1
|
Ahn S, Kaipparettu BA. G-protein coupled receptors in metabolic reprogramming and cancer. Pharmacol Ther 2025; 270:108849. [PMID: 40204142 DOI: 10.1016/j.pharmthera.2025.108849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/09/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
G-protein coupled receptors (GPCR) are one of the frequently investigated drug targets. GPCRs are involved in many human pathophysiologies that lead to various disease conditions, such as cancer, diabetes, and obesity. GPCR receptor activates multiple signaling pathways depending on the ligand and tissue type. However, this review will be limited to the GPCR-mediated metabolic modulations and the activation of relevant signaling pathways in cancer therapy. Cancer cells often have reprogrammed cell metabolism to support tumor growth and metastatic plasticity. Many aggressive cancer cells maintain a hybrid metabolic status, using both glycolysis and mitochondrial metabolism for better metabolic plasticity. In addition to glucose and glutamine pathways, fatty acid is a key mitochondrial energy source in some cancer subtypes. Recently, targeting alternative energy pathways like fatty acid beta-oxidation (FAO) has attracted great interest in cancer therapy. Several in vitro and in vivo experiments in different cancer models reported encouraging responses to FAO inhibitors. However, due to the potential liver toxicity of FAO inhibitors in clinical trials, new approaches to indirectly target metabolic reprogramming are necessary for in vivo targeting of cancer cells. This review specifically focused on free fatty acid receptors (FFAR) and β-adrenergic receptors (β-AR) because of their reported significance in mitochondrial metabolism and cancer. Further understanding the pharmacology of GPCRs and their role in cancer metabolism will help repurpose GPCR-targeting drugs for cancer therapy and develop novel drug discovery strategies to combine them with standard cancer therapy to increase anticancer potential and overcome drug resistance.
Collapse
Affiliation(s)
- Songyeon Ahn
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Benny Abraham Kaipparettu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
Dei Cas M, Mantovani S, Oliviero B, Zulueta A, Montavoci L, Falleni M, Tosi D, Morano C, Penati S, Chiocchetti A, Sinella R, Barbero Mazzucca C, Donadon M, Soldani C, Piccolo G, Barabino M, Bianchi PP, Maestri M, Lleo A, Banales JM, Mondelli MU, Caretti A. Cholangiocarcinoma cells direct fatty acids to support membrane synthesis and modulate macrophage phenotype. Hepatol Commun 2025; 9:e0717. [PMID: 40408281 DOI: 10.1097/hc9.0000000000000717] [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: 09/05/2024] [Accepted: 03/01/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND AND AIMS Cholangiocarcinoma (CCA) is a globally rare, increasingly incident cancer. Metabolic reprogramming is common in cancer cells, and altered lipid homeostasis favors tumor development and progression. Previous studies have described lipid deregulation in HCC cells, while in CCA, the lipidome profile is still poorly characterized. METHODS We used liquid chromatography-tandem mass spectrometry to examine the lipid level profile of intrahepatic CCA (iCCA) and non-tumor surrounding tissue from patients, as well as in patients' and healthy controls' sera. RESULTS All lipid classes were upregulated in tumor specimens and iCCA-derived sera. Newly synthesized fatty acids (FAs) accumulated in iCCA and were only marginally directed to mitochondrial β-oxidation and scarcely folded in lipid droplets as neutral species. Metabolic flux assay showed that FAs were instead redirected toward plasma membrane formation and remodeling, being incorporated into phospholipids and sphingomyelin. A distinct lipid droplet and macrophage distribution was revealed by immunohistochemistry and Imaging Mass Cytometry. Lipid droplets were fewer in iCCA than in normal tissue and present mainly in the intratumoral fibrous septa and in M2 macrophages. Monocytes modified their lipid content and phenotype in the presence of iCCA cells, and the same effect could be recapitulated by FA supplementation. CONCLUSIONS Our results reveal a profound alteration in the lipid content of iCCA tissues and demonstrate that FA accumulation prompts iCCA aggressiveness by supporting membrane biogenesis, generating bioactive lipids that boost proliferation, and by modifying macrophage phenotype.
Collapse
Affiliation(s)
- Michele Dei Cas
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Stefania Mantovani
- Research Department, Division of Clinical Immunology-Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Barbara Oliviero
- Research Department, Division of Clinical Immunology-Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Aida Zulueta
- Istituti Clinici Scientifici Maugeri IRCCS, Neurorehabilitation Unit of Milan Institute, Milan, Italy
| | - Linda Montavoci
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Monica Falleni
- Health Sciences Department, Pathology Division, University of Milan, Milan, Italy
| | - Delfina Tosi
- Health Sciences Department, Pathology Division, University of Milan, Milan, Italy
| | - Camillo Morano
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Sara Penati
- Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Riccardo Sinella
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | | | - Matteo Donadon
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Department of Surgery, University Maggiore Hospital della Carità, Novara, Italy
| | - Cristiana Soldani
- Laboratory of Hepatobiliary Immunopathology, IRCCS Humanitas Research Hospital, Rozzano, Milan
| | - Gaetano Piccolo
- Department of Health Sciences, General Surgery Unit, University of Milan, Milan, Italy
| | - Matteo Barabino
- Department of Health Sciences, General Surgery Unit, University of Milan, Milan, Italy
| | - Paolo Pietro Bianchi
- Department of Health Sciences, General Surgery Unit, University of Milan, Milan, Italy
| | - Marcello Maestri
- Division of General Surgery 1, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute-Donostia University Hospital, University of the Basque Country (UPV/EHU), Ikerbasque, San Sebastian, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Mario U Mondelli
- Research Department, Division of Clinical Immunology-Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Anna Caretti
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
3
|
Inoue FSR, Concato-Lopes VM, Bortoleti BTDS, Cruz EMS, Detoni MB, Tomiotto-Pellissier F, Gonçalves-Lens MD, Morais-Valentim JMBD, Machado RRB, Santiago-Silva KM, Bispo MDLF, Schirmann JG, Barbosa-Dekker AM, Dekker RFH, Assis MCTD, Conchon-Costa I, Mantovani MS, Lazarin-Bidóia D, Panis C, Pavanelli WR. 3,3',5,5'-Tetramethoxybiphenyl-4,4'-diol exerts a cytotoxic effect on hepatocellular carcinoma cell lines by inducing morphological and ultrastructural alterations, G2/M cell cycle arrest and death by apoptosis via CDK1 interaction. Biomed Pharmacother 2025; 187:118082. [PMID: 40280030 DOI: 10.1016/j.biopha.2025.118082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignancy with frequent recurrence and chemoresistance, underscoring the need for new treatment strategies. 3,3',5,5'-Tetramethoxybiphenyl-4,4'-diol (TMBP) showed cytotoxicity against lung cancer cell lines without harming normal cells. Thus, we investigated the antitumoral effect of TMBP on HCC cell lines, HuH7.5 (p53-mutant) and HepG2/C3A (p53-wild type). Cells were treated with TMBP (12.5-150 µM) for 24 and 48 h, and metabolic cellular activity (MTT) were used to determine the 50 % inhibitory concentration (IC50) values. TMBP cytotoxicity were assessed by Trypan blue assay, scanning and transmission electron microscopy. Cell migration (wound healing), total ROS (H2DCFDA), mitochondrial dysfunction (TMRE), lipid droplets (Nile Red), and autophagic vacuoles (MDC) were assessed. Flow cytometry characterized cell cycle distribution and cell death. Caspase 3/7 activity and CASP3 expression confirmed apoptosis. Molecular docking and gene expression analysis validated TMBP-CDK1 interaction. TMBP reduced cell viability, with IC50 values of 68 and 55 µM (HuH7.5) and 50 and 42 µM (HepG2/C3A) at 24 and 48 h. TMBP induced severe morphological alterations, impaired migration, increased ROS, mitochondrial dysfunction, increased lipid droplets and autophagic vacuoles. TMBP also led to G2/M arrest and apoptosis, likely via CDK1 inhibition through hydrogen bonding at Tyr15. These findings highlight TMBP as a promising therapeutic candidate targeting CDK1 in HCC.
Collapse
Affiliation(s)
- Fabricio Seidy Ribeiro Inoue
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil.
| | - Virginia Marcia Concato-Lopes
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil
| | - Bruna Taciane da Silva Bortoleti
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil
| | - Ellen Mayara Souza Cruz
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil
| | - Mariana Barbosa Detoni
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil
| | - Fernanda Tomiotto-Pellissier
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil; Department of Medical Pathology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Manoela Daiele Gonçalves-Lens
- Laboratory of Biotransformation and Phytochemical, Department of Chemistry, State University of Londrina, Londrina, PR, Brazil
| | - Juliana Maria Bitencourt de Morais-Valentim
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil
| | - Rayanne Regina Beltrame Machado
- Laboratory of Technological Innovation in the Development of Drugs and Cosmetics, Department of Basic Health Sciences, State University of Maringá, Maringá, PR, Brazil
| | - Kaio Maciel Santiago-Silva
- Laboratório de Síntese de Moléculas Medicinais (LaSMMed), Department of Chemistry, State University of Londrina, Londrina, PR, Brazil
| | - Marcelle de Lima Ferreira Bispo
- Laboratório de Síntese de Moléculas Medicinais (LaSMMed), Department of Chemistry, State University of Londrina, Londrina, PR, Brazil
| | - Jéseka Gabriela Schirmann
- Laboratory of Research of Bioactive Molecules, Department of Chemistry, State University of Londrina, Londrina, PR, Brazil
| | - Aneli M Barbosa-Dekker
- Laboratory of Research of Bioactive Molecules, Department of Chemistry, State University of Londrina, Londrina, PR, Brazil; Beta-Glucan Produtos Farmoquímicos-EIRELI, Lote 24A - Bloco Zircônia, Universidade Tecnológica Federal do Paraná, Londrina, PR CEP: 86036-700, Brazil
| | - Robert F H Dekker
- Beta-Glucan Produtos Farmoquímicos-EIRELI, Lote 24A - Bloco Zircônia, Universidade Tecnológica Federal do Paraná, Londrina, PR CEP: 86036-700, Brazil
| | | | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil
| | - Mário Sérgio Mantovani
- Laboratory of Toxicological Genetics, Department of General Biology, State University of Londrina, Londrina, PR, Brazil
| | - Danielle Lazarin-Bidóia
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil; Laboratory of Technological Innovation in the Development of Drugs and Cosmetics, Department of Basic Health Sciences, State University of Maringá, Maringá, PR, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, Center of Health Sciences, State University of Western Parana, Francisco Beltrão, PR, Brazil
| | - Wander Rogério Pavanelli
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina, PR, Brazil
| |
Collapse
|
4
|
Liu C, He S, Guo XF, Wang H. A novel near-infrared AIE probe for sensitive imaging of lipid droplet and dual-parameter cancer diagnosis. Anal Chim Acta 2025; 1352:343916. [PMID: 40210274 DOI: 10.1016/j.aca.2025.343916] [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: 01/09/2025] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Lipid droplets (LDs) are vital intracellular organelles for lipid storage, closely associated with various metabolic disorders and cancers. Fluorescence imaging offers a powerful, non-invasive approach to study LDs in real time, but many existing probes suffer from non-specific staining and aggregation-caused quenching (ACQ), compromising their imaging specificity and contrast. RESULT In this study, we synthesized a novel LD fluorescent probe TPC-AN that takes advantage of near-infrared emission, large Stokes shift, high lipophilicity, polarity response and aggregation-induced emission (AIE) characteristics. TPC-AN effectively addresses issues of non-specific staining and ACQ commonly observed with traditional probes, enabling highly specific and high-contrast imaging of LDs. Utilizing TPC-AN, imaging of LDs in several kinds of cells was performed, and discrimination of cancerous and normal cells was achieved using dual-parameter through differences in LD fluorescence area and intensity. SIGNIFICANCE This work provides a promising tool for studying LDs in diseases and offers a reliable method for cancer diagnosis, with excellent LD-specificity, low cytotoxicity, and dual-parameter imaging capabilities.
Collapse
Affiliation(s)
- Cong Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Shan He
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiao-Feng Guo
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Hong Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
5
|
Liu L, Wang H, Chen R, Song Y, Wei W, Baek D, Gillin M, Kurabayashi K, Chen W. Cancer-on-a-chip for precision cancer medicine. LAB ON A CHIP 2025. [PMID: 40376718 DOI: 10.1039/d4lc01043d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Many cancer therapies fail in clinical trials despite showing potent efficacy in preclinical studies. One of the key reasons is the adopted preclinical models cannot recapitulate the complex tumor microenvironment (TME) and reflect the heterogeneity and patient specificity in human cancer. Cancer-on-a-chip (CoC) microphysiological systems can closely mimic the complex anatomical features and microenvironment interactions in an actual tumor, enabling more accurate disease modeling and therapy testing. This review article concisely summarizes and highlights the state-of-the-art progresses in CoC development for modeling critical TME compartments including the tumor vasculature, stromal and immune niche, as well as its applications in therapying screening. Current dilemma in cancer therapy development demonstrates that future preclinical models should reflect patient specific pathophysiology and heterogeneity with high accuracy and enable high-throughput screening for anticancer drug discovery and development. Therefore, CoC should be evolved as well. We explore future directions and discuss the pathway to develop the next generation of CoC models for precision cancer medicine, such as patient-derived chip, organoids-on-a-chip, and multi-organs-on-a-chip with high fidelity. We also discuss how the integration of sensors and microenvironmental control modules can provide a more comprehensive investigation of disease mechanisms and therapies. Next, we outline the roadmap of future standardization and translation of CoC technology toward real-world applications in pharmaceutical development and clinical settings for precision cancer medicine and the practical challenges and ethical concerns. Finally, we overview how applying advanced artificial intelligence tools and computational models could exploit CoC-derived data and augment the analytical ability of CoC.
Collapse
Affiliation(s)
- Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - Huishu Wang
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - Ruiqi Chen
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Yujing Song
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - William Wei
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - David Baek
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Mahan Gillin
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Katsuo Kurabayashi
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
- Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
6
|
Lv Y, Jin H, Liu Z, Li N, Liao YX, Shen J, Hou JT. A polarity-sensitive fluorescent probe for visualizing lipid droplets in ferroptosis, cuproptosis, and autophagy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 332:125854. [PMID: 39922069 DOI: 10.1016/j.saa.2025.125854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/12/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Dynamics of lipid droplets (LDs) in various pathological processes provides important information about lipid metabolism during theses biological processes, while only a few reports focused on this field. In this work, a benzothiazine-fused coumarin chromophore BCLD with strong fluorescence in low-polarity environment is described. It is confirmed that cyclization-induced rigidification might be a promising approach to enhance the LDs specificity of phenothiazine-based strucutres.The probe is found to enter cells through a clathrin-mediated endocytosis, and is able to monitor LDs variations in living cells, especially during various pathological processes. It is found that obvious increase in polarity of LDs during ferroptosis and cuproptosis was visualized while a dramatic decrease in the number of LDs was recorded during autophagy, indicating different lipid metabolism manners and LD dynamics in these pathological processes. This work supports the potentials of LDs as markers for drug design targeting ferroptosis, cuproptosis, and autophagy.
Collapse
Affiliation(s)
- Yang Lv
- School of Chemistry and Chemical Engineering, Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Minzu University, Nanning 530008 China
| | - Haoyu Jin
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027 China
| | - Zhe Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027 China
| | - Na Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027 China
| | - Ye-Xin Liao
- School of Chemistry and Chemical Engineering, Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Minzu University, Nanning 530008 China.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027 China; Zhejiang Engineering Research Center for Tissue Repair Materials, University of Chinese Academy of Sciences, Wenzhou 325001 China.
| | - Ji-Ting Hou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027 China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000 China.
| |
Collapse
|
7
|
Michurina S, Goltseva Y, Ratner E, Dergilev K, Shestakova E, Minniakhmetov I, Rumyantsev S, Stafeev I, Shestakova M, Parfyonova Y. Artificial intelligence-enabled lipid droplets quantification: Comparative analysis of NIS-elements Segment.ai and ZeroCostDL4Mic StarDist networks. Methods 2025; 237:9-18. [PMID: 40023351 DOI: 10.1016/j.ymeth.2025.02.013] [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: 11/08/2024] [Revised: 01/29/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Lipid droplets (LDs) are dynamic organelles that are present in almost all cell types, with a particularly high prevalence in adipocytes. The phenotype of LDs in these cells reflects their maturity, metabolic activity and function. Although LDs quantification in adipocytes is significant for understanding the origins of obesity and associated complications, it remains challenging and requires the implementation of computer science innovations. This article outlines a practical workflow for application of Segment.ai neural network from the commercial software NIS-Elements and the open-source StarDist Jupyter notebook from the ZeroCostDL4Mic platform for the analysis of LDs number and morphology. To generate a training dataset, 3T3-L1 cells were differentiated into adipocytes and stained with lipophilic dye BODIPY493/503. Subsequently, confocal live cell images were acquired, annotated and used for training. As an example task, deep learning models were tested on their ability to detect LDs enlargement on images of adipocytes with inhibited lipolysis. We demonstrated that both Segment.ai and StarDist models are capable of accurately recognising LDs on microphotographs, thereby significantly accelerating the processing of imaging data. The advantage of the Segment.ai model is its integration into NIS-Elements General Analysis 3, which performs quantitative and statistical data interpretation. Alternatively, StarDist is a more accessible and transparent tool, enabling precise model evaluation. In conclusion, both created approaches have the potential to accelerate the exploration of LDs dynamics, thus paving the way for further insights into how these organelles regulate energy homeostasis and contribute to the development of metabolic abnormalities.
Collapse
Affiliation(s)
- S Michurina
- National Medical Research Centre for Cardiology Named After Academician E.I.Chazov, Moscow, Russia
| | - Y Goltseva
- National Medical Research Centre for Cardiology Named After Academician E.I.Chazov, Moscow, Russia
| | - E Ratner
- National Medical Research Centre for Cardiology Named After Academician E.I.Chazov, Moscow, Russia; Endocrinology Research Centre, Moscow, Russia
| | - K Dergilev
- National Medical Research Centre for Cardiology Named After Academician E.I.Chazov, Moscow, Russia
| | | | | | | | - I Stafeev
- National Medical Research Centre for Cardiology Named After Academician E.I.Chazov, Moscow, Russia; Endocrinology Research Centre, Moscow, Russia.
| | - M Shestakova
- Endocrinology Research Centre, Moscow, Russia; Lomonosov Moscow State University, Moscow, Russia
| | - Ye Parfyonova
- National Medical Research Centre for Cardiology Named After Academician E.I.Chazov, Moscow, Russia; Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
8
|
Li C, Zhang D, Huang J, Zhou H, Song T, Wang X, Kong Q, Li L, Liu Z, Zhang N, Lu Y, Tan J, Zhang J. From non-coding RNAs to cancer regulators: The fascinating world of micropeptides. Int J Cancer 2025. [PMID: 40279117 DOI: 10.1002/ijc.35459] [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: 12/17/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Micropeptides are commonly identified as peptides encoded by non-coding RNAs (ncRNAs). In the short open reading frame (sORF) of ncRNAs, there is a base sequence encoding functional micropeptides, which is of great significance in the biological field. Recently, micropeptides regulate diverse processes, including mitochondrial metabolism, calcium transport, mRNA splicing, signal transduction, myocyte fusion, and cellular senescence, regulating the homeostasis of the internal environment and cancer's incidence and progression. Especially, the study of micropeptides in cancer about the potential regulatory mechanism will be conducive to further understanding of the process of cancer initiation and development. More and more research shows micropeptides have been confirmed to play an essential role in the emergence of multiple kinds of cancers, including Breast cancer, Colon cancer, Colorectal cancer, Glioma, Glioblastoma, and Liver cancer. This review presents a comprehensive synthesis of the latest advancements in our understanding of the biological roles of micropeptides in cancer cells, with a particular focus on the regulatory networks involving micropeptides in oncogenesis. The new mode of action of micropeptides provides innovative ideas for cancer diagnosis and treatment. Moreover, we explored the significant capacity of micropeptides as diagnostic biomarkers and targets for anti-cancer therapies in cancer clinical settings, highlighting their role in the development of innovative micropeptide-based diagnostic tools and anti-cancer therapeutics.
Collapse
Affiliation(s)
- Can Li
- Nanshan Class, Zunyi Medical University, Zunyi, China
| | - Dan Zhang
- Library, Zunyi Medical University, Zunyi, China
| | - Jinxi Huang
- Nanshan Class, Zunyi Medical University, Zunyi, China
| | - He Zhou
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Qinghong Kong
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, China
| | - Liujin Li
- Department of Otolaryngology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhaohui Liu
- Department of Otolaryngology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Neng Zhang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yanxin Lu
- Basic Medical Science Department, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Jun Tan
- Department of Histology and Embryology, Zunyi Medical University, Zunyi, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| |
Collapse
|
9
|
Zeng J, Liu C, Shi Q, Zhang S, Li Y, Liu J, Wang N, Wu G, Wang J, Liu S, Yue S. Stimulated Raman Scattering Microscopy Facilitates the Discovery of Diacylglycerol O-Acyltransferase 2 as a Target to Enhance Iodine Uptake in Papillary Thyroid Carcinoma. Anal Chem 2025; 97:8452-8458. [PMID: 40208310 DOI: 10.1021/acs.analchem.5c00308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Radioactive 131I therapy is a primary treatment for papillary thyroid carcinoma (PTC), with approximately 30% of patients developing iodine-refractory disease. There is an urgent clinical need to improve iodine uptake in PTC. Previous research suggested a connection between triglyceride (TG) accumulation and decreased iodine uptake in benign thyroid cells. Notably, TG accumulation has been found to be a marker of aggressive human PTC. Therefore, it is crucial to elucidate whether TG accumulation affects iodine uptake in PTC, which may lead to a new way for enhancement of iodine uptake. Here, by combining stimulated Raman scattering (SRS) microscopy and deuterated Raman tags, we first quantitatively analyzed the level of TG and its source in the K1 cell with low iodine uptake and the TPC-1 cell with high iodine uptake. It was found that K1 cells had significantly greater TG accumulation than TPC-1 cells, primarily due to an increased exogenous uptake of fatty acids. Further RNA-seq transcriptome experiments revealed that the underlying mechanism could be upregulation of lipid biosynthesis, uptake, and transport-related genes, along with down-regulation of fatty acid β-oxidation and lipolysis-related genes in K1 cells. Among the upregulated lipid biosynthesis genes, diacylglycerol O-acyltransferase 2 (DGAT2) is of great importance as the rate-limiting enzyme in TG biosynthesis. Notably, the inhibition of DGAT2 led to a significant increase in the expression of iodine uptake-related proteins, namely, sodium iodide symporter (NIS) and thyroglobulin (Tg), in K1 cells. Further Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses unraveled that DGAT2 inhibition could enhance thyroid hormone synthesis, for which iodine is an essential raw material, by alleviating endoplasmic reticulum stress and upregulating the pathways related to protein glycosylation and transmembrane transport. In summary, our study has shown that SRS microscopy facilitates the discovery of DGAT2 as a potential target to enhance iodine uptake in PTC, which holds promise for improving treatment outcome of iodine-refractory PTC.
Collapse
Affiliation(s)
- Junjie Zeng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Changjian Liu
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiusheng Shi
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shuo Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yuhui Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Jianlin Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Nana Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Guoliang Wu
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jian Wang
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shaoyan Liu
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuhua Yue
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
10
|
Shen D, Zhao Q, Zhang H, Wu C, Jin H, Guo K, Sun R, Guo H, Zhao Q, Feng H, Dong X, Gao Z, Zhang L, Liu Y. A hydrophobic photouncaging reaction to profile the lipid droplet interactome in tissues. Proc Natl Acad Sci U S A 2025; 122:e2420861122. [PMID: 40238459 PMCID: PMC12037041 DOI: 10.1073/pnas.2420861122] [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: 10/10/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Most bioorthogonal photouncaging reactions preferentially occur in polar environments to accommodate biological applications in the aqueous cellular milieu. However, they are not precisely designed to chemically adapt to the diverse microenvironments of the cell. Herein, we report a hydrophobic photouncaging reaction with tailored photolytic kinetics toward solvent polarity. Structural modulations of the aminobenzoquinone-based photocage reveal the impact of cyclic ring size, steric substituent, and electronic substituent on the individual uncaging kinetics (kH2O and kdioxane) and polarity preference (kdioxane/kH2O). Rational incorporation of optimized moieties leads to up to 20.2-fold nonpolar kinetic selectivity (kdioxane/kH2O). Further photochemical spectroscopic characterizations and theoretical calculations together uncover the mechanism underlying the polarity-dependent uncaging kinetics. The uncaged ortho-quinone methide product bears covalent reactivity toward diverse nucleophiles of a protein revealed by tandem mass spectrometry. Finally, we demonstrate the application of such lipophilic photouncaging chemistry toward selective labeling and profiling of proteins in proximity to lipid droplets inside human fatty liver tissues. Together, this work studies the solvent polarity effects of a photouncaging reaction and chemically adapts it toward suborganelle-targeted protein proximity labeling and profiling.
Collapse
Affiliation(s)
- Di Shen
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Qun Zhao
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Huaiyue Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Ci Wu
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Hao Jin
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Kun Guo
- The Second Hospital of Dalian Medical University, Dalian116023, China
| | - Rui Sun
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Hengke Guo
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Qi Zhao
- The Second Hospital of Dalian Medical University, Dalian116023, China
| | - Huan Feng
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University, Dalian116023, China
| | - Zhenming Gao
- The Second Hospital of Dalian Medical University, Dalian116023, China
| | - Lihua Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Yu Liu
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| |
Collapse
|
11
|
Cotton K, Comer C, Caporali S, Butera A, Gurres S, Capradossi F, D'Alessandro A, Amelio I, Niklison-Chirou MV. Lipidome atlas of p53 mutant variants in pancreatic cancer. Biol Direct 2025; 20:51. [PMID: 40217553 PMCID: PMC11992884 DOI: 10.1186/s13062-025-00635-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/15/2025] [Indexed: 04/14/2025] Open
Abstract
Mutations in the tumour suppressor protein p53 are present in 70% of human pancreatic ductal adenocarcinomas (PDAC), subsequently to highly common activation mutation of the oncogene KRAS. These p53 mutations generate stable expression of mutant proteins, such as p53R175H and p53R273H, which do not retain p53 wild type function. In this study, we investigated the impact of two specific p53 mutant variants on lipid metabolism of pancreatic cancer. Lipids critically participate to tumorigenesis with to their roles in membrane biosynthesis, energy storage and production of signalling molecules. Using cell lines derived from mouse models of PDAC generated by knock-in p53 alleles carrying point mutations at codons R172H and R270H (equivalent to R175H and R273H in humans), we found that silencing p53R172H and p53R270H in pancreatic cancer cells significantly alters lipid metabolism, with patterns of common and variant specific changes. Specifically, loss of p53R172H in these cells reduces lipid storage. Additionally, silencing either p53R172H or p53R270H individually leads to marked increases in lysophospholipid levels. These findings offer new insights into the lipidome reprogramming induced by the loss of mutant p53 and underscore changes in lipid storage as a potential key molecular mechanism in PDAC pathogenesis.
Collapse
Affiliation(s)
- Kian Cotton
- Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Charley Comer
- Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany
| | - Sabrina Caporali
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany
| | - Alessio Butera
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany
| | - Stephanie Gurres
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany
| | | | | | - Ivano Amelio
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany.
| | - Maria Victoria Niklison-Chirou
- Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
12
|
Rezaei A, Kocsis-Jutka V, Gunes ZI, Zeng Q, Kislinger G, Bauernschmitt F, Isilgan HB, Parisi LR, Kaya T, Franzenburg S, Koppenbrink J, Knogler J, Arzberger T, Farny D, Nuscher B, Katona E, Dhingra A, Yang C, Gouna G, LaClair KD, Janjic A, Enard W, Zhou Q, Hagan N, Ofengeim D, Beltrán E, Gokce O, Simons M, Liebscher S, Edbauer D. Correction of dysregulated lipid metabolism normalizes gene expression in oligodendrocytes and prolongs lifespan in female poly-GA C9orf72 mice. Nat Commun 2025; 16:3442. [PMID: 40216746 PMCID: PMC11992041 DOI: 10.1038/s41467-025-58634-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Clinical and genetic research links altered cholesterol metabolism with ALS development and progression, yet pinpointing specific pathomechanisms remain challenging. We investigated how cholesterol dysmetabolism interacts with protein aggregation, demyelination, and neuronal loss in ALS. Bulk RNAseq transcriptomics showed decreased cholesterol biosynthesis and increased cholesterol export in ALS mouse models (GA-Nes, GA-Camk2a GA-CFP, rNLS8) and patient samples (spinal cord), suggesting an adaptive response to cholesterol overload. Consequently, we assessed the efficacy of the cholesterol-binding drug 2-hydroxypropyl-β-cyclodextrin (CD) in a fast-progressing C9orf72 ALS mouse model with extensive poly-GA expression and myelination deficits. CD treatment normalized cholesteryl ester levels, lowered neurofilament light chain levels, and prolonged lifespan in female but not male GA-Nes mice, without impacting poly-GA aggregates. Single nucleus transcriptomics indicated that CD primarily affected oligodendrocytes, significantly restored myelin gene expression, increased density of myelinated axons, inhibited the disease-associated oligodendrocyte response, and downregulated the lipid-associated genes Plin4 and ApoD. These results suggest that reducing excess free cholesterol in the CNS could be a viable ALS treatment strategy.
Collapse
Affiliation(s)
- Ali Rezaei
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | | | - Zeynep I Gunes
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Munich, Germany
- Biomedical Center, Ludwig Maximilians University Munich, Munich, Germany
| | - Qing Zeng
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Georg Kislinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Franz Bauernschmitt
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Munich, Germany
- Biomedical Center, Ludwig Maximilians University Munich, Munich, Germany
| | | | - Laura R Parisi
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA, USA
| | - Tuğberk Kaya
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Jonas Koppenbrink
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Julia Knogler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Daniel Farny
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Brigitte Nuscher
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Eszter Katona
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Ashutosh Dhingra
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Chao Yang
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Garyfallia Gouna
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | | | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, Munich, Germany
| | - Qihui Zhou
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Nellwyn Hagan
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA, USA
| | | | - Eduardo Beltrán
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Munich, Germany
- Biomedical Center, Ludwig Maximilians University Munich, Munich, Germany
| | - Ozgun Gokce
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Sabine Liebscher
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Munich, Germany
- Biomedical Center, Ludwig Maximilians University Munich, Munich, Germany
- Institute of Neurobiochemistry, Medical University of Innsbruck, Innsbruck, Austria
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
| |
Collapse
|
13
|
Zhang R, Zhou H, Wang C, Weng X, Liu L, Xi P, Qu J. Dual-Bessel-Beam Stimulated Emission Depletion Microscopy for Super-resolution Volumetric Projection Imaging of Lipid Droplet Dynamics. NANO LETTERS 2025; 25:5557-5564. [PMID: 40094473 DOI: 10.1021/acs.nanolett.4c04867] [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: 03/19/2025]
Abstract
Volumetric imaging efficiently captures comprehensive spatial structures and dynamic function information on organisms in biomedical research. However, optical diffraction limit restricts the visualization of fine structure details at nanoscale. To address this limitation, we developed dual-Bessel-beam stimulated emission depletion (DB-STED) microscopy to enhance the information throughput and lateral resolution. This technique combines a zeroth-order Bessel beam for excitation with a first-order hollow Bessel beam for depletion, aligned both spatially and temporally to achieve super-resolution volumetric projection imaging. We validated this approach using fluorescent beads embedded in agarose, achieving a resolution of 69 nm over a depth of 10 μm with a numerical aperture of 1.4. The high-throughput and super-resolution capability enables detailed observation of lipid droplet motion within entire cells, providing valuable insights into lipid dynamics.
Collapse
Affiliation(s)
- Renlong Zhang
- State Key Laboratory of Radio Frequency Heterogeneous Integration and Key Laboratory of Optoelectronic Devices and Systems, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haoxian Zhou
- State Key Laboratory of Radio Frequency Heterogeneous Integration and Key Laboratory of Optoelectronic Devices and Systems, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Chenguang Wang
- State Key Laboratory of Integrated Optoelectronics, Key Laboratory of Advanced Gas Sensors of Jilin Province, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Xiaoyu Weng
- State Key Laboratory of Radio Frequency Heterogeneous Integration and Key Laboratory of Optoelectronic Devices and Systems, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Liwei Liu
- State Key Laboratory of Radio Frequency Heterogeneous Integration and Key Laboratory of Optoelectronic Devices and Systems, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Peng Xi
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Junle Qu
- State Key Laboratory of Radio Frequency Heterogeneous Integration and Key Laboratory of Optoelectronic Devices and Systems, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
14
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [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: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
15
|
Zhou Y, Xu Y, Hou X, Xia D. Raman analysis of lipids in cells: Current applications and future prospects. J Pharm Anal 2025; 15:101136. [PMID: 40242217 PMCID: PMC11999598 DOI: 10.1016/j.jpha.2024.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 04/18/2025] Open
Abstract
Lipids play an important role in the regulation of cell life processes. Although there are various lipid detection methods, Raman spectroscopy, a non-invasive technique, provides the detailed chemical composition of lipid profiles without a complex sample preparation procedure and possesses greater potential in basic biology, clinical diagnosis and disease therapy. In this review, we summarized the characteristics and advantages of Raman-based techniques and their primary contribution to illustrating cellular lipid metabolism.
Collapse
Affiliation(s)
- Yixuan Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuelin Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaoli Hou
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Daozong Xia
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
16
|
Wang T, Perelló Amorós M, Lopez Llao G, Porte C. Distinctive lipidomic responses induced by polystyrene micro- and nano-plastics in zebrafish liver cells. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 281:107291. [PMID: 39999695 DOI: 10.1016/j.aquatox.2025.107291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/27/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Despite growing awareness of the size-dependent toxicity caused by micro- and nano-plastics (MNPs) in fish, the modulation of the liver lipidome as a function of particle size has not been thoroughly investigated. This study explores the subcellular and molecular responses induced by polystyrene microplastics (MPs, 1 µm) and nano-plastics (NPs, 52 nm) in zebrafish liver (ZFL) cells, with a focus on the modulation of the cell's lipidome and gene expression profiles. Both particle sizes are readily internalized by ZFL cells; however, NPs had a more pronounced impact compared to MPs. Lipidomic analysis revealed that MPs decreased polyunsaturated phospholipids, while NPs increased ether-linked phosphatidylcholines (PC-Ps/PCOs). Gene expression analysis showed that high concentrations of MPs down-regulated the expression of fatty acid synthesis related genes, and significantly downregulated the microsomal triglyceride transfer protein (mtp) gene, indicating a perturbation in lipid storage metabolism, which was not observed for NP exposure. In contrast, NPs induced a dose-dependent accumulation of lipids, suggesting increased lipid droplet formation and an activation of ceramide-mediated apoptosis pathway. These findings provide new insights into the molecular mechanisms of MNP toxicity and emphasize the importance of considering particle size when assessing environmental and health risks. Furthermore, this study highlights the potential of lipidomics for elucidating the mechanisms underlying MNP toxicity, prompting further research into of the long-term consequences of exposure.
Collapse
Affiliation(s)
- Tiantian Wang
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona, 18-26, 08034, Barcelona, Spain; PhD Program Aquaculture, University of Barcelona, Spain.
| | - Miquel Perelló Amorós
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona, 18-26, 08034, Barcelona, Spain
| | - Gemma Lopez Llao
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona, 18-26, 08034, Barcelona, Spain; PhD Program Aquaculture, University of Barcelona, Spain
| | - Cinta Porte
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona, 18-26, 08034, Barcelona, Spain.
| |
Collapse
|
17
|
Madesh S, Murugan R, Sau A, Jubie S, Swaroop AK, Rajagopal R, Kumaradoss KM, Arockiaraj J. Nano-Encapsulated Coumarin Derivative, CS-QM2 Inhibits Neoplasm Growth: Experimented in Zebrafish Model. J Biochem Mol Toxicol 2025; 39:e70239. [PMID: 40143626 DOI: 10.1002/jbt.70239] [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: 12/25/2024] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025]
Abstract
Cancer remains a significant global health challenge with limited therapeutic success, prompting the need for innovative treatment strategies. This study investigates the anticancer potential of nano-encapsulated metal derivatives (CS-QM2) using a zebrafish model with chemically induced cellular neoplasia. Characterization of CS-QM2 nanoparticles revealed successful synthesis with a high entrapment efficiency and enhanced drug release under acidic conditions. Zebrafish embryos exposed to 7,12-Dimethylbenz[a]anthracene (DMBA) exhibited significant malformations, macrophage accumulation, and abnormal tissue growth, which were markedly reduced by CS-QM2 treatment. CS-QM2 significantly increases intracellular ROS, resulting in higher LPO and induces apoptosis in neoplasm tissues. Furthermore, CS-QM2 treatment alters the tumor microenvironment, reducing macrophage accumulation by decreasing neutral lipid droplets, disrupting TAM metabolic support and limiting their protumorigenic activities. Biochemical assays demonstrated restored activities of antioxidant enzymes SOD, CAT, and GSH. Gene expression analysis showed upregulation of apoptosis and tumor suppressor genes (cas3, p53) and downregulation of inflammatory genes (cox-2, nf-kb). Histological assessment and SEM analysis confirmed reduced neoplasm occurrence and tissue abnormalities. These findings suggest that CS-QM2 nanoparticles effectively inhibit neoplasm growth and modulate the tumor microenvironment through oxidative stress induction and gene expression regulation.
Collapse
Affiliation(s)
- S Madesh
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Raghul Murugan
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Avra Sau
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - S Jubie
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Kathiravan Muthu Kumaradoss
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
18
|
Bhoumik S, Lee Y. Core Molecular Clock Factors Regulate Osteosarcoma Stem Cell Survival and Behavior via CSC/EMT Pathways and Lipid Droplet Biogenesis. Cells 2025; 14:517. [PMID: 40214471 PMCID: PMC11988071 DOI: 10.3390/cells14070517] [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: 01/22/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The circadian clock, an intrinsic 24 h cellular timekeeping system, regulates fundamental biological processes, including tumor physiology and metabolism. Cancer stem cells (CSCs), a subpopulation of cancer cells with self-renewal and tumorigenic capacities, are implicated in tumor initiation, recurrence, and metastasis. Despite growing evidence for the circadian clock's involvement in regulating CSC functions, its precise regulatory mechanisms remain largely unknown. Here, using a human osteosarcoma (OS) model (143B), we have shown that core molecular clock factors are critical for OS stem cell survival and behavior via direct modulation of CSC and lipid metabolic pathways. In single-cell-derived spheroid formation assays, 143B OS cells exhibited robust spheroid-forming capacity under 3D culture conditions. Furthermore, siRNA-mediated depletion of core clock components (i.e., BMAL1, CLOCK, CRY1/2, PER1/2)-essential positive and negative elements of the circadian clock feedback loop-significantly reduced spheroid formation in 143B CSCs isolated from in vivo OS xenografts. In contrast, knockdown of the secondary clock-stabilizing factor genes NR1D1 and NR1D2 had little effect. We also found that knockdown of BMAL1, CLOCK, or CRY1/2 markedly impaired the migration and invasion capacities of 143B CSCs. At the molecular level, silencing of BMAL1, CLOCK, or CRY1/2 distinctly altered the expression of genes associated with stem cell properties and the epithelial-mesenchymal transition (EMT) in 143B CSCs. In addition, disruption of BMAL1, CLOCK, or CRY1/2 expression significantly reduced lipid droplet formation by downregulating the expression of genes involved in lipogenesis (e.g., DGAT1, FASN, ACSL4, PKM2, CHKA, SREBP1), which are closely linked to CSC/EMT processes. Furthermore, transcriptomic analysis of human OS patient samples revealed that compared with other core clock genes, CRY1 was highly expressed in OS tumors relative to controls, and its expression exhibited strong positive correlations with patient prognosis, survival, and LD biogenesis gene expression. These findings highlight the critical role of the molecular circadian clock in regulating CSC properties and metabolism, underscoring the therapeutic potential of targeting the core clock machinery to enhance OS treatment outcomes.
Collapse
Affiliation(s)
- Sukanya Bhoumik
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
- Sleep and Performance Research Center, Washington State University, Spokane, WA 99202, USA
- Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
19
|
Wright ZJ, Tharp NE, Bartel B. ER nests are specialized ER subdomains in Arabidopsis where peroxisomes and lipid droplets form. Dev Cell 2025:S1534-5807(25)00152-2. [PMID: 40157364 DOI: 10.1016/j.devcel.2025.03.005] [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: 11/16/2023] [Revised: 09/08/2024] [Accepted: 03/06/2025] [Indexed: 04/01/2025]
Abstract
Organelles are defining features of eukaryotic cells, yet much remains to be learned about organelle biogenesis. Lipid droplets and peroxisomes, which play opposing roles in storing and catabolizing fats, form from a mysterious domain in the endoplasmic reticulum (ER). We used live-cell fluorescence microscopy to visualize peroxisome and lipid droplet biogenesis in young Arabidopsis seedlings, where lipid catabolism is active, and peroxisomes can be unusually large. We found that the ER domains where these organelles are born, which we term ER nests, are complex, dynamic structures that exclude general ER proteins but accumulate other proteins, including lipid biosynthetic enzymes and the COPII component SAR1. Furthermore, ER nests appear to define peroxisome-lipid droplet contact sites. Our findings provide a framework for understanding how these domains form and sort their protein components, illuminate eukaryotic lipid biosynthesis, and elucidate how distinct organelles arise from the ER.
Collapse
Affiliation(s)
| | - Nathan E Tharp
- Biosciences Department, Rice University, Houston, TX 77005, USA
| | - Bonnie Bartel
- Biosciences Department, Rice University, Houston, TX 77005, USA.
| |
Collapse
|
20
|
Liu J, Aye Y. Tools to Dissect Lipid Droplet Regulation, Players, and Mechanisms. ACS Chem Biol 2025; 20:539-552. [PMID: 40035358 PMCID: PMC11934092 DOI: 10.1021/acschembio.4c00835] [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: 12/13/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 03/05/2025]
Abstract
Spurred by the authors' own recent discovery of reactive metabolite-regulated nexuses involving lipid droplets (LDs), this perspective discusses the latest knowledge and multifaceted approaches toward deconstructing the function of these dynamic organelles, LD-associated localized signaling networks, and protein players. Despite accumulating knowledge surrounding protein families and pathways of conserved importance for LD homeostasis surveillance and maintenance across taxa, much remains to be understood at the molecular level. In particular, metabolic stress-triggered contextual changes in LD-proteins' localized functions, crosstalk with other organelles, and feedback signaling loops and how these are specifically rewired in disease states remain to be illuminated with spatiotemporal precision. We hope this perspective promotes an increased interest in these essential organelles and innovations of new tools and strategies to better understand context-specific LD regulation critical for organismal health.
Collapse
Affiliation(s)
- Jinmin Liu
- University
of Oxford, Oxford OX1 3TA, United
Kingdom
| | - Yimon Aye
- University
of Oxford, Oxford OX1 3TA, United
Kingdom
| |
Collapse
|
21
|
Tong W, Sun J, Shen B, Hu Y, Wang C, Rao M, Li J, Xia D, Dong J, Wang H, Zhu D, Wu H, Cai Z. Transcription Factor FOSL1 Promotes Cisplatin Resistance in Non-Small Cell Lung Cancer Cells by Modulating the Wnt3a/β-Catenin Signaling through Upregulation of PLIN3 Expression. FRONT BIOSCI-LANDMRK 2025; 30:26898. [PMID: 40152390 DOI: 10.31083/fbl26898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is the most prevalent histological subtype of lung cancer, accounting for 45.3% of all cases and serving as a major cause of cancer-related mortality. Although cisplatin (DDP) is a cornerstone in LUAD therapy, its efficacy is often compromised by resistance, leading to therapeutic failure and poor patient outcomes. Lipid metabolism and associated proteins, such as perilipin 3 (PLIN3), have been increasingly implicated in cancer progression and chemoresistance. However, the precise mechanisms through which PLIN3 contributes to cisplatin (DDP) resistance in LUAD remain poorly understood. METHODS To investigate the role of PLIN3 in DDP resistance, its expression in LUAD tissues and its correlation with patient prognosis were analyzed using bioinformatics databases and validated through clinical sample analysis. The effects of PLIN3 knockdown and overexpression on DDP resistance and Wnt3a/β-catenin signaling were assessed using quantitative real-time PCR (qPCR), western blotting, cytotoxicity assays, and colony formation assays. Bioinformatics screening identified FOS-like antigen 1 (FOSL1) as a transcription factor positively correlated with PLIN3, and its involvement in DDP resistance was further examined both in vitro and in vivo. RESULTS PLIN3 expression is significantly elevated in LUAD tissues and correlates with poor overall survival. In LUAD cells, PLIN3 overexpression enhanced DDP resistance by upregulating Wnt3a expression and promoting β-catenin nuclear translocation. Bioinformatics analysis identified FOSL1 as a key transcription factor regulating PLIN3 expression. Experimental validation confirmed that FOSL1 directly binds to the PLIN3 promoter, activating the Wnt3a/β-catenin pathway and promoting DDP resistance. Knockdown of PLIN3 or inhibition of Wnt3a signaling reversed the effects of FOSL1 overexpression on DDP resistance. CONCLUSION This study demonstrates that PLIN3 contributes to DDP resistance in LUAD by activating the Wnt3a/β-catenin signaling pathway, with FOSL1 acting as a critical upstream regulator. Targeting the FOSL1/PLIN3/Wnt/β-catenin axis may provide a promising therapeutic strategy for overcoming chemoresistance in LUAD.
Collapse
Affiliation(s)
- Wanning Tong
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Jianjun Sun
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Bin Shen
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Yaohua Hu
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Chenxing Wang
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Min Rao
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Jin Li
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Delin Xia
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Jiagui Dong
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Hong Wang
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Dongmei Zhu
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Haibo Wu
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| | - Zhigang Cai
- Department of Respiratory and Critical Care Medicine, PLA Navy Medical Center, 200052 Shanghai, China
| |
Collapse
|
22
|
Montagne A, Kotta K, Kielbassa-Elkadi K, Martins I, Martinez-Climent JÁ, Kroemer G, Thieblemont C, Baud V. Fatty Acid Metabolism Provides an Essential Survival Signal in OxPhos and BCR DLBCL Cells. Biomedicines 2025; 13:707. [PMID: 40149683 PMCID: PMC11940118 DOI: 10.3390/biomedicines13030707] [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: 11/13/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Backgroung/objectives: Diffuse large B-cell lymphoma (DLBCL) is the most frequent subtype of malignant lymphoma and is a heterogeneous disease with various gene and chromosomal abnormalities. The development of novel therapeutic treatments has improved DLBCL prognosis, but patients with early relapse or refractory disease have a poor outcome (with a mortality of around 40%). Metabolic reprogramming is a hallmark of cancer cells. Fatty acid (FA) metabolism is frequently altered in cancer cells and recently emerged as a critical survival path for cancer cell survival. Methods: We first performed the metabolic characterization of an extended panel of DLBCL cell lines, including lipid droplet content. Then, we investigated the effect of drugs targeting FA metabolism on DLBCL cell survival. Further, we studied how the combination of drugs targeting FA and either mitochondrial metabolism or mTOR pathway impacts on DLBCL cell death. Results: Here, we reveal, using a large panel of DLBCL cell lines characterized by their metabolic status, that targeting of FA metabolism induces massive DLBCL cell death regardless of their OxPhos or BCR/glycolytic subtype. Further, FA drives resistance of DLBCL cell death induced by mitochondrial stress upon treatment with either metformin or L-asparaginase, two FDA-approved antimetabolic drugs. Interestingly, combining inhibition of FA metabolism with that of the mTOR oncogenic pathway strongly potentiates DLBCL cell death. Conclusion: Altogether, our data highlight the central role played by FA metabolism in DLBCL cell survival, independently of their metabolic subtype, and provide the framework for the use of drugs targeting this metabolic vulnerability to overcome resistance in DLBCL patients.
Collapse
Affiliation(s)
- Aurélie Montagne
- NF-κB, Differentiation and Cancer, Université Paris Cité, 75006 Paris, France; (A.M.); (K.K.); (K.K.-E.); (C.T.)
| | - Konstantina Kotta
- NF-κB, Differentiation and Cancer, Université Paris Cité, 75006 Paris, France; (A.M.); (K.K.); (K.K.-E.); (C.T.)
| | - Karoline Kielbassa-Elkadi
- NF-κB, Differentiation and Cancer, Université Paris Cité, 75006 Paris, France; (A.M.); (K.K.); (K.K.-E.); (C.T.)
| | - Isabelle Martins
- Equipe Labellisée Ligue contre le Cancer, Cordeliers Research Center, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; (I.M.); (G.K.)
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94800 Villejuif, France
| | - José Ángel Martinez-Climent
- Department of Hematology, Center for Applied Medical Research, University of Navarra, IDISNA, CIBERONC, 31071 Pamplona, Spain;
| | - Guido Kroemer
- Equipe Labellisée Ligue contre le Cancer, Cordeliers Research Center, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; (I.M.); (G.K.)
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94800 Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Catherine Thieblemont
- NF-κB, Differentiation and Cancer, Université Paris Cité, 75006 Paris, France; (A.M.); (K.K.); (K.K.-E.); (C.T.)
- Hemato-Oncology, AP-HP, Hôpital Saint-Louis, Université Paris Cité, 75006 Paris, France
| | - Véronique Baud
- NF-κB, Differentiation and Cancer, Université Paris Cité, 75006 Paris, France; (A.M.); (K.K.); (K.K.-E.); (C.T.)
| |
Collapse
|
23
|
Saczuk K, Kassem A, Dudek M, Sánchez DP, Khrouz L, Allain M, Welch GC, Sabouri N, Monnereau C, Josse P, Cabanetos C, Deiana M. Organelle-Specific Thiochromenocarbazole Imide Derivative as a Heavy-Atom-Free Type I Photosensitizer for Biomolecule-Triggered Image-Guided Photodynamic Therapy. J Phys Chem Lett 2025; 16:2273-2282. [PMID: 39988904 PMCID: PMC11891978 DOI: 10.1021/acs.jpclett.5c00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Modern photodynamic therapy (PDT) demands next-generation photosensitizers (PSs) that overcome heavy-atom dependency and enhance efficacy beyond traditional, highly oxygen-dependent type II mechanisms. We introduce herein TCI-NH, as a thiochromenocarbazole imide derivative designed for type I photodynamic action. Upon light activation, TCI-NH efficiently favors superoxide (O2•-) and PS-centered radical formation instead of singlet oxygen (1O2) generation. Its high luminescence efficiency and selective localization in both the endoplasmic reticulum and mitochondria enable precise, image-guided PDT. Notably, interactions with biomolecules, such as serum albumin or DNA, enhance TCI-NH's emission by up to 40-fold and amplify radical generation by up to 5-fold. With negligible dark toxicity, this results in ∼120 nM photocytotoxicity along with an impressive phototherapeutic index exceeding 200. Real-time live-cell imaging revealed rapid, light-triggered cytotoxicity characterized by apoptotic body formation and extensive cellular damage. With its small size, heavy-atom-free structure, exceptional, organelle specificity, and therapeutic efficacy, TCI-NH sets a new benchmark for anticancer type I PDT.
Collapse
Affiliation(s)
- Karolina Saczuk
- Institute
of Advanced Materials, Faculty of Chemistry, Wrocław University of Science and Technology, Wyb. Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Ahmad Kassem
- CNRS,
MOLTECH-ANJOU, SFR-MATRIX, F-49000 Angers, France
| | - Marta Dudek
- Institute
of Advanced Materials, Faculty of Chemistry, Wrocław University of Science and Technology, Wyb. Wyspiańskiego 27, 50-370 Wrocław, Poland
| | | | - Lhoussain Khrouz
- ENS
de Lyon, CNRS, Laboratoire de Chimie, UMR 5182, 46 allée d’Italie, F-69342 Lyon, France
| | - Magali Allain
- CNRS,
MOLTECH-ANJOU, SFR-MATRIX, F-49000 Angers, France
| | - Gregory C. Welch
- Department
of Chemistry, University of Calgary, 731 Campus Place NW, Calgary, Alberta T2N 1N4, Canada
| | - Nasim Sabouri
- Department
of Medical Biochemistry and Biophysics, Science for Life Laboratory, Umeå University, 90187 Umeå, Sweden
| | - Cyrille Monnereau
- ENS
de Lyon, CNRS, Laboratoire de Chimie, UMR 5182, 46 allée d’Italie, F-69342 Lyon, France
| | - Pierre Josse
- CNRS,
MOLTECH-ANJOU, SFR-MATRIX, F-49000 Angers, France
| | | | - Marco Deiana
- Institute
of Advanced Materials, Faculty of Chemistry, Wrocław University of Science and Technology, Wyb. Wyspiańskiego 27, 50-370 Wrocław, Poland
- Department
of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden
| |
Collapse
|
24
|
Ma W, Chen Y, Chen G, Yang L, Lu Y, Dong X, Li D, Gan W. TFE3 fusion proteins promote the progression of TFE3 rearranged renal cell carcinoma via enhancing chaperone-mediated lipophagy. Cell Commun Signal 2025; 23:122. [PMID: 40050998 PMCID: PMC11887198 DOI: 10.1186/s12964-025-02117-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND TFE3 rearranged renal cell carcinoma (TFE3 rRCC), classified as a distinct entity of RCCs, exhibits aggressive biological behavior and possesses unique metabolic characteristics. In the present study, TFE3 rRCC with high expression of TFE3 fusion proteins was employed to investigate the features of lipid metabolism and its underlying mechanism in cancer progression. METHODS Fluorescence microscope and flow cytometry were employed to detect lipid droplets (LDs). GPO-PAP method and Oil Red O staining were used to quantify triacylglycerol levels. The data for bioinformatics analysis were sourced from GEO and iProX. The biological roles of TFE3 and LAMP2A were investigated by CCK8 assay, EdU staining, seahorse, transwell assay, colony, and sphere formation assay. The regulatory mechanisms involving TFE3, LAMP2A and Hsc70 were investigated using western blotting, immunohistochemistry, qRT-PCR, luciferase assays, Co-IP techniques, and ChIP analyses. RESULTS The level of LDs accumulation in TFE3 rRCC was relatively low, and the knockdown of TFE3 led to an increase in LDs accumulation while inhibiting tumor progression. The underlying mechanism revealed that TFE3 fusion proteins inhibited the biosynthesis of LDs within the endoplasmic reticulum by promoting the degradation of DGAT1 and DGAT2 via autophagy. Furthermore, TFE3 fusion proteins upregulated LAMP2A, thereby enhancing chaperone-mediated autophagy pathways. The process facilitated the degradation of LDs and promoted oxidative metabolism of long-chain fatty acids in mitochondria. CONCLUSIONS TFE3 fusion proteins facilitated the progression of TFE3 rRCC through enhancing the degradation of LDs via chaperone-mediated lipophagy. LAMP2A could serve as a novel potential prognostic biomarker and therapeutic targets.
Collapse
Affiliation(s)
- Wenliang Ma
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, 210008, China
| | - Yi Chen
- Department of Cardiology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, China
| | - Guijuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Lei Yang
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Yanwen Lu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, 210008, China
| | - Xiang Dong
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, 210008, China
| | - Dongmei Li
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Weidong Gan
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, 210008, China.
| |
Collapse
|
25
|
Lian C, He M, Zhao C, Wang T, Tong F, Chen J, Ju R. PLIN2: a potential prognostic markers of early-stage atypical endometrial hyperplasia. J Gynecol Oncol 2025; 36:36.e84. [PMID: 40114556 DOI: 10.3802/jgo.2025.36.e84] [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: 08/09/2024] [Revised: 12/04/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE In the background of endometrial hyperplasia triggered by obesity and estrogen, could the perilipin 2 (PLIN2) serve as a possible prognostic marker for early atypical endometrial hyperplasia (AEH)? METHODS A retrospective study examined blood lipid levels in endometrial cancer (EC) or AEH patients. An AEH mice model was established administrating with estradiol and/or high-fat (HF) diet. Hematoxylin and eosin staining were employed to assess pathological changes in the endometrium. Immunohistochemical staining were employed to evaluate the expression of adipose metabolism and endometrial hyperplasia proteins. The Cell Counting Kit-8 assay, cell colony-forming assays, and western blotting were utilized to verify the impact of oleic acid (OA) on HEC-1A cells. RESULTS The retrospective study revealed elevated blood lipid levels among patients with EC or AEH. Prolonged HF diet stimulated the endometrium to exhibit features of complex atypical hyperplasia. In the early stage, PLIN2 (p=0.006) expression significantly increased with endometrial glandular hyperplasia. Both PLIN2 (p=0.008) and progesterone receptor (PR; p=0.019) exhibited elevated expression concurrent with simple endometrial hyperplasia. When AEH occurred, there were notable rise in the expression of PLIN2 (p<0.001), PR (p=0.044), and estrogen receptor (p=0.045). The atypical hyperplasia glands demonstrated notably elevated PLIN2 expression in comparison to surrounding normal glands in AEH lesions. OA was found to enhance the proliferation and clonal formation of HEC-1A cells. HEC-1A cells induced by OA demonstrated elevated autophagy levels accompanied by enhanced expression of PLIN2. CONCLUSION PLIN2 may potentially serve as a biomarker for early development of AEH and EC, facilitating diagnosis and intervention and contributing to the determination of prognosis.
Collapse
Affiliation(s)
- Chao Lian
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Minling He
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Chengcheng Zhao
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Tianming Wang
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Fang Tong
- Department of Pathology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China.
| | - Rong Ju
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
26
|
Wan M, Pan S, Shan B, Diao H, Jin H, Wang Z, Wang W, Han S, Liu W, He J, Zheng Z, Pan Y, Han X, Zhang J. Lipid metabolic reprograming: the unsung hero in breast cancer progression and tumor microenvironment. Mol Cancer 2025; 24:61. [PMID: 40025508 PMCID: PMC11874147 DOI: 10.1186/s12943-025-02258-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Aberrant lipid metabolism is a well-recognized hallmark of cancer. Notably, breast cancer (BC) arises from a lipid-rich microenvironment and depends significantly on lipid metabolic reprogramming to fulfill its developmental requirements. In this review, we revisit the pivotal role of lipid metabolism in BC, underscoring its impact on the progression and tumor microenvironment. Firstly, we delineate the overall landscape of lipid metabolism in BC, highlighting its roles in tumor progression and patient prognosis. Given that lipids can also act as signaling molecules, we next describe the lipid signaling exchanges between BC cells and other cellular components in the tumor microenvironment. Additionally, we summarize the therapeutic potential of targeting lipid metabolism from the aspects of lipid metabolism processes, lipid-related transcription factors and immunotherapy in BC. Finally, we discuss the possibilities and problems associated with clinical applications of lipid‑targeted therapy in BC, and propose new research directions with advances in spatiotemporal multi-omics.
Collapse
Affiliation(s)
- Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haizhou Diao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Ziqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Shuya Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wan Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaying He
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Graduate School of Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zihan Zheng
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
27
|
Zhang J, Xu S, Fang H, Wu D, Ouyang C, Shi Y, Hu Z, Zhang M, Zhong Y, Zhao J, Gan Y, Zhang S, Liu X, Yin J, Li Y, Tang M, Wang Y, Li L, Chan WC, Horne D, Feng M, Huang W, Gu Y. CAMKIIδ Reinforces Lipid Metabolism and Promotes the Development of B Cell Lymphoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409513. [PMID: 39840457 PMCID: PMC11905072 DOI: 10.1002/advs.202409513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/14/2024] [Indexed: 01/23/2025]
Abstract
The most prevalent types of lymphomas are B cell lymphomas (BCL). Newer therapies for BCL have improved the prognosis for many patients. However, approximately 30% with aggressive BCL either remain refractory or ultimately relapse. These patients urgently need other options. This study shows how calcium/calmodulin-dependent protein kinase II delta (CAMKIIδ) is pivotal for BCL development. In BCL cells, ablation of CAMKIIδ inhibits both lipolysis from lipid droplets and oxidative phosphorylation (OXPHOS). With lipolysis blocked, BCL progression is markedly suppressed in two distinct BCL mouse models: MYC-driven EµMyc mice and Myc/Bcl2 double-expressed mice. When CAMKIIδ is present, it destabilizes transcription factor Forkhead Box O3A (FOXO3A) by phosphorylating it at Ser7 and Ser12. This then permits transcription of downstream gene IRF4 - a master transcription factor of lipid metabolism. The CAMKIIδ/FOXO3A axis bolsters lipid metabolism, mitochondrial respiration, and tumor fitness in BCL under metabolic stress. This study also evaluates Tetrandrine (TET), a small molecule compound, as a potent CAMKIIδ inhibitor. TET attenuates metabolic fitness and elicits therapeutic responses both in vitro and in vivo. Collectively, this study highlights how CAMKIIδ is critical in BCL progression. The results also pave the way for innovative therapeutic strategies for treating aggressive BCL.
Collapse
|
28
|
Weng X, Gonzalez M, Angelia J, Piroozmand S, Jamehdor S, Behrooz AB, Latifi-Navid H, Ahmadi M, Pecic S. Lipidomics-driven drug discovery and delivery strategies in glioblastoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167637. [PMID: 39722408 DOI: 10.1016/j.bbadis.2024.167637] [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: 09/28/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
With few viable treatment options, glioblastoma (GBM) is still one of the most aggressive and deadly types of brain cancer. Recent developments in lipidomics have demonstrated the potential of lipid metabolism as a therapeutic target in GBM. The thorough examination of lipids in biological systems, or lipidomics, is essential to comprehending the changed lipid profiles found in GBM, which are linked to the tumor's ability to grow, survive, and resist treatment. The use of lipidomics in drug delivery and discovery is examined in this study, focusing on how it may be used to find new biomarkers, create multi-target directed ligands, and improve drug delivery systems. We also cover the use of FDA-approved medications, clinical trials that use lipid-targeted medicines, and the integration of lipidomics with other omics technologies. This study emphasizes lipidomics as a possible tool in developing more effective treatment methods for GBM by exploring various lipid-centric techniques.
Collapse
Affiliation(s)
- Xiaohui Weng
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Michael Gonzalez
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Jeannes Angelia
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran; School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry and Petroleum Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States.
| |
Collapse
|
29
|
Yamamoto T, Okuno M, Kuwano K, Ogura Y. Mycoplasma pneumoniae drives macrophage lipid uptake via GlpD-mediated oxidation, facilitating foam cell formation. Int J Med Microbiol 2025; 318:151646. [PMID: 39862618 DOI: 10.1016/j.ijmm.2025.151646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Cardiovascular diseases, primarily caused by atherosclerosis, are a major public health concern worldwide. Atherosclerosis is characterized by chronic inflammation and lipid accumulation in the arterial wall, leading to plaque formation. In this process, macrophages play a crucial role by ingesting lipids and transforming into foam cells, which contribute to plaque instability and cardiovascular events. Recent studies have suggested that various pathogens are involved in the development of atherosclerosis, with Mycoplasma pneumoniae considered one of the potential candidates. Therefore, this study investigated whether this bacterium induces lipid accumulation in macrophages, which play a crucial role in the development of atherosclerosis, using the Raw264.7 model. Our findings revealed that M. pneumoniae infection promotes lipid droplet formation in macrophages. Glycerol 3-phosphate oxidase, GlpD, in the bacterium is involved in this process by producing reactive oxygen species, which in turn causes the oxidation of low-density lipoprotein. Furthermore, the significant increase in the expression of oxidized lipid receptors involved in the uptake of this oxidized lipid indicates that the bacteria promote lipid uptake in infected macrophages. These results suggest that M. pneumoniae has a direct pro-atherogenic effect, promoting the formation of atherosclerotic lesions through foam cell formation. Understanding the mechanisms by which M. pneumoniae influences atherosclerosis provides valuable insights for devising new therapeutic strategies for the prevention and management of cardiovascular diseases.
Collapse
Affiliation(s)
- Takeshi Yamamoto
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan.
| | - Miki Okuno
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Koichi Kuwano
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Yoshitoshi Ogura
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| |
Collapse
|
30
|
Luo H, She X, Zhang Y, Xie B, Zhang S, Li Q, Zhou Y, Guo S, Zhang S, Jiang Y, Dong Y, He J, Wang L, Zhang Q, Zhuang Y, Deng P, Wang F, Liu J, Chen X, Nie H, He H. PLIN2 Promotes Lipid Accumulation in Ascites-Associated Macrophages and Ovarian Cancer Progression by HIF1α/SPP1 Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411314. [PMID: 39921309 PMCID: PMC11948008 DOI: 10.1002/advs.202411314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/21/2025] [Indexed: 02/10/2025]
Abstract
A major characteristic of ovarian cancer (OC) is its unique route of metastasis via ascites. The immune microenvironment in ascites remains understudied, leaving the mechanism of ascites-mediated abdominal metastasis obscure. Here, a single-cell transcriptomic landscape of CD45+ immune cells across multiple anatomical sites is depicted, including primary tumors, metastatic lesions, and ascites, from patients diagnosed with high-grade serous ovarian carcinoma (HGSOC). A novel subset of perilipin 2 high (PLIN2hi) macrophages are identified that are enriched in ascites and positively correlated with OC progression, hence being designated as "ascites-associated macrophages (AAMs)". AAMs are lipid-loaded with overexpression of the lipid droplet protein PLIN2. Overexpression or suppression of PLIN2 can enhance or inhibit tumor cell migration, invasion, and vascular permeability in vitro, which is also confirmed in vivo. Mechanistically, it is demonstrated that PLIN2 boosts HIF1α/SPP1 signaling in macrophages, thereby exerting pro-tumor functions. Finally, a PLIN2-targeting liposome is designed to efficiently suppress ascites production and tumor metastasis. Taken together, this work provides a comprehensive characterization of the cancer-promoting function and lipid-rich property of ascites-enriched PLIN2hi macrophages, establishes a link between lipid metabolism and hypoxia within the context of the ascites microenvironment, and elucidates the pivotal role of ascites in trans-coelomic metastasis of OC.
Collapse
Affiliation(s)
- Hui Luo
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Xiaolu She
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yubo Zhang
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Bingfan Xie
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Shibo Zhang
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Qianqian Li
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yangyang Zhou
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Shuang Guo
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Shushan Zhang
- Department of UltrasoundThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yanhui Jiang
- Cancer CenterThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yingying Dong
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Jianzhong He
- Cancer CenterThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Lijie Wang
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Qianqian Zhang
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Yuan Zhuang
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Panxia Deng
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Feng Wang
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Jihong Liu
- Department of Gynecology OncologyState Key Laboratory of Oncology in South ChinaSun Yat‐Sen University Cancer CenterGuangzhouGuangdong510060China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacau999078China
| | - Huilong Nie
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| | - Huanhuan He
- Guangdong Provincial Engineering Research Center of Molecular ImagingGuangdong‐Hong Kong‐Macao University Joint Laboratory of Interventional MedicineThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiGuangdong519000China
| |
Collapse
|
31
|
Wang J, Wang M, Zeng X, Li Y, Lei L, Chen C, Lin X, Fang P, Guo Y, Jiang X, Wang Y, Chen L, Long J. Targeting membrane contact sites to mediate lipid dynamics: innovative cancer therapies. Cell Commun Signal 2025; 23:89. [PMID: 39955542 PMCID: PMC11830217 DOI: 10.1186/s12964-025-02089-z] [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: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
Membrane contact sites (MCS) are specialized regions where organelles are closely interconnected through membrane structures, facilitating the transfer and exchange of ions, lipids, and other molecules. This proximity enables a synergistic regulation of cellular homeostasis and functions. The formation and maintenance of these contact sites are governed by specific proteins that bring organelle membranes into close apposition, thereby enabling functional crosstalk between cellular compartments. In eukaryotic cells, lipids are primarily synthesized and metabolized within distinct organelles and must be transported through MCS to ensure proper cellular function. Consequently, MCS act as pivotal platforms for lipid synthesis and trafficking, particularly in cancer cells and immune cells within the tumor microenvironment, where dynamic alterations are critical for maintaining lipid homeostasis. This article provides a comprehensive analysis of how these cells exploit membrane contact sites to modulate lipid synthesis, metabolism, and transport, with a specific focus on how MCS-mediated lipid dynamics influence tumor progression. We also examine the differences in MCS and associated molecules across various cancer types, exploring novel therapeutic strategies targeting MCS-related lipid metabolism for the development of anticancer drugs, while also addressing the challenges involved.
Collapse
Affiliation(s)
- Jie Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China.
| | - Meifeng Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Xueni Zeng
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Yanhan Li
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Lingzhi Lei
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Changan Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Xi Lin
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Peiyuan Fang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China
| | - Yuxuan Guo
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, 410013, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Yian Wang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Department of Pathophysiology, School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, 410013, China
| | - Lihong Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, University Town, Fuzhou, Fujian, 350122, China.
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China.
| | - Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
32
|
Cortini M, Ilieva E, Massari S, Bettini G, Avnet S, Baldini N. Uncovering the protective role of lipid droplet accumulation against acid-induced oxidative stress and cell death in osteosarcoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167576. [PMID: 39561857 DOI: 10.1016/j.bbadis.2024.167576] [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: 04/29/2024] [Revised: 10/14/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Extracellular acidosis stemming from altered tumor metabolism promotes cancer progression by enabling tumor cell adaptation to the hostile microenvironment. In osteosarcoma, we have previously shown that acidosis increases tumor cell survival alongside substantial lipid droplet accumulation. In this study, we explored the role of lipid droplet formation in mitigating cellular stress induced by extracellular acidosis in osteosarcoma cells, thereby enhancing tumor survival during progression. Specifically, we examined how lipid droplets shield against reactive oxygen species induced by extracellular acidosis. We demonstrated that lipid droplet biogenesis is critical for acid-exposed tumor cell survival, as it starts shortly after acid exposure (24 h) and inversely correlates with ROS levels (DCFH-DA assay), lipid peroxidation (Bodipy assay), and the antioxidant response, as also revealed by NRF2 transcript. Additionally, extracellular metabolites, such as lactate, and interaction with mesenchymal stromal cells within the tumor microenvironment intensify lipid droplet build-up in osteosarcoma cells. Critically, upon targeting two key proteins implicated in LD formation - PLIN2 and DGAT1 - cell viability significantly declined while ROS production escalated. In summary, our findings underscore the vital reliance of acid-exposed tumor cells on lipid droplet formation to scavenge oxidative stress. We conclude that the rewiring of lipid metabolism driven by microenvironmental cues is of paramount importance for the survival of metabolically altered osteosarcoma cells in acidic condition. Overall, we suggest that targeting key members of lipid droplet biogenesis may eradicate more aggressive and resistant tumor cells, uncovering potential new treatment strategies for osteosarcoma.
Collapse
Affiliation(s)
- Margherita Cortini
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy.
| | - Elizabeta Ilieva
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy.
| | - Stefania Massari
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy
| | - Giuliano Bettini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, 40100 Ozzano dell'Emilia, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy.
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy; Biomedical Science, Technology and Nanobiotechnology Laboratory, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| |
Collapse
|
33
|
Jog E, Jainarayanan AK, La Ferlita A, Chakraborty A, Dalwai A, Yahya S, Shivashankar A, Choudhary BS, Chandramouli A, Kazi M, Jain D, Khapare N, B A, Khan BK, Gera P, Patil P, Thorat R, Verma N, Sehgal L, Saklani A, Kamat SS, Dalal SN, Chaudhary N. Inhibiting de novo lipogenesis identifies a therapeutic vulnerability in therapy-resistant colorectal cancer. Redox Biol 2025; 79:103458. [PMID: 39705849 PMCID: PMC11729006 DOI: 10.1016/j.redox.2024.103458] [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: 09/30/2024] [Revised: 11/18/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
A significant clinical challenge in patients with colorectal cancer (CRC), which adversely impacts patient survival, is the development of therapy resistance leading to a relapse. Therapy resistance and relapse in CRC is associated with the formation of lipid droplets (LD) by stimulating de novo lipogenesis (DNL). However, the molecular mechanisms underlying the increase in DNL and the susceptibility to DNL-targeted therapies remain unclear. Our study demonstrates that colorectal drug-tolerant persister cells (DTPs) over-express Lipin1 (LPIN1), which facilitates the sequestration of free fatty acids into LDs. The increased expression is mediated by the ETS1-PTPN1-c-Src-CEBPβ pathway. Blocking the conversion of free fatty acids into LDs by treatment with statins or inhibiting lipin1 expression disrupts lipid homeostasis, leading to lipotoxicity and ferroptotic cell death in both DTPs and patient-derived organoids (PDOs) in vitro. Ferroptosis inhibitors or N-acetylcysteine (NAC) can alleviate lipid ROS and cell death resulting from lipin1 inhibition. This strategy also significantly reduces tumor growth in CRC DTP mouse xenograft and patient-derived xenograft (PDX) models. Our findings highlight a new metabolic vulnerability in CRC DTPs, PDO, and PDX models and provide a framework for the rational repurposing of statins. Targeting the phosphatidic acid (PA) to diacylglycerol (DAG) conversion to prevent lipid droplet formation could be an effective therapeutic approach for therapy-resistant CRC.
Collapse
Affiliation(s)
- Eeshrita Jog
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Ashwin Kumar Jainarayanan
- Interdisciplinary Bioscience Doctoral Training Program and Exeter College, University of Oxford, Oxford, UK
| | - Alessandro La Ferlita
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Arnab Chakraborty
- Department of Biology, Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Afiya Dalwai
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Showket Yahya
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Anusha Shivashankar
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Bhagya Shree Choudhary
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Aakash Chandramouli
- Department of Biology, Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Mufaddal Kazi
- Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Department of Gastrointestinal Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Darshan Jain
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Nileema Khapare
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Akshaya B
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Bushra K Khan
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Poonam Gera
- Biorepository, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Prachi Patil
- Department of Digestive Disease and Clinical Nutrition India, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Nandini Verma
- TNBC Precision Medicine Group, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Lalit Sehgal
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Avanish Saklani
- Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Department of Gastrointestinal Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Siddhesh S Kamat
- Department of Biology, Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Sorab N Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Nazia Chaudhary
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India.
| |
Collapse
|
34
|
Jung W, Yang MJ, Kang MS, Lim J, Choi H, Lee JA, Yoon KS, Kim JB, Park EJ. Didecyldimethylammonium chloride-induced lung fibrosis may be associated with phospholipidosis. Toxicol Appl Pharmacol 2025; 495:117211. [PMID: 39710153 DOI: 10.1016/j.taap.2024.117211] [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: 11/15/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
In the current study, we dosed didecyldimethylammonium chloride (DDAC) in mice by pharyngeal aspiration for 28 days or 90 days (weekly) and tried to elucidate the relationship between lamellar body formation and the lesions. When exposed for 28 days (0, 5, 10, 50, and 100 μg/head), all the mice in the 50 and 100 μg/head groups died since Day 2 after the third dosing (Day 16 after the first dosing). Edema, necrosis of bronchiolar and alveolar epithelium, and fibrinous exudate were observed in the lungs of all the dead mice, and chronic inflammatory lesions were observed in the lung tissues of alive mice. When dosed with DDAC of 0, 1, 4, and 8 μg/head for 13 weeks, the total number of pulmonary cells and the pulmonary levels of pro- and anti-inflammatory cytokines significantly increased, and chronic inflammatory lesions were detected with the production of collagen, collagen fibers, and lamellar body-like structures. Swelling of the nuclear envelope and nucleoplasmic components and generation of lipid droplets were also notably observed in the lung tissues of DDAC (8 μg/head)-treated mice. Furthermore, transcriptomic analysis performed using human bronchial epithelial cells showed that DDAC affected the expression of DNA damage, ER stress, lipid metabolism, and transcription regulation-related genes at 6 h after treatment, as it did 24 h treatment and that early growth response factor 1 gene was added to a list of the most up-regulated genes. Meanwhile, cytokines that are associated with the pathology of chronic lung diseases (IL-11, IL-24, and TGF-β) were slightly increased in the lung of DDAC-treated mice, and only the pulmonary level of CCL-2, but not CXCL-1 and CCL-3, increased in both sexes of mice. More importantly, the GM-CSF level increased dose-dependently in the lungs of both sexes of mice exposed to DDAC. Considering that the wound-healing process can take several weeks to complete, we suggest that DDAC-induced pulmonary fibrosis may be attributable to disruption of the wound-healing process due to continuous exposure to DDAC. We also hypothesize that the formation of lamellar bodies may be attributable to lysosomal accumulation of phospholipids separated from the destroyed lung tissue membrane.
Collapse
Affiliation(s)
- Wonkyun Jung
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea
| | - Mi-Jin Yang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongup 56212, Republic of Korea
| | - Min-Sung Kang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongup 56212, Republic of Korea
| | - Jiyun Lim
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea
| | - Hyosun Choi
- National Instrumentation Center for Environmental management, Seoul National University
| | - Ji Ae Lee
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea
| | - Kyung-Sik Yoon
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea; Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea
| | - Jin-Bae Kim
- Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea; Division of Cardiology, Department of Internal Medicine, Kyung-Hee University Hospital, Kyung Hee University, 02447, Republic of Korea
| | - Eun-Jung Park
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea; Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea.
| |
Collapse
|
35
|
Smiriglia A, Lorito N, Bacci M, Subbiani A, Bonechi F, Comito G, Kowalik MA, Perra A, Morandi A. Estrogen-dependent activation of TRX2 reverses oxidative stress and metabolic dysfunction associated with steatotic disease. Cell Death Dis 2025; 16:57. [PMID: 39890799 PMCID: PMC11785963 DOI: 10.1038/s41419-025-07331-7] [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: 04/12/2024] [Revised: 12/09/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a spectrum of hepatic disorders, ranging from simple steatosis to steatohepatitis, with the most severe outcomes including cirrhosis, liver failure, and hepatocellular carcinoma. Notably, MASLD prevalence is lower in premenopausal women than in men, suggesting a potential protective role of estrogens in mitigating disease onset and progression. In this study, we utilized preclinical in vitro models-immortalized cell lines and hepatocyte-like cells derived from human embryonic stem cells-exposed to clinically relevant steatotic-inducing agents. These exposures led to lipid droplet (LD) accumulation, increased reactive oxygen species (ROS) levels, and mitochondrial dysfunction, along with decreased expression of markers associated with hepatocyte functionality and differentiation. Estrogen treatment in steatotic-induced liver cells resulted in reduced ROS levels and LD content while preserving mitochondrial integrity, mediated by the upregulation of mitochondrial thioredoxin 2 (TRX2), an antioxidant system regulated by the estrogen receptor. Furthermore, disruption of TRX2, either pharmacologically using auranofin or through genetic interference, was sufficient to counteract the protective effects of estrogens, highlighting a potential mechanism through which estrogens may prevent or slow MASLD progression.
Collapse
Affiliation(s)
- Alfredo Smiriglia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Nicla Lorito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Marina Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Angela Subbiani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Francesca Bonechi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy
| | - Marta Anna Kowalik
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Andrea Perra
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134, Florence, Italy.
| |
Collapse
|
36
|
Zhang JL, Wang XF, Li JL, Duan C, Wang JF. The cholesterol metabolite 25-hydroxycholesterol suppresses porcine deltacoronavirus via lipophagy inhibition and mTORC1 modulation. Vet Res 2025; 56:23. [PMID: 39891192 PMCID: PMC11786589 DOI: 10.1186/s13567-025-01452-9] [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: 09/18/2024] [Accepted: 11/29/2024] [Indexed: 02/03/2025] Open
Abstract
25-Hydroxycholesterol (25HC) is a hydroxylated cholesterol with multiple antiviral activities, however, little is known about the mechanisms by which 25HC correlates antiviral ability with lipid droplet (LD) dynamic balance to ensure cholesterol homeostasis. In the present study, 25HC was applied to porcine deltacoronavirus (PDCoV)-infected LLC-PK1 (Lilly Laboratories Culture-Porcine Kidney 1) cells and piglets to explore its antiviral capacity and underlying mechanism. The results revealed that 25HC decreased free cholesterol (FC) levels but increased triglyceride (TG) levels in PDCoV-infected cells and piglets. The accumulation of LDs induced by oleic acid (OA) impedes PDCoV replication. In addition, 25HC administration increases LD accumulation and declines protein expression associated with lipophagy and lysosomes to facilitate LD accumulation. Moreover, 25HC inhibited TFEB (transcription factor-EB) expression, blocked its translocation into the nucleus and reversed Mechanistic Target of Rapamycin Complex 1 (mTORC1) activity, which in turn hindered lipophagy and PDCoV replication. Additionally, 25HC treatment ameliorated the clinical symptoms and intestinal injury of PDCoV-infected piglets. These findings reveal the beneficial effect of lipophagy on PDCoV infection and uncover the antiviral mechanism of 25HC, by which lipophagy and mTOR activity are tightly controlled by 25HC.
Collapse
Affiliation(s)
- Jia-Lu Zhang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
- College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
| | - Xue-Fei Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jia-Lin Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Cong Duan
- China Institute of Veterinary Drug Control, Beijing, 100081, China.
| | - Jiu-Feng Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
37
|
Zhang J, Peng J, Wang S, Wang L, Sun Y, Xia J, Cheng B, Hu Q. Perilipin2-dependent lipid droplets accumulation promotes metastasis of oral squamous cell carcinoma via epithelial-mesenchymal transition. Cell Death Discov 2025; 11:30. [PMID: 39875372 PMCID: PMC11775315 DOI: 10.1038/s41420-025-02314-1] [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: 07/26/2024] [Revised: 12/28/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025] Open
Abstract
Emerging evidence shows that lipid metabolic reprogramming plays a vital role in tumor metastasis. The effect and mechanism of fatty acids and lipid droplets (LDs), the core products of lipid metabolism, on the metastasis of oral squamous cell carcinoma (OSCC), need further exploration. In this study, the influence of palmitic acid (PA) and oleic acid (OA) on the migration and invasion ability of OSCC cells was determined by in vitro experiments. Genetic manipulation of PLIN2 was performed to explore its effect on the accumulation of LDs and OSCC metastasis. Possible mechanisms of these biological effects were clarified by detecting the levels of epithelial-mesenchymal transition (EMT) markers and phosphatidylinositol 3-kinase (PI3K) pathway proteins as well as conducting various bioinformatics analyses. The results indicated that PA/OA promoted the migration and invasion of OSCC cells and induced PLIN2-dependent LDs accumulation in vitro. Knockdown of PLIN2 inhibited the LDs accumulation and the migration and invasion of OSCC cells in vitro, while overexpression of PLIN2 enhanced those of OSCC cells in vitro and also promoted the metastasis of OSCC in vivo. Besides, PLIN2 up-regulation activated the PI3K pathway and subsequently enhanced EMT in OSCC cells in vitro. OSCC patients with higher PLIN2 expression possessed poorer prognosis and higher sensitivity to chemotherapy drugs (1S,3 R)-RSL3 and ML-210. In conclusion, PLIN2-dependent LDs accumulation could promote the metastasis of OSCC cells by regulating EMT. PLIN2 might be a potential therapeutic target for OSCC patients, especially those with obesity.
Collapse
Affiliation(s)
- Jiayu Zhang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Jianmin Peng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Siyu Wang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Li Wang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Yutong Sun
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Qinchao Hu
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
38
|
Gonzalez GA, Osuji EU, Fiur NC, Clark MG, Ma S, Lukov LL, Zhang C. Alteration of Lipid Metabolism in Hypoxic Cancer Cells. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:25-34. [PMID: 39886224 PMCID: PMC11775851 DOI: 10.1021/cbmi.4c00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 02/01/2025]
Abstract
Due to uncontrolled cell proliferation and disrupted vascularization, many cancer cells in solid tumors have limited oxygen supply. The hypoxic microenvironments of tumors lead to metabolic reprogramming of cancer cells, contributing to therapy resistance and metastasis. To identify better targets for the effective removal of hypoxia-adaptive cancer cells, it is crucial to understand how cancer cells alter their metabolism in hypoxic conditions. Here, we studied lipid metabolic changes in cancer cells under hypoxia using coherent Raman scattering (CRS) microscopy. We discovered the accumulation of lipid droplets (LDs) in the endoplasmic reticulum (ER) in hypoxia. Time-lapse CRS microscopy revealed the release of old LDs and the reaccumulated LDs in the ER during hypoxia exposure. Additionally, we explored the impact of carbon sources on LD formation and found that MIA PaCa2 cells preferred fatty acid uptake for LD formation, while glucose was essential to alleviate lipotoxicity. Hyperspectral-stimulated Raman scattering (SRS) microscopy revealed a reduction in cholesteryl ester content and a decrease in lipid saturation levels of LDs in hypoxic MIA PaCa2 cancer cells. This alteration in LD content is linked to reduced efficacy of treatments targeting cholesteryl ester formation. This study unveils important lipid metabolic changes in hypoxic cancer cells, providing insights that could lead to better treatment strategies for hypoxia-resistant cancer cells.
Collapse
Affiliation(s)
- Gil A. Gonzalez
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Ezinne U. Osuji
- College
of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, 201 S. University Street, West Lafayette, Indiana 47907, United States
| | - Natalie C. Fiur
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, 201 S. University Street, West Lafayette, Indiana 47907, United States
| | - Matthew G. Clark
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Seohee Ma
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Laura L. Lukov
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Chi Zhang
- Department
of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
- Purdue
Center for Cancer Research, 201 S. University Street, West Lafayette, Indiana 47907, United States
- Purdue
Institute of Inflammation, Immunology, and Infectious Disease, 207 S. Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
39
|
Kalarikkal C, Anjali, Bhattacharjee S, Mapa K, P CAS. Lipid droplet specific BODIPY based rotors with viscosity sensitivity to distinguish normal and cancer cells: impact of molecular conformation. J Mater Chem B 2025; 13:1474-1486. [PMID: 39698835 DOI: 10.1039/d4tb02405b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Lipid droplets (LDs) are dynamic, multifunctional organelles critical for regulating energy balance, cell signaling, membrane formation, and trafficking. Recent studies have highlighted LDs as emerging cancer biomarkers, with cancer cells typically exhibiting a higher number and viscosity of LDs compared to normal cells. This discovery paves the way for developing molecular probes that can monitor intracellular viscosity changes within LDs, offering a powerful tool for early cancer diagnosis, recurrence monitoring, and therapeutic interventions. In this study, we designed and synthesized two series of donor-acceptor (D-A) conjugated BODIPY-cyanostilbene based fluorophores (5a-c and 6a-c) by fine-tuning the cyanostilbene unit with three distinct substituents (OMe, H, Cl) and modulating the molecular conformation via rigidifying the indacene core. While the terminal substituents had a minimal effect on the optical properties, changes in molecular conformation significantly impacted the photophysical behavior of the fluorophores. Compounds 5a-c function as molecular rotors, with the free rotation of the meso-biphenyl rings leading to non-radiative deactivation of the excited state, resulting in weak emission. Additionally, this structural feature makes them highly responsive to changes in viscosity. As the glycerol concentration increased from 0% to 99%, the fluorescence intensity of compounds 5a, 5b, and 5c increased dramatically by 17-fold, 78-fold, and 43-fold, respectively. In contrast, compounds 6a-c, with restricted phenyl ring rotation due to tetra-methyls on the indacene unit, showed only a modest 2-3-fold increment in fluorescence intensity under similar conditions. These fluorophores possess several key advantages, including high selectivity for LDs, good photostability, sensitivity to viscosity, and responsiveness to polarity and pH. Moreover, they effectively differentiate between normal and cancer cells, making them valuable tools for cancer diagnosis and potential therapeutic applications.
Collapse
Affiliation(s)
- Charutha Kalarikkal
- Main group Organometallics Optoelectronic Materials and Catalysis lab, Department of Chemistry, National Institute of Technology, Calicut, 673601, India.
| | - Anjali
- Protein Homeostasis Laboratory, Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Delhi-NCR, Greater Noida, Gautam Buddha Nagar, Uttar Pradesh 201314, India
| | - Sarbani Bhattacharjee
- Protein Homeostasis Laboratory, Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Delhi-NCR, Greater Noida, Gautam Buddha Nagar, Uttar Pradesh 201314, India
| | - Koyeli Mapa
- Protein Homeostasis Laboratory, Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Delhi-NCR, Greater Noida, Gautam Buddha Nagar, Uttar Pradesh 201314, India
| | - Chinna Ayya Swamy P
- Main group Organometallics Optoelectronic Materials and Catalysis lab, Department of Chemistry, National Institute of Technology, Calicut, 673601, India.
| |
Collapse
|
40
|
Chadokiya J, Chang K, Sharma S, Hu J, Lill JR, Dionne J, Kirane A. Advancing precision cancer immunotherapy drug development, administration, and response prediction with AI-enabled Raman spectroscopy. Front Immunol 2025; 15:1520860. [PMID: 39850874 PMCID: PMC11753970 DOI: 10.3389/fimmu.2024.1520860] [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: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 01/25/2025] Open
Abstract
Molecular characterization of tumors is essential to identify predictive biomarkers that inform treatment decisions and improve precision immunotherapy development and administration. However, challenges such as the heterogeneity of tumors and patient responses, limited efficacy of current biomarkers, and the predominant reliance on single-omics data, have hindered advances in accurately predicting treatment outcomes. Standard therapy generally applies a "one size fits all" approach, which not only provides ineffective or limited responses, but also an increased risk of off-target toxicities and acceleration of resistance mechanisms or adverse effects. As the development of emerging multi- and spatial-omics platforms continues to evolve, an effective tumor assessment platform providing utility in a clinical setting should i) enable high-throughput and robust screening in a variety of biological matrices, ii) provide in-depth information resolved with single to subcellular precision, and iii) improve accessibility in economical point-of-care settings. In this perspective, we explore the application of label-free Raman spectroscopy as a tumor profiling tool for precision immunotherapy. We examine how Raman spectroscopy's non-invasive, label-free approach can deepen our understanding of intricate inter- and intra-cellular interactions within the tumor-immune microenvironment. Furthermore, we discuss the analytical advances in Raman spectroscopy, highlighting its evolution to be utilized as a single "Raman-omics" approach. Lastly, we highlight the translational potential of Raman for its integration in clinical practice for safe and precise patient-centric immunotherapy.
Collapse
Affiliation(s)
- Jay Chadokiya
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, Stanford, CA, United States
| | - Kai Chang
- Department of Electrical Engineering, Stanford University,
Stanford, CA, United States
| | - Saurabh Sharma
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, Stanford, CA, United States
| | - Jack Hu
- Pumpkinseed Technologies, Palo Alto, CA, United States
| | | | - Jennifer Dionne
- Pumpkinseed Technologies, Palo Alto, CA, United States
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, United States
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, United States
| | - Amanda Kirane
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, Stanford, CA, United States
| |
Collapse
|
41
|
Fathi Kazerooni A, Kraya A, Rathi KS, Kim MC, Vossough A, Khalili N, Familiar AM, Gandhi D, Khalili N, Kesherwani V, Haldar D, Anderson H, Jin R, Mahtabfar A, Bagheri S, Guo Y, Li Q, Huang X, Zhu Y, Sickler A, Lueder MR, Phul S, Koptyra M, Storm PB, Ware JB, Song Y, Davatzikos C, Foster JB, Mueller S, Fisher MJ, Resnick AC, Nabavizadeh A. Multiparametric MRI along with machine learning predicts prognosis and treatment response in pediatric low-grade glioma. Nat Commun 2025; 16:340. [PMID: 39747214 PMCID: PMC11697432 DOI: 10.1038/s41467-024-55659-z] [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: 04/14/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Pediatric low-grade gliomas (pLGGs) exhibit heterogeneous prognoses and variable responses to treatment, leading to tumor progression and adverse outcomes in cases where complete resection is unachievable. Early prediction of treatment responsiveness and suitability for immunotherapy has the potential to improve clinical management and outcomes. Here, we present a radiogenomic analysis of pLGGs, integrating MRI and RNA sequencing data. We identify three immunologically distinct clusters, with one group characterized by increased immune activity and poorer prognosis, indicating potential benefit from immunotherapies. We develop a radiomic signature that predicts these immune profiles with over 80% accuracy. Furthermore, our clinicoradiomic model predicts progression-free survival and correlates with treatment response. We also identify genetic variants and transcriptomic pathways associated with progression risk, highlighting links to tumor growth and immune response. This radiogenomic study in pLGGs provides a framework for the identification of high-risk patients who may benefit from targeted therapies.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Adam Kraya
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Komal S Rathi
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Meen Chul Kim
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Arastoo Vossough
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Radiology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ariana M Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Deep Gandhi
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Neda Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Varun Kesherwani
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Debanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Hannah Anderson
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Run Jin
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aria Mahtabfar
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sina Bagheri
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yiran Guo
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Qi Li
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xiaoyan Huang
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuankun Zhu
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alex Sickler
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew R Lueder
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Saksham Phul
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mateusz Koptyra
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey B Ware
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuanquan Song
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica B Foster
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sabine Mueller
- Department of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Michael J Fisher
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
42
|
Wu G, Ying L, Zhang Q, Xiong H, Wang J, Chen S, Yang C, Jin Y, Lai Z, Feng N, Ge Y. Lipid droplet formation induced by icaritin derivative IC2 promotes a combination strategy for cancer therapy. Chin Med 2024; 19:178. [PMID: 39725994 PMCID: PMC11670343 DOI: 10.1186/s13020-024-01050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Lipid metabolism is crucial in cancer progression. Lipid droplets (LDs) generated in cancer cells can act as protective mechanisms through alleviating lipotoxicity under stress conditions. We previously developed IC2 from the Chinese medicine icaritin as an inhibitor of stearoyl-CoA desaturase 1 (SCD1). IC2 has been shown to disrupt lipid metabolism and inhibits cancer cell proliferation. However, the impact of IC2 on intracellular LDs and the potential of targeting LD formation for combination cancer therapy remain unexplored. METHODS LD formation in cancer cells was analyzed with oil red O or BODIPY staining by microscopy. LD quantification was normalized to the cell number. IC2-induced cellular responses were revealed by transcriptional analysis, real-time PCR, and immunoblotting. Mitochondrial functions were assessed by measuring ATP production and oxygen consumption. The lipid source for LD formation was studied using lipid transporter inhibitors or lipid deprivation. The effect of inhibiting LD formation on IC2's anti-tumor effects was evaluated using MTT assays and apoptosis assays, which was subsequently validated in an in vivo xenografted tumor model. RESULTS IC2 exerted anti-tumor effects, resulting in LD formation in various cancer cells. LD formation stimulated by IC2 was independent of extracellular lipid sources and did not result from increased de novo fatty acid (FA) synthesis within the cancer cells. Transcriptional analysis indicated that IC2 disturbed mitochondrial functions, which was confirmed by impaired mitochondrial membrane potential (MMP) and reduced capacity for ATP production and oxygen consumption. Moreover, IC2 treatment led to a greater accumulation of lipids in LDs outside the mitochondria compared with the control group. IC2 inhibited the proliferation of PC3 cells and promoted the apoptosis of the cancer cells. These effects were further enhanced after inhibiting the diacylglycerol acyltransferase 1 (DGAT1), a key intracellular enzyme involved in LD formation. In PC3-xenografted mice, the DGAT1 inhibitor augmented the IC2-induced reduction in tumor growth by modulating LD formation. CONCLUSION LD formation is a feedback response to IC2's anti-tumor effects, which compromises the anti-tumor actions. IC2's anti-tumor efficacy can be enhanced by combining it with inhibitors targeting LD formation. This strategy may be extended to other anti-tumor agents that regulate lipid metabolism.
Collapse
Affiliation(s)
- Guosheng Wu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Liang Ying
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Qian Zhang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - He Xiong
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jie Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Sitao Chen
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Chen Yang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou, Anhui, China
| | - Yiyuan Jin
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Taizhou Center for Disease Control and Prevention, Taizhou, China
| | - Zengwei Lai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Ninghan Feng
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Yunjun Ge
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
43
|
Chaudhary A, Patil P, Raina P, Kaul-Ghanekar R. Matairesinol repolarizes M2 macrophages to M1 phenotype to induce apoptosis in triple-negative breast cancer cells. Immunopharmacol Immunotoxicol 2024:1-15. [PMID: 39722605 DOI: 10.1080/08923973.2024.2425028] [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: 06/05/2024] [Accepted: 10/27/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Triple-Negative Breast Cancer (TNBC), the most challenging subtype of Breast Cancer (BC), currently lacks targeted therapy, presenting a significant therapeutic gap in its management. Tumor Associated Macrophages (TAMs) play a significant role in TNBC progression and could be targeted by repolarizing them from M2 to M1 phenotype. Matairesinol (MAT), a plant lignan, has been shown to exhibit anticancer, anti-inflammatory and immunomodulatory activities. In this study, we explored how MAT-induced repolarization of THP-1-derived M2 macrophages towards the M1 phenotype, which could effectively target the TNBC cell line, MDA-MB-231. METHODS The differential expression of genes in THP-1-derived macrophages at mRNA levels was evaluated by RNAseq assay. An inverted microscope equipped with a CMOS camera was utilized to capture the morphological variations in THP-1 cells and THP-1-derived macrophages. Relative mRNA expression of M1 and M2 specific marker genes was quantified by qRT-PCR. Cell viability and induction of apoptosis were evaluated by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1 dye) assays, respectively. RESULTS MAT reduced the viability of M2a and M2d macrophages and repolarized them to M1 phenotype. Conditioned medium (CM) from MAT-treated M2a and M2d macrophages significantly reduced the viability of TNBC cells by apoptosis. CONCLUSION Targeting M2 macrophages is an important strategy to regulate cancer progression. Our study provides evidence that MAT may be a promising drug candidate for developing novel anti-TNBC therapy. However, further studies are warranted to thoroughly elucidate the molecular mechanism of action of MAT and evaluate its therapeutic potential in TNBC in vitro and in vivo models.
Collapse
Affiliation(s)
- Amol Chaudhary
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prajakta Patil
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prerna Raina
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Analytical Department (ADT), Lupin Limited, Pune, India
| | - Ruchika Kaul-Ghanekar
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Symbiosis Centre for Research and Innovation (SCRI); Symbiosis International Deemed University (SIU), Pune, India
- Cancer Research Lab, Symbiosis School of Biological Sciences (SSBS), Symbiosis International Deemed University (SIU), Pune, India
| |
Collapse
|
44
|
Mao Y, Xia Z, Xia W, Jiang P. Metabolic reprogramming, sensing, and cancer therapy. Cell Rep 2024; 43:115064. [PMID: 39671294 DOI: 10.1016/j.celrep.2024.115064] [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: 07/04/2024] [Revised: 10/30/2024] [Accepted: 11/21/2024] [Indexed: 12/15/2024] Open
Abstract
The metabolic reprogramming of tumor cells is a crucial strategy for their survival and proliferation, involving tissue- and condition-dependent remodeling of certain metabolic pathways. While it has become increasingly clear that tumor cells integrate extracellular and intracellular signals to adapt and proliferate, nutrient and metabolite sensing also exert direct or indirect influences, although the underlying mechanisms remain incompletely understood. Furthermore, metabolic changes not only support the rapid growth and dissemination of tumor cells but also promote immune evasion by metabolically "educating" immune cells in the tumor microenvironment (TME). Recent studies have highlighted the profound impact of metabolic reprogramming on the TME and the potential of targeting metabolic pathways as a therapeutic strategy, with several enzyme inhibitors showing promising results in clinical trials. Thus, understanding how tumor cells alter their metabolic pathways and metabolically remodel the TME to support their survival and proliferation may offer new strategies for metabolic therapy and immunotherapy.
Collapse
Affiliation(s)
- Youxiang Mao
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ziyan Xia
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Wenjun Xia
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Peng Jiang
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
45
|
Lagal DJ, Ortiz-Alcántara Á, Pedrajas JR, McDonagh B, Bárcena JA, Requejo-Aguilar R, Padilla CA. Loss of peroxiredoxin 6 alters lipid composition and distribution resulting in increased sensitivity to ferroptosis. Biochem J 2024; 481:1997-2015. [PMID: 39601357 DOI: 10.1042/bcj20240445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/03/2024] [Accepted: 11/27/2024] [Indexed: 11/29/2024]
Abstract
Peroxiredoxin 6 (PRDX6) is a multifunctional enzyme involved in phospholipid peroxide repair and metabolism. In this study we investigated the global lipid composition of a human hepatocarcinoma cell line SNU475 lacking PRDX6 and lipid related cellular processes. There was a general decrease in multiple lipids species upon loss of PRDX6, in particular sphingomyelins and acylcarnitines, consistent with previously observed alterations in cell signaling pathways and mitochondrial dysfunction. Deprivation of docosahexaenoic acid and related species was also evident. However, a few striking exceptions are worth highlighting: (1) Three specific arachidonic acid (AA) containing phophatidylcholines (PC) increased significantly. The increase of sn1-stearic/sn2-PUFA containing PC and sn2-AA containing plasmenyls are indicative of a preference of PRDX6 iPLA2 activity for these AA storage glycerophospholipids. (2) Several polyunsaturated fatty acids (PUFA) and PUFA containing triacylglycerols accumulated together with increased formation of lipid droplets, an indication of altered FA flux and PUFA sequestration in PRDX6 knockout cells. Loss of PRDX6 resulted in increased sensitivity to erastin-induced ferroptosis, independent of selenium and GPX4, as a consequence of increased levels of lipid hydroperoxides, that reverted to normal levels upon rescue with PRDX6. The results presented demonstrate that all three enzymatic activities of PRDX6 contribute to the role of this multifunctional enzyme in diverse cellular processes, including membrane phospholipid remodeling and glycerophospholipid functional diversity, resulting in altered lipid peroxides and modulation of AA disposition and traffic. These contributions highlight the complexity of the changes that loss of PRDX6 exerts on cell functionality.
Collapse
Affiliation(s)
- Daniel J Lagal
- Department of Biochemistry and Molecular Biology, University of Córdoba, Cordoba, Spain
| | - Ángel Ortiz-Alcántara
- Department of Biochemistry and Molecular Biology, University of Córdoba, Cordoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Cordoba, Spain
| | - José R Pedrajas
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, University Institute of Research in Olive Groves and Olive Oils, University of Jaén, Jaen, Spain
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland
| | - J Antonio Bárcena
- Department of Biochemistry and Molecular Biology, University of Córdoba, Cordoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Cordoba, Spain
| | - Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Córdoba, Cordoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Cordoba, Spain
| | - C Alicia Padilla
- Department of Biochemistry and Molecular Biology, University of Córdoba, Cordoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Cordoba, Spain
| |
Collapse
|
46
|
Zhang J, Liu H, Wu Q, Liu T, Liu X, Cai J, Yi X, Wang Z, Gao L. Exosomal ANXA2 facilitates ovarian cancer peritoneal metastasis by activating peritoneal mesothelial cells through binding with TLR2. Cell Commun Signal 2024; 22:616. [PMID: 39709496 DOI: 10.1186/s12964-024-01987-y] [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: 08/21/2024] [Accepted: 12/07/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Peritoneal dissemination of ovarian cancer (OvCa) can be largely attributed to the formation of a metastatic microenvironment driven by tumoral exosomes. Here, we aimed to elucidate the mechanisms through which exosomal annexin A2 (ANXA2) derived from OvCa cells induces an HPMC phenotypic shift in favour of peritoneal metastasis. METHODS Immunohistochemistry and orthotopic and intraperitoneal OvCa xenograft mouse models were used to clarify the relationship between tumour ANXA2 expression and peritoneal metastasis. Exosomes were isolated from OvCa cell lines via ultracentrifugation. Functional experiments on cell proliferation and motility, and western blot were performed to investigate the activation of HPMCs and its impact on tumour cell in vitro. High-throughput transcriptional sequencing and rescue experiments in which ANXA2 inhibitor (LCKLSL) or the toll-like receptor 2 (TLR2) inhibitor (C29) was used to co-culture the HPMCs with exosome were employed to identify the crucial functional molecules through which exosomal ANXA2 activates HPMCs. The impact of exosomal ANXA2-activated HPMCs on tumour progression was assessed via functional experiments. RESULTS Primary OvCa samples with high ANXA2 expression exhibited a stronger tendency to metastasize to the abdominal cavity. Tumoral ANXA2 promoted OvCa peritoneal metastasis through the secretion of exosomes carrying ANXA2. ANXA2-loaded exosomes activated HPMCs through exosomal ANXA2 binding to TLR2, shifting the phenotype of HPMCs towards mesenchymal cells, increasing their migration and invasion capacities, and elevating the expression of lipocalin 2 (LCN2). High LCN2 expression in HPMCs promoted OvCa cell adhesion, proliferation, motility, and lipid metabolism reprogramming. CONCLUSION Exosomal ANXA2 secreted by tumour cells activates HPMCs and induces the expression of LCN2, which in turn promotes the peritoneal metastasis of OvCa.
Collapse
Affiliation(s)
- Jingni Zhang
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongmei Liu
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiulei Wu
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tong Liu
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoli Liu
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Cai
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoqing Yi
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zehua Wang
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Lingling Gao
- Departmentof Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
47
|
Ko K, Bandara SR, Zhou W, Svenningsson L, Porras-Gómez M, Kambar N, Dreher-Threlkeld J, Topgaard D, Hernández-Saavedra D, Anakk S, Leal C. Diet-Induced Obesity Modulates Close-Packing of Triacylglycerols in Lipid Droplets of Adipose Tissue. J Am Chem Soc 2024; 146:34796-34810. [PMID: 39644234 DOI: 10.1021/jacs.4c13420] [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: 12/09/2024]
Abstract
Adipose-derived lipid droplets (LDs) are rich in triacylglycerols (TAGs), which regulate essential cellular processes, such as energy storage. Although TAG accumulation and LD expansion in adipocytes occur during obesity, how LDs dynamically package TAGs in response to excessive nutrients remains elusive. Here, we found that LD lipidomes display a remarkable increase in TAG acyl chain saturation under calorie-dense diets, turning them conducive to close-packing. Using high-resolution X-ray diffraction, solid-state NMR, and imaging, we show that beyond size expansion LDs from mice under varied obesogenic diets govern fat accumulation by packing TAGs in different crystalline polymorphs. Consistently, LDs and tissue stiffen for high-calorie-fed mice with more than a 2-fold increase in elastic moduli compared to normal diet. Our data suggest that in addition to expanding, adipocyte LDs undergo structural remodeling by close-packing rigid and highly saturated TAGs in response to caloric overload, as opposed to liquid TAGs in a low-calorie diet. This work provides insights into how lipid packing within LDs can allow for the rapid and optimal expansion of fat during the initial stages of obesity.
Collapse
Affiliation(s)
- Kyungwon Ko
- Department of Bioengineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Sarith R Bandara
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Weinan Zhou
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Leo Svenningsson
- Division of Physical Chemistry, Lund University, Lund 22100, Sweden
| | - Marilyn Porras-Gómez
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Nurila Kambar
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Julia Dreher-Threlkeld
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Daniel Topgaard
- Division of Physical Chemistry, Lund University, Lund 22100, Sweden
| | - Diego Hernández-Saavedra
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Sayeepriyadarshini Anakk
- Department of Bioengineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Cecília Leal
- Department of Bioengineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Materials Science and Engineering, Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
48
|
Sadhukhan P, Feng M, Illingworth E, Sloma I, Ooki A, Matoso A, Sidransky D, Johnson BA, Marchionni L, Sillé FC, Choi W, McConkey D, Hoque MO. YAP1 induces bladder cancer progression and promotes immune evasion through IL-6/STAT3 pathway and CXCL deregulation. J Clin Invest 2024; 135:e171164. [PMID: 39630608 PMCID: PMC11735109 DOI: 10.1172/jci171164] [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: 04/05/2023] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
The Hippo signaling pathway plays a key role in tumorigenesis in different cancer types. We investigated the role of the Hippo effector YAP1 in the tumor immune microenvironment (TIME) of urothelial carcinoma of the bladder (UCB) and evaluated the efficacy of immunotherapy in the context of YAP1 signaling. We performed numerous in vitro and in vivo experiments to determine the role of YAP1 using genetic and pharmacological attenuation of YAP1 activity. Briefly, RNA sequencing was carried out with mouse and human cell lines to identify novel YAP1-regulated downstream targets unbiasedly. We then experimentally confirmed that YAP1 regulates the TIME through the IL-6/STAT3 signaling pathway and varied C-X-C motif chemokine regulation. We analyzed several human sample sets to explore the TIME status in the context of YAP1 expression. Our data indicate that YAP1 attenuation decreases M2 macrophages and myeloid-derived suppressor cells in the TIME compared with YAP1-expressing cells. In summary, this study provides insights into YAP1 signaling as a driver for cancer stemness and an inducer of immunosuppressive TIME. Moreover, the therapeutic efficacy of YAP1 attenuation indicates that combined blockade of YAP1 and immune checkpoints may yield clinical value for treating patients with UCB.
Collapse
Affiliation(s)
| | - Mingxiao Feng
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Emily Illingworth
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ido Sloma
- Champions Oncology, R&D, Baltimore, Maryland, USA
| | - Akira Ooki
- Department of Otolaryngology–Head and Neck Surgery and
| | | | - David Sidransky
- Department of Otolaryngology–Head and Neck Surgery and
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Burles A. Johnson
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Luigi Marchionni
- Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Fenna C.M. Sillé
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Woonyoung Choi
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David McConkey
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohammad O. Hoque
- Department of Otolaryngology–Head and Neck Surgery and
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
49
|
Bisogno S, Depciuch J, Gulzar H, Heber MF, Kobiałka M, Gąsior Ł, Bereta A, Pieczara A, Fic K, Musson R, Garcia Gamero G, Pardo Martinez M, Fornés Pérez A, Tatíčková M, Holubcova Z, Barańska M, Ptak GE. Female-age-dependent changes in the lipid fingerprint of the mammalian oocytes. Hum Reprod 2024; 39:2754-2767. [PMID: 39366679 PMCID: PMC11630086 DOI: 10.1093/humrep/deae225] [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: 01/15/2024] [Revised: 09/09/2024] [Indexed: 10/06/2024] Open
Abstract
STUDY QUESTION Can oocyte functionality be assessed by observing changes in their intracytoplasmic lipid droplets (LDs) profiles? SUMMARY ANSWER Lipid profile changes can reliably be detected in human oocytes; lipid changes are linked with maternal age and impaired developmental competence in a mouse model. WHAT IS KNOWN ALREADY In all cellular components, lipid damage is the earliest manifestation of oxidative stress (OS), which leads to a cascade of negative consequences for organelles and DNA. Lipid damage is marked by the accumulation of LDs. We hypothesized that impaired oocyte functionality resulting from aging and associated OS could be assessed by changes in LDs profile, hereafter called lipid fingerprint (LF). STUDY DESIGN, SIZE, DURATION To investigate if it is possible to detect differences in oocyte LF, we subjected human GV-stage oocytes to spectroscopic examinations. For this, a total of 48 oocytes derived from 26 young healthy women (under 33 years of age) with no history of infertility, enrolled in an oocyte donation program, were analyzed. Furthermore, 30 GV human oocytes from 12 women were analyzed by transmission electron microscopy (TEM). To evaluate the effect of oocytes' lipid profile changes on embryo development, a total of 52 C57BL/6 wild-type mice and 125 Gnpat+/- mice were also used. PARTICIPANTS/MATERIALS, SETTING, METHODS Human oocytes were assessed by label-free cell imaging via coherent anti-Stokes Raman spectroscopy (CARS). Further confirmation of LF changes was conducted using spontaneous Raman followed by Fourier transform infrared (FTIR) spectroscopies and TEM. Additionally, to evaluate whether LF changes are associated with developmental competence, mouse oocytes and blastocysts were evaluated using TEM and the lipid dyes BODIPY and Nile Red. Mouse embryonic exosomes were evaluated using flow cytometry, FTIR and FT-Raman spectroscopies. MAIN RESULTS AND THE ROLE OF CHANCE Here we demonstrated progressive changes in the LF of oocytes associated with the woman's age consisting of increased LDs size, area, and number. LF variations in oocytes were detectable also within individual donors. This finding makes LF assessment a promising tool to grade oocytes of the same patient, based on their quality. We next demonstrated age-associated changes in oocytes reflected by lipid peroxidation and composition changes; the accumulation of carotenoids; and alterations of structural properties of lipid bilayers. Finally, using a mouse model, we showed that LF changes in oocytes are negatively associated with the secretion of embryonic exosomes prior to implantation. Deficient exosome secretion disrupts communication between the embryo and the uterus and thus may explain recurrent implantation failures in advanced-age patients. LIMITATIONS, REASONS FOR CAUTION Due to differences in lipid content between different species' oocytes, the developmental impact of lipid oxidation and consequent LF changes may differ across mammalian oocytes. WIDER IMPLICATIONS OF THE FINDINGS Our findings open the possibility to develop an innovative tool for oocyte assessment and highlight likely functional connections between oocyte LDs and embryonic exosome secretion. By recognizing the role of oocyte LF in shaping the embryo's ability to implant, our original work points to future directions of research relevant to developmental biology and reproductive medicine. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by National Science Centre of Poland, Grants: 2021/41/B/NZ3/03507 and 2019/35/B/NZ4/03547 (to G.E.P.); 2022/44/C/NZ4/00076 (to M.F.H.) and 2019/35/N/NZ3/03213 (to Ł.G.). M.F.H. is a National Agency for Academic Exchange (NAWA) fellow (GA ULM/2019/1/00097/U/00001). K.F. is a Diamond Grant fellow (Ministry of Education and Science GA 0175/DIA/2019/28). The open-access publication of this article was funded by the Priority Research Area BioS under the program "Excellence Initiative - Research University" at the Jagiellonian University in Krakow. The authors declare no competing interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Simona Bisogno
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Joanna Depciuch
- Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Hafsa Gulzar
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Kraków, Poland
| | - Maria Florencia Heber
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Michał Kobiałka
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Łukasz Gąsior
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Adrianna Bereta
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Anna Pieczara
- Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Kraków, Poland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University in Krakow, Kraków, Poland
| | - Kinga Fic
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Richard Musson
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Kraków, Poland
| | - Gabriel Garcia Gamero
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Maria Pardo Martinez
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Alba Fornés Pérez
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Martina Tatíčková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Holubcova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Reprofit International, Brno, Czech Republic
| | - Małgorzata Barańska
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University in Krakow, Kraków, Poland
- Faculty of Chemistry, Jagiellonian University in Kraków, Kraków, Poland
| | - Grażyna Ewa Ptak
- Malopolska Centre of Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| |
Collapse
|
50
|
Soares VC, Dias SSG, Santos JC, Bozza PT. Unlocking secrets: lipid metabolism and lipid droplet crucial roles in SARS-CoV-2 infection and the immune response. J Leukoc Biol 2024; 116:1254-1268. [PMID: 39087951 DOI: 10.1093/jleuko/qiae170] [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: 01/31/2024] [Revised: 06/11/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024] Open
Abstract
Lipid droplets (LDs) are crucial for maintaining lipid and energy homeostasis within cells. LDs are highly dynamic organelles that present a phospholipid monolayer rich in neutral lipids. Additionally, LDs are associated with structural and nonstructural proteins, rapidly mobilizing lipids for various biological processes. Lipids play a pivotal role during viral infection, participating during viral membrane fusion, viral replication, and assembly, endocytosis, and exocytosis. SARS-CoV-2 infection often induces LD accumulation, which is used as a source of energy for the replicative process. These findings suggest that LDs are a hallmark of viral infection, including SARS-CoV-2 infection. Moreover, LDs participate in the inflammatory process and cell signaling, activating pathways related to innate immunity and cell death. Accumulating evidence demonstrates that LD induction by SARS-CoV-2 is a highly coordinated process, aiding replication and evading the immune system, and may contribute to the different cell death process observed in various studies. Nevertheless, recent research in the field of LDs suggests these organelles according to the pathogen and infection conditions may also play roles in immune and inflammatory responses, protecting the host against viral infection. Understanding how SARS-CoV-2 influences LD biogenesis is crucial for developing novel drugs or repurposing existing ones. By targeting host lipid metabolic pathways exploited by the virus, it is possible to impact viral replication and inflammatory responses. This review seeks to discuss and analyze the role of LDs during SARS-CoV-2 infection, specifically emphasizing their involvement in viral replication and the inflammatory response.
Collapse
Affiliation(s)
- Vinicius Cardoso Soares
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Program of Immunology and Inflammation, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Suelen Silva Gomes Dias
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Julia Cunha Santos
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| |
Collapse
|