1
|
Zhang Y, Chen Y, Zhuang C, Qi J, Zhao RC, Wang J. Lipid droplets in the nervous system: involvement in cell metabolic homeostasis. Neural Regen Res 2025; 20:740-750. [PMID: 38886939 PMCID: PMC11433920 DOI: 10.4103/nrr.nrr-d-23-01401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/20/2024] [Indexed: 06/20/2024] Open
Abstract
Lipid droplets serve as primary storage organelles for neutral lipids in neurons, glial cells, and other cells in the nervous system. Lipid droplet formation begins with the synthesis of neutral lipids in the endoplasmic reticulum. Previously, lipid droplets were recognized for their role in maintaining lipid metabolism and energy homeostasis; however, recent research has shown that lipid droplets are highly adaptive organelles with diverse functions in the nervous system. In addition to their role in regulating cell metabolism, lipid droplets play a protective role in various cellular stress responses. Furthermore, lipid droplets exhibit specific functions in neurons and glial cells. Dysregulation of lipid droplet formation leads to cellular dysfunction, metabolic abnormalities, and nervous system diseases. This review aims to provide an overview of the role of lipid droplets in the nervous system, covering topics such as biogenesis, cellular specificity, and functions. Additionally, it will explore the association between lipid droplets and neurodegenerative disorders. Understanding the involvement of lipid droplets in cell metabolic homeostasis related to the nervous system is crucial to determine the underlying causes and in exploring potential therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Yuchen Zhang
- School of Life Sciences, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Yiqing Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Cheng Zhuang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jingxuan Qi
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, China
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
2
|
Fu S, Yu R, Yang B, Han X, Xu Y, Miao J. Hypoxia-inducible lipid droplet-associated protein (HILPDA) and cystathionine β-synthase (CBS) co-contribute to protecting intestinal epithelial cells from Staphylococcus aureus via regulating lipid droplets formation. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159558. [PMID: 39173873 DOI: 10.1016/j.bbalip.2024.159558] [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: 04/13/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Despite Staphylococcus aureus (S. aureus) being a highly studied zoontic bacterium, its enteropathogenicity remains elusive. Herein, our findings demonstrated that S. aureus infection led to the accumulation of lipid droplets (LDs) in intestinal epithelial cells, accompanied by marked elevation inflammatory response that ultimately decreases intracellular bacterial load. The aforestated phenomenon may be partly attributed to the up-regulation of hypoxia-inducible lipid droplet-associated protein (HILPDA) and the concomitant down-regulation of cystathionine β-synthase (CBS) protein. Moreover, S. aureus infection up-regulated the expression of HILPDA, thereby promoting LDs accumulation, and down-regulated that of CBS, consequently inhibiting microsomal triglyceride transfer protein (MTTP) expression. This process may suppress the transport of LDs to the extracellular environment, further contributing to the formation of intracellular LDs. In summary, the results of this study provide significant insights into the intricate mechanisms through which the host organism combats pathogens and maintains the balance of sulfur and lipid metabolism. These findings not only enhance our understanding of the host's defense mechanisms but also offer promising avenues for the development of novel strategies to combat intestinal infectious diseases.
