspase-3 activation. Analysis of endoplasmic reticulum (ER)-associated proteins shows that sterculic acid pretreatment reduced the fenretinide-induced upregulation of heme oxygenase-1, ATF3 and GADD153 expression that are in response to reactive oxygen species (ROS) generation. Sterculic acid is as effective as allopurinol in inhibition of xanthine oxidase (XDH), and this may play a role in reducing the potential role of XDH in fenretinide-induced ROS generation. Sterculic acid pretreatment also results in a reduction in SOD2 mRNA expression. Dihydroceramide accumulation, compared to ceramide, and ROS generation indicate that a ceramide-independent pathway mediates fenretinide-induced apoptosis, and ROS mediation is borne out by activation of the NF-κBp50 and NF-κBp65 downstream signaling cascade. Its prevention by sterculic acid pretreatment further indicates the latter's antioxidant/anti-inflammatory effect. Taken together, our results suggest that sterculic acid pretreatment can mitigate ROS-mediated fenretinide-induced apoptosis. Thus, sterculic acid may serve as a potential antioxidant and therapeutic agent. These effects may be independent of its effects on SCD activity.
Collapse
Affiliation(s)
- Samuel William
- grid.280030.90000 0001 2150 6316Laboratory of Retinal Cell and Molecular Biology, Bldg. 6/Room 112A, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892-0608 USA
| | - Todd Duncan
- grid.280030.90000 0001 2150 6316Laboratory of Retinal Cell and Molecular Biology, Bldg. 6/Room 112A, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892-0608 USA
| | - T. Michael Redmond
- grid.280030.90000 0001 2150 6316Laboratory of Retinal Cell and Molecular Biology, Bldg. 6/Room 112A, National Eye Institute, National Institutes of Health, 6 Center Drive, Bethesda, MD 20892-0608 USA
| |
Collapse
|
43
|
Day ZI, Mayfosh AJ, Giel MC, Hong Y, Williams SA, Santavanond JP, Rau TF, Poon IK, Hulett MD. Novel Formulation of Undecylenic Acid induces Tumor Cell Apoptosis. Int J Mol Sci 2022; 23:ijms232214170. [PMID: 36430646 PMCID: PMC9692760 DOI: 10.3390/ijms232214170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/28/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
Undecylenic acid, a monounsaturated fatty acid, is currently in clinical use as a topical antifungal agent, however the potential for therapeutic application in other disease settings has not been investigated. In this study, we describe a novel platform for the solubilization of fatty acids using amino acids and utilize this approach to define a tumoricidal activity and underlying mechanism for undecylenic acid. We examined a novel formulation of undecylenic acid compounded with L-Arginine, called GS-1, that induced concentration-dependent tumor cell death, with undecylenic acid being the cytotoxic component. Further investigation revealed that GS-1-mediated cell death was caspase-dependent with a reduction in mitochondrial membrane potential, suggesting a pro-apoptotic mechanism of action. Additionally, GS-1 was found to localize intracellularly to lipid droplets. In contrast to previous studies where lipid droplets have been shown to be protective against fatty acid-induced cell death, we showed that lipid droplets could not protect against GS-1-induced cytotoxicity. We also found a role for Fatty Acid Transport Protein 2 (FATP2) in the uptake of this compound. Collectively, this study demonstrates that GS-1 has effective pro-apoptotic antitumor activity in vitro and, together with the novel platform of fatty acid solubilization, contributes to the re-emerging field of fatty acids as potential anti-cancer therapeutics.
Collapse
Affiliation(s)
- Zoe I. Day
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Alyce J. Mayfosh
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
- Wintermute Biomedical, 789 Bauer Lane, Corvallis, MT 59828, USA
| | - Marie-Claire Giel
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Yuning Hong
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Scott A. Williams
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Jascinta P. Santavanond
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Thomas F. Rau
- Wintermute Biomedical, 789 Bauer Lane, Corvallis, MT 59828, USA
| | - Ivan K. Poon
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Mark D. Hulett
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
- Correspondence: ; Tel.: +61-9479-1266
| |
Collapse
|
44
|
Huang LJ, Chen RH. Lipid saturation induces degradation of squalene epoxidase for sterol homeostasis and cell survival. Life Sci Alliance 2022; 6:6/1/e202201612. [PMID: 36368908 PMCID: PMC9652772 DOI: 10.26508/lsa.202201612] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022] Open
Abstract
A fluid membrane containing a mix of unsaturated and saturated lipids is essential for life. However, it is unclear how lipid saturation might affect lipid homeostasis, membrane-associated proteins, and membrane organelles. Here, we generate temperature-sensitive mutants of the sole fatty acid desaturase gene OLE1 in the budding yeast Saccharomyces cerevisiae Using these mutants, we show that lipid saturation triggers the endoplasmic reticulum-associated degradation (ERAD) of squalene epoxidase Erg1, a rate-limiting enzyme in sterol biosynthesis, via the E3 ligase Doa10-Ubc7 complex. We identify the P469L mutation that abolishes the lipid saturation-induced ERAD of Erg1. Overexpressed WT or stable Erg1 mutants all mislocalize into foci in the ole1 mutant, whereas the stable Erg1 causes aberrant ER and severely compromises the growth of ole1, which are recapitulated by doa10 deletion. The toxicity of the stable Erg1 and doa10 deletion is due to the accumulation of lanosterol and misfolded proteins in ole1 Our study identifies Erg1 as a novel lipid saturation-regulated ERAD target, manifesting a close link between lipid homeostasis and proteostasis that maintains sterol homeostasis under the lipid saturation condition for cell survival.
Collapse
Affiliation(s)
| | - Rey-Huei Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
45
|
Cherubini A, Zito E. ER stress as a trigger of UPR and ER-phagy in cancer growth and spread. Front Oncol 2022; 12:997235. [PMID: 36408145 PMCID: PMC9667062 DOI: 10.3389/fonc.2022.997235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/12/2022] [Indexed: 01/25/2023] Open
Abstract
Tumors can survive environmental and metabolic stress by triggering homeostatic responses that re-establish the pre-stress status and permit them to grow and thrive. The endoplasmic reticulum (ER) is the organelle where proteins undergo post-translational modifications and are folded and exported to the secretory pathway. Its environment and activity are therefore fundamental for proteostasis, i.e., the plethora of mechanisms controlling protein formation, folding, degradation, and secretion, needed to assure protein balance and cellular health. In different tumor-related conditions, such as after the activation of oncogenes or under hypoxia and nutrient deprivation, the ER experiences stress, triggered by a high load of proteins to be folded compared to the limited folding capacity of the organelle. As a consequence, three ER membrane sensors and the related unfolded protein response (UPR) are activated. The UPR comprises a complex interconnection between signal transduction pathways that promote a homeostatic response that acts by increasing the amount of protein chaperones and of proteins involved in ER-associated protein degradation (ERAD) on one hand and attenuating protein translation on the other. ER-phagy, literally "eating" the ER, is part of another homeostatic response consisting of the clearance of non-functional ER portions including misfolded proteins. This response is also activated by a set of dedicated ER-phagy receptors after ER stimuli, which overlap the stimuli generating ER stress. Thus, the UPR and ER-phagy are two closely related homeostatic mechanisms that cooperate in re-establishing ER homeostasis. However, while the role of the UPR in favoring cancer growth and thriving by promoting angiogenesis, metastasis, chemotherapy resistance, and epithelial-to-mesenchymal transition is consolidated, that of ER-phagy is still in its infancy. This essay provides an overview of emerging concepts on ER stress, the UPR, and ER-phagy and their crosstalk in tumorigenesis. We also critically review new findings on their pharmacological targeting in cancer.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy,Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy,Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy,*Correspondence: Ester Zito,
| |
Collapse
|
46
|
Lpcat3 deficiency promotes palmitic acid-induced 3T3-L1 mature adipocyte inflammation through enhanced ROS generation. Acta Biochim Biophys Sin (Shanghai) 2022; 55:117-130. [PMID: 36331295 PMCID: PMC10157521 DOI: 10.3724/abbs.2022161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphatidylcholines (PCs) are major phospholipids in the mammalian cell membrane. Structural remodeling of PCs is associated with many biological processes. Lysophosphatidylcholine acyltransferase 3 (Lpcat3), which catalyzes the incorporation of polyunsaturated fatty acyl chains into the sn-2 site of PCs, plays an important role in maintaining plasma membrane fluidity. Adipose tissue is one of the main distribution organs of Lpcat3, while the relationship between Lpcat3 and adipose tissue dysfunction during overexpansion remains unknown. In this study, we reveal that both polyunsaturated PC content and Lpcat3 expression are increased in abdominal adipose tissues of high-fat diet-fed mice when compared with chow-diet-fed mice, indicating that Lpcat3 is involved in adipose tissue overexpansion and dysfunction. Our experiments in 3T3-L1 adipocytes show that inhibition of Lpcat3 does not change triglyceride accumulation but increases palmitic acid-induced inflammation and lipolysis. Conversely, Lpcat3 overexpression exhibits anti-inflammatory and anti-lipolytic effects. Furthermore, mechanistic studies demonstrate that Lpcat3 deficiency promotes reactive oxygen species (ROS) generation by increasing NOX enzyme activity by facilitating the translocation of NOX4 to lipid rafts, thereby aggregating 3T3-L1 adipocyte inflammation induced by palmitic acid. Moreover, overexpression of Lpcat3 exhibits the opposite effects. These findings suggest that Lpcat3 protects adipocytes from inflammation during adipose tissue overexpansion by reducing ROS generation. In conclusion, our study demonstrates that Lpcat3 deficiency promotes palmitic acid-induced inflammation in 3T3-L1 adipocytes by enhancing ROS generation.
Collapse
|
47
|
Taskinen JH, Ruhanen H, Matysik S, Käkelä R, Olkkonen VM. Global effects of pharmacologic inhibition of OSBP in human umbilical vein endothelial cells. Steroids 2022; 185:109053. [PMID: 35623602 DOI: 10.1016/j.steroids.2022.109053] [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: 12/17/2021] [Revised: 04/14/2022] [Accepted: 05/23/2022] [Indexed: 11/26/2022]
Abstract
Oxysterol-binding protein (OSBP) is a cholesterol/PI4P exchanger at contacts of the endoplasmic reticulum (ER) with trans-Golgi network (TGN) and endosomes. Several central endothelial cell (EC) functions depend on adequate cholesterol distribution in cellular membranes. Here we elucidated the effects of pharmacologic OSBP inhibition on the lipidome and transcriptome of human umbilical vein endothelial cells (HUVECs). OSBP was inhibited for 24 h with 25 nM Schweinfurthin G (SWG) or Orsaponin (OSW-1), followed by analyses of cellular cholesterol, 27-hydroxy-cholesterol, and triacylglycerol concentration, phosphatidylserine synthesis rate, the lipidome, as well as lipid droplet staining and western analysis of OSBP protein. Next-generation RNA sequencing of the SWG-treated and control HUVECs and angiogenesis assays were performed. Protein-normalized lipidomes of the inhibitor-treated cells revealed decreases in glycerophospholipids, the most pronounced effect being on phosphatidylserines and the rate of their synthesis, as well as increases in cholesteryl esters, triacylglycerols and lipid droplet number. Transcriptome analysis of SWG-treated cells suggested ER stress responses apparently caused by disturbed cholesterol exit from the ER, as indicated by suppression of cholesterol biosynthetic genes. OSBP was associated with the TGN in the absence of inhibitors and disappeared therefrom in inhibitor-treated cells in a time-dependent manner, coinciding with OSBP reduction on western blots. Prolonged treatment with SWG or OSW-1 inhibited angiogenesis in vitro. To conclude, inhibition of OSBP in primary endothelial cells induced multiple effects on the lipidome, transcriptome changes suggesting ER stress, and disruption of in vitro angiogenic capacity. Thus, OSBP is a crucial regulator of EC lipid homeostasis and angiogenic capacity.
Collapse
Affiliation(s)
- Juuso H Taskinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland.
| | - Hanna Ruhanen
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Molecular and Integrative Biosciences Research Programme, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014 University of Helsinki, Finland.
| | - Silke Matysik
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany.
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Molecular and Integrative Biosciences Research Programme, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014 University of Helsinki, Finland.
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland.
| |
Collapse
|
48
|
Katoh Y, Yaguchi T, Kubo A, Iwata T, Morii K, Kato D, Ohta S, Satomi R, Yamamoto Y, Oyamada Y, Ouchi K, Takahashi S, Ishioka C, Matoba R, Suematsu M, Kawakami Y. Inhibition of stearoyl-CoA desaturase 1 (SCD1) enhances the antitumor T cell response through regulating β-catenin signaling in cancer cells and ER stress in T cells and synergizes with anti-PD-1 antibody. J Immunother Cancer 2022; 10:jitc-2022-004616. [PMID: 35793868 PMCID: PMC9260842 DOI: 10.1136/jitc-2022-004616] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 11/26/2022] Open
Abstract
Background Understanding the mechanisms of non-T cell inflamed tumor microenvironment (TME) and their modulation are important to improve cancer immunotherapies such as immune checkpoint inhibitors. The involvement of various immunometabolisms has recently been indicated in the formation of immunosuppressive TME. In this study, we investigated the immunological roles of stearoyl-CoA desaturase 1 (SCD1), which is essential for fatty acid metabolism, in the cancer immune response. Methods We investigated the roles of SCD1 by inhibition with the chemical inhibitor or genetic manipulation in antitumor T cell responses and the therapeutic effect of anti-programmed cell death protein 1 (anti-PD-1) antibody using various mouse tumor models, and their cellular and molecular mechanisms. The roles of SCD1 in human cancers were also investigated by gene expression analyses of colon cancer tissues and by evaluating the related free fatty acids in sera obtained from patients with non-small cell lung cancer who were treated with anti-PD-1 antibody. Results Systemic administration of a SCD1 inhibitor in mouse tumor models enhanced production of CCL4 by cancer cells through reduction of Wnt/β-catenin signaling and by CD8+ effector T cells through reduction of endoplasmic reticulum stress. It in turn promoted recruitment of dendritic cells (DCs) into the tumors and enhanced the subsequent induction and tumor accumulation of antitumor CD8+ T cells. SCD1 inhibitor was also found to directly stimulate DCs and CD8+ T cells. Administration of SCD1 inhibitor or SCD1 knockout in mice synergized with an anti-PD-1 antibody for its antitumor effects in mouse tumor models. High SCD1 expression was observed in one of the non-T cell-inflamed subtypes in human colon cancer, and serum SCD1 related fatty acids were correlated with response rates and prognosis of patients with non-small lung cancer following anti-PD-1 antibody treatment. Conclusions SCD1 expressed in cancer cells and immune cells causes immunoresistant conditions, and its inhibition augments antitumor T cells and therapeutic effects of anti-PD-1 antibody. Therefore, SCD1 is an attractive target for the development of new diagnostic and therapeutic strategies to improve current cancer immunotherapies including immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Yuki Katoh
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, Tokyo, Japan
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Akiko Kubo
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Iwata
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Morii
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Daiki Kato
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.,Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shigeki Ohta
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Ryosuke Satomi
- National Hospital Organisation Tokyo Medical Center, Tokyo, Japan
| | - Yasuhiro Yamamoto
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Kota Ouchi
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan.,Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shin Takahashi
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan.,Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chikashi Ishioka
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan.,Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan .,Department of Immunology, International University of Health and Welfare, Chiba, Japan
| |
Collapse
|
49
|
Tiszlavicz Á, Gombos I, Péter M, Hegedűs Z, Hunya Á, Dukic B, Nagy I, Peksel B, Balogh G, Horváth I, Vígh L, Török Z. Distinct Cellular Tools of Mild Hyperthermia-Induced Acquired Stress Tolerance in Chinese Hamster Ovary Cells. Biomedicines 2022; 10:1172. [PMID: 35625909 PMCID: PMC9138356 DOI: 10.3390/biomedicines10051172] [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: 04/12/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/20/2022] Open
Abstract
Mild stress could help cells to survive more severe environmental or pathophysiological conditions. In the current study, we investigated the cellular mechanisms which contribute to the development of stress tolerance upon a prolonged (0-12 h) fever-like (40 °C) or a moderate (42.5 °C) hyperthermia in mammalian Chinese Hamster Ovary (CHO) cells. Our results indicate that mild heat triggers a distinct, dose-dependent remodeling of the cellular lipidome followed by the expression of heat shock proteins only at higher heat dosages. A significant elevation in the relative concentration of saturated membrane lipid species and specific lysophosphatidylinositol and sphingolipid species suggests prompt membrane microdomain reorganization and an overall membrane rigidification in response to the fluidizing heat in a time-dependent manner. RNAseq experiments reveal that mild heat initiates endoplasmic reticulum stress-related signaling cascades resulting in lipid rearrangement and ultimately in an elevated resistance against membrane fluidization by benzyl alcohol. To protect cells against lethal, protein-denaturing high temperatures, the classical heat shock protein response was required. The different layers of stress response elicited by different heat dosages highlight the capability of cells to utilize multiple tools to gain resistance against or to survive lethal stress conditions.
Collapse
Affiliation(s)
- Ádám Tiszlavicz
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - Imre Gombos
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - Mária Péter
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - Zoltán Hegedűs
- Core Facilities, Biological Research Centre, 6726 Szeged, Hungary; (Z.H.); (I.N.)
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Ákos Hunya
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - Barbara Dukic
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - István Nagy
- Core Facilities, Biological Research Centre, 6726 Szeged, Hungary; (Z.H.); (I.N.)
- Seqomics Biotechnology Ltd., 6782 Mórahalom, Hungary
| | - Begüm Peksel
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - Gábor Balogh
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - Ibolya Horváth
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - László Vígh
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| | - Zsolt Török
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary; (Á.T.); (I.G.); (M.P.); (Á.H.); (B.D.); (B.P.); (G.B.); (I.H.); (L.V.)
| |
Collapse
|
50
|
Kanehara K, Cho Y, Yu CY. A lipid viewpoint on the plant endoplasmic reticulum stress response. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:2835-2847. [PMID: 35560195 DOI: 10.1093/jxb/erac063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/15/2022] [Indexed: 06/15/2023]
Abstract
Organisms, including humans, seem to be constantly exposed to various changes, which often have undesirable effects, referred to as stress. To keep up with these changes, eukaryotic cells may have evolved a number of relevant cellular processes, such as the endoplasmic reticulum (ER) stress response. Owing to presumably intimate links between human diseases and the ER function, the ER stress response has been extensively investigated in various organisms for a few decades. Based on these studies, we now have a picture of the molecular mechanisms of the ER stress response, one of which, the unfolded protein response (UPR), is highly conserved among yeasts, mammals, higher plants, and green algae. In this review, we attempt to highlight the plant UPR from the perspective of lipids, especially membrane phospholipids. Phosphatidylcholine (PtdCho) and phosphatidylethanolamine (PtdEtn) are the most abundant membrane phospholipids in eukaryotic cells. The ratio of PtdCho to PtdEtn and the unsaturation of fatty acyl tails in both phospholipids may be critical factors for the UPR, but the pathways responsible for PtdCho and PtdEtn biosynthesis are distinct in animals and plants. We discuss the plant UPR in comparison with the system in yeasts and animals in the context of membrane phospholipids.
Collapse
Affiliation(s)
- Kazue Kanehara
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Yueh Cho
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Chao-Yuan Yu
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, 11529, Taiwan
| |
Collapse
|