Collapse
Affiliation(s)
- Shaodong Fu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Rui Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Bo Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
3
|
Chen L, Hao J, Zhang J, Wu J, Ren Z. Rosiglitazone-induced white adipocyte browning is regulated by actin and Myh9. Life Sci 2024; 359:123217. [PMID: 39510170 DOI: 10.1016/j.lfs.2024.123217] [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: 08/03/2024] [Revised: 10/22/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
AIMS This study investigates the role of actin polymerization and Myh9 in mediating lipid droplet (LD) fission during rosiglitazone-induced browning of white adipocytes. The aim is to understand how LD splitting might contribute to the beige conversion of white adipose tissue, providing insights into adipocyte plasticity and metabolic regulation. MATERIALS AND METHODS C3H10 T1/2-differentiated adipocytes were used as a classical model to study white adipocyte browning. Rosiglitazone was applied to induce browning, and the interactions between LDs and actin, as well as the distribution of Myh9, were assessed using immunofluorescence and Western blotting. In vivo, we employed a microfilament inhibitor to block actin polymerization in cold-stimulated mice and evaluated changes in LD morphology and browning. Furthermore, dynamic live-cell imaging using confocal microscopy was conducted to observe the real-time behavior of LDs during the browning process and to determine whether they undergo fission. MAIN FINDINGS Our results demonstrate that rosiglitazone significantly induces LD size reduction, a process correlated with the increased contact of LDs with microfilaments. Inhibition of actin polymerization prevented both the reduction in LD size and the browning of white adipocytes, indicating that actin plays a critical role. Myh9 was enriched at the LD fission sites, forming a structure resembling a contractile ring. Overexpression of Myh9 promoted the shrinkage of LD, suggesting that it may be involved in LD fission. SIGNIFICANCE This study identifies actin and Myh9 as key regulators of LD fission in rosiglitazone-induced browning of white adipocytes, offering new insights into the cellular mechanisms of adipocyte plasticity. The findings propose a novel pathway by which LD dynamics contribute to the beige conversion of white fat, with potential implications for metabolic disease therapies targeting adipocyte function and energy expenditure.
Collapse
Affiliation(s)
- Lupeng Chen
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jingjie Hao
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Junzhi Zhang
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China.
| |
Collapse
|
4
|
Li M, Zhao P, Wang J, Zhang X, Li J. Functional antimicrobial peptide-loaded 3D scaffolds for infected bone defect treatment with AI and multidimensional printing. MATERIALS HORIZONS 2024. [PMID: 39484845 DOI: 10.1039/d4mh01124d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infection is the most prevalent complication of fractures, particularly in open fractures, and often leads to severe consequences. The emergence of bacterial resistance has significantly exacerbated the burden of infection in clinical practice, making infection control a significant treatment challenge for infectious bone defects. The implantation of a structural stent is necessary to treat large bone defects despite the increased risk of infection. Therefore, there is a need for the development of novel antibacterial therapies. The advancement in antibacterial biomaterials and new antimicrobial drugs offers fresh perspectives on antibacterial treatment. Although antimicrobial 3D scaffolds are currently under intense research focus, relying solely on material properties or antibiotic action remains insufficient. Antimicrobial peptides (AMPs) are one of the most promising new antibacterial therapy approaches. This review discusses the underlying mechanisms behind infectious bone defects and presents research findings on antimicrobial peptides, specifically emphasizing their mechanisms and optimization strategies. We also explore the potential prospects of utilizing antimicrobial peptides in treating infectious bone defects. Furthermore, we propose that artificial intelligence (AI) algorithms can be utilized for predicting the pharmacokinetic properties of AMPs, including absorption, distribution, metabolism, and excretion, and by combining information from genomics, proteomics, metabolomics, and clinical studies with computational models driven by machine learning algorithms, scientists can gain a comprehensive understanding of AMPs' mechanisms of action, therapeutic potential, and optimizing treatment strategies tailored to individual patients, and through interdisciplinary collaborations between computer scientists, biologists, and clinicians, the full potential of AI in accelerating the discovery and development of novel AMPs will be realized. Besides, with the continuous advancements in 3D/4D/5D/6D technology and its integration into bone scaffold materials, we anticipate remarkable progress in the field of regenerative medicine. This review summarizes relevant research on the optimal future for the treatment of infectious bone defects, provides guidance for future novel treatment strategies combining multi-dimensional printing with new antimicrobial agents, and provides a novel and effective solution to the current challenges in the field of bone regeneration.
Collapse
Affiliation(s)
- Mengmeng Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Peizhang Zhao
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jingwen Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xincai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
5
|
Rouvray S, Drummond RA. The role of lipids in regulating macrophage antifungal immunity. mBio 2024; 15:e0305723. [PMID: 39207168 PMCID: PMC11481918 DOI: 10.1128/mbio.03057-23] [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: 09/04/2024] Open
Abstract
Macrophages are critical components of the antifungal immune response. Disturbance in the number or function of these innate immune cells can significantly increase susceptibility to invasive fungal infections. Pathogenic fungi cause billions of infections every year and have an unmet clinical need, with many infections associated with unacceptably high mortality rates that primarily affect vulnerable patients with underlying immune defects. Lipid metabolism has been increasingly appreciated to significantly influence macrophage function, particularly of macrophages residing in lipid-rich organs, such as the brain, or macrophages specialized at clearing dead cells including alveolar macrophages in the lungs. In this review, we provide an overview of macrophage lipid metabolism, and discuss how lipid recycling and dysregulation affect key macrophage functions relevant for antifungal immunity including phagocytosis, functional polarization, and inflammasome activation. We focus on the fungal pathogen Cryptococcus neoformans, as this is the most common cause of death from fungal infection in humans and because several lines of evidence have already linked lipid metabolism in the regulation of C. neoformans and macrophage interactions.
Collapse
Affiliation(s)
- Sophie Rouvray
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Rebecca A. Drummond
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
6
|
Simwela NV, Jaecklein E, Sassetti CM, Russell DG. Impaired fatty acid import or catabolism in macrophages restricts intracellular growth of Mycobacterium tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604660. [PMID: 39091727 PMCID: PMC11291043 DOI: 10.1101/2024.07.22.604660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Mycobacterium tuberculosis (Mtb) infection of macrophages reprograms cellular metabolism to promote lipid retention. While it is clearly known that intracellular Mtb utilize host derived lipids to maintain infection, the role of macrophage lipid processing on the bacteria's ability to access the intracellular lipid pool remains undefined. We utilized a CRISPR-Cas9 genetic approach to assess the impact of sequential steps in fatty acid metabolism on the growth of intracellular Mtb. Our analyzes demonstrate that mutated macrophages that cannot either import, store or catabolize fatty acids restrict Mtb growth by both common and divergent anti-microbial mechanisms, including increased glycolysis, increased oxidative stress, production of pro-inflammatory cytokines, enhanced autophagy and nutrient limitation. We also show that impaired macrophage lipid droplet biogenesis is restrictive to Mtb replication, but increased induction fails to rescue Mtb growth. Our work expands our understanding of how host fatty acid homeostasis impacts Mtb growth in the macrophage.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Eleni Jaecklein
- Department of Microbiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | | | - David G. Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
7
|
Zhang F, Jiang Q, Cai J, Meng F, Tang W, Liu Z, Lin X, Liu W, Zhou Y, Shen X, Xue R, Dong L, Zhang S. Activation of NOD1 on tumor-associated macrophages augments CD8 + T cell-mediated antitumor immunity in hepatocellular carcinoma. SCIENCE ADVANCES 2024; 10:eadp8266. [PMID: 39356756 PMCID: PMC11446285 DOI: 10.1126/sciadv.adp8266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024]
Abstract
The efficacy of immunotherapy targeting the PD-1/PD-L1 pathway in hepatocellular carcinoma (HCC) is limited. NOD-like receptors (NLRs) comprise a highly evolutionarily conserved family of cytosolic bacterial sensors, yet their impact on antitumor immunity against HCC remains unclear. In this study, we uncovered that NOD1, a well-studied member of NLR family, exhibits predominant expression in tumor-associated macrophages (TAMs) and correlates positively with improved prognosis and responses to anti-PD-1 treatments in patients with HCC. Activation of NOD1 in vivo augments antitumor immunity and enhances the effectiveness of anti-PD-1 therapy. Mechanistically, NOD1 activation resulted in diminished expression of perilipin 5, thereby hindering fatty acid oxidation and inducing free fatty acid accumulation in TAMs. This metabolic alteration promoted membrane localization of the costimulatory molecule OX40L in a lipid modification-dependent manner, thereby activating CD8+ T cells. These findings unveil a previously unrecognized role for NOD1 in fortifying antitumor T cell immunity in HCC, potentially advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Qiuyu Jiang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Jialiang Cai
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Fansheng Meng
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Wenqing Tang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Zhiyong Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Xiahui Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Wenfeng Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Yi Zhou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Liver Disease, 180 Fenglin Road, Shanghai, 200032, P.R. China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dongan Road, Shanghai, 200030, P.R. China
| |
Collapse
|
8
|
Corbo JH, Chung J. Mechanisms of lipid droplet degradation. Curr Opin Cell Biol 2024; 90:102402. [PMID: 39053179 DOI: 10.1016/j.ceb.2024.102402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Lipid droplets (LDs) are subcellular organelles that play an integral role in lipid metabolism by regulating the storage and release of fatty acids, which are essential for energy production and various cellular processes. Lipolysis and lipophagy are the two major LD degradation pathways that mediate the utilization of lipids stored in these organelles. Recent studies have further uncovered alternative pathways, including direct lysosomal LD degradation and LD exocytosis. Here, we highlight recent findings that dissect the molecular basis of these diverse LD degradation pathways. Then, we discuss speculations on the crosstalk among these pathways and the potential unconventional roles of LD degradation.
Collapse
Affiliation(s)
- J H Corbo
- Department of Molecular and Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - J Chung
- Department of Molecular and Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA.
| |
Collapse
|
9
|
Liu D, Fang G, Wang Y, Meng C, Liu Z, Chen Q, Shao X. Facile construction of dual-response super-resolution probes for tracking organelles dynamics. EXPLORATION (BEIJING, CHINA) 2024; 4:20230145. [PMID: 39439499 PMCID: PMC11491301 DOI: 10.1002/exp.20230145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/09/2023] [Indexed: 10/25/2024]
Abstract
Super-resolution imaging techniques, such as structured illumination microscopy (SIM), have enabled researchers to obtain nanoscale organelle-level outputs in living systems, but they impose additional stringent requirements on fluorescence probes. However, high-performance, custom-designed SIM probes that can explain underlying biological processes remain unavailable. Herein, a customizable engineering toolkit is developed for the facile assembly of SIM probes suitable for subcellular component detection. This toolkit is used to customize a fluorescent molecule, CPC (coumarin-phenylhydrazine-carboxyl), capable of simultaneously monitoring peroxynitrite (ONOO-) and polarity distribution in mitochondria and lipid droplets (LDs), respectively, through functional ON-OFF mechanisms. The customized CPC molecule demonstrated excellent imaging capabilities under SIM, enabled the successful localization of multiple organelles, and reliably tracked the distribution of different components, thus facilitating the study of the interplay between organelles. Using CPC, the physical transition of intracellular LDs is demonstrated from heterogeneity to homogeneity. This was specifically observed during ferroptosis where the polarity of the LDs increased and their morphology became more contracted. Furthermore, the loss of LDs functionality could not counteract the accumulation of ONOO- within the mitochondria, leading to the decoupling of mitochondrial LDs during ferroptosis. These results confirmed the potential mechanism of LDs dysfunction and decoupling triggered via cumulative mitochondrial oxidative stress during ferroptosis. To summarize, this toolkit will be a powerful tool for examining subtle variations among components during the interplay between different organelles, thus offering novel avenues for understanding and treating related diseases.
Collapse
Affiliation(s)
- Daili Liu
- School of Chinese Materia MedicaTianjin University of Traditional Chinese MedicineTianjinChina
- Institute of Materia MedicaScience and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Guiqian Fang
- Institute of Materia MedicaScience and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Yanfeng Wang
- Institute of Materia MedicaScience and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Caicai Meng
- School of Life SciencesScience and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Zhidong Liu
- School of Chinese Materia MedicaTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Qixin Chen
- Institute of Materia MedicaScience and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
- Departments of Diagnostic RadiologyChemical and Biomolecular Engineeringand Biomedical EngineeringYong Loo Lin School of Medicine and Faculty of EngineeringNational University of SingaporeSingaporeSingapore
| | - Xintian Shao
- School of Life SciencesScience and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|
10
|
Lan Z, Lv S, Ge Z, Zhao B, Li L, Li C. Lactic acid regulates lipid droplet aggregation through a microglia-neuron axis in neuroinflammation. J Lipid Res 2024; 65:100629. [PMID: 39182605 PMCID: PMC11437955 DOI: 10.1016/j.jlr.2024.100629] [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: 01/20/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/27/2024] Open
Abstract
Neuroinflammation, marked by the release of proinflammatory cytokines and resulting neuronal death, is a multifaceted process extending beyond traditional inflammatory pathways. Microglia, primary cells in the inflammatory response, rapidly activate during neuroinflammation and produce proinflammatory and cytotoxic factors that affect neuronal function. Recent evidence highlights the significant role of abnormal lipid droplet (LD) deposition in the pathogenesis of neuroinflammation. While microglia are known to influence LD aggregation during neuroinflammation, the regulatory mechanism within neurons is not well understood. Our study demonstrates that lipopolysaccharide-activated microglia induce the accumulation of LD in neurons, identifying microglial-derived lactic acid as a key mediator in this process. Excessive lipid accumulation threatens neuronal function, a phenomenon reversed by eliminating microglia. Our study demonstrates that lipopolysaccharide-activated microglia induce the accumulation of LD in neurons, identifying microglial-derived lactic acid as a key mediator in this process. Excessive lipid accumulation threatens neuronal function, a phenomenon reversed by eliminating microglia.
Collapse
Affiliation(s)
- Zhuoqing Lan
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Shukai Lv
- Department of General Medicine, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ziyi Ge
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bing Zhao
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Leilei Li
- Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Caixia Li
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
11
|
Tang J, Song H, Li S, Lam SM, Ping J, Yang M, Li N, Chang T, Yu Z, Liu W, Lu Y, Zhu M, Tang Z, Liu Z, Guo YR, Shui G, Veillette A, Zeng Z, Wu N. TMEM16F Expressed in Kupffer Cells Regulates Liver Inflammation and Metabolism to Protect Against Listeria Monocytogenes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402693. [PMID: 39136057 PMCID: PMC11497084 DOI: 10.1002/advs.202402693] [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: 03/14/2024] [Revised: 06/20/2024] [Indexed: 10/25/2024]
Abstract
Infection by bacteria leads to tissue damage and inflammation, which need to be tightly controlled by host mechanisms to avoid deleterious consequences. It is previously reported that TMEM16F, a calcium-activated lipid scramblase expressed in various immune cell types including T cells and neutrophils, is critical for the control of infection by bacterium Listeria monocytogenes (Lm) in vivo. This function correlated with the capacity of TMEM16F to repair the plasma membrane (PM) damage induced in T cells in vitro, by the Lm toxin listeriolysin O (LLO). However, whether the protective effect of TMEM16F on Lm infection in vivo is mediated by an impact in T cells, or in other cell types, is not determined. Herein, the immune cell types and mechanisms implicated in the protective effect of TMEM16F against Lm in vivo are elucidated. Cellular protective effects of TMEM16F correlated with its capacity of lipid scrambling and augment PM fluidity. Using cell type-specific TMEM16F-deficient mice, the indication is obtained that TMEM16F expressed in liver Kupffer cells (KCs), but not in T cells or B cells, is key for protection against Listeria in vivo. In the absence of TMEM16F, Listeria induced PM rupture and fragmentation of KCs in vivo. KC death associated with greater liver damage, inflammatory changes, and dysregulated liver metabolism. Overall, the results uncovered that TMEM16F expressed in Kupffer cells is crucial to protect the host against Listeria infection. This influence is associated with the capacity of Kupffer cell-expressed TMEM16F to prevent excessive inflammation and abnormal liver metabolism.
Collapse
Affiliation(s)
- Jianlong Tang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Hua Song
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
| | - Shimin Li
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - Jieming Ping
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Mengyun Yang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Na Li
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Teding Chang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ze Yu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Weixiang Liu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Yan Lu
- Department of Clinical ImmunologyThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Min Zhu
- Department of Thoracic SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zhaohui Tang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zheng Liu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Yusong R. Guo
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - André Veillette
- Laboratory of Molecular OncologyInstitut de recherches cliniques de Montréal (IRCM)MontréalQuébecH2W1R7Canada
- Department of MedicineUniversity of MontréalMontréalQuébecH3T 1J4Canada
- Department of MedicineMcGill UniversityMontréalQuébecH3G 1Y6Canada
| | - Zhutian Zeng
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Department of OncologyThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefei230001China
| | - Ning Wu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
12
|
Sweet MJ, Ramnath D, Singhal A, Kapetanovic R. Inducible antibacterial responses in macrophages. Nat Rev Immunol 2024:10.1038/s41577-024-01080-y. [PMID: 39294278 DOI: 10.1038/s41577-024-01080-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/20/2024]
Abstract
Macrophages destroy bacteria and other microorganisms through phagocytosis-coupled antimicrobial responses, such as the generation of reactive oxygen species and the delivery of hydrolytic enzymes from lysosomes to the phagosome. However, many intracellular bacteria subvert these responses, escaping to other cellular compartments to survive and/or replicate. Such bacterial subversion strategies are countered by a range of additional direct antibacterial responses that are switched on by pattern-recognition receptors and/or host-derived cytokines and other factors, often through inducible gene expression and/or metabolic reprogramming. Our understanding of these inducible antibacterial defence strategies in macrophages is rapidly evolving. In this Review, we provide an overview of the broad repertoire of antibacterial responses that can be engaged in macrophages, including LC3-associated phagocytosis, metabolic reprogramming and antimicrobial metabolites, lipid droplets, guanylate-binding proteins, antimicrobial peptides, metal ion toxicity, nutrient depletion, autophagy and nitric oxide production. We also highlight key inducers, signalling pathways and transcription factors involved in driving these different antibacterial responses. Finally, we discuss how a detailed understanding of the molecular mechanisms of antibacterial responses in macrophages might be exploited for developing host-directed therapies to combat antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| | - Divya Ramnath
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Amit Singhal
- Infectious Diseases Labs (ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ronan Kapetanovic
- INRAE, Université de Tours, Infectiologie et Santé Publique (ISP), Nouzilly, France
| |
Collapse
|
13
|
Miguel V, Shaw IW, Kramann R. Metabolism at the crossroads of inflammation and fibrosis in chronic kidney disease. Nat Rev Nephrol 2024:10.1038/s41581-024-00889-z. [PMID: 39289568 DOI: 10.1038/s41581-024-00889-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2024] [Indexed: 09/19/2024]
Abstract
Chronic kidney disease (CKD), defined as persistent (>3 months) kidney functional loss, has a growing prevalence (>10% worldwide population) and limited treatment options. Fibrosis driven by the aberrant accumulation of extracellular matrix is the final common pathway of nearly all types of chronic repetitive injury in the kidney and is considered a hallmark of CKD. Myofibroblasts are key extracellular matrix-producing cells that are activated by crosstalk between damaged tubules and immune cells. Emerging evidence indicates that metabolic alterations are crucial contributors to the pathogenesis of kidney fibrosis by affecting cellular bioenergetics and metabolite signalling. Immune cell functions are intricately connected to their metabolic characteristics, and kidney cells seem to undergo cell-type-specific metabolic shifts in response to damage, all of which can determine injury and repair responses in CKD. A detailed understanding of the heterogeneity in metabolic reprogramming of different kidney cellular subsets is essential to elucidating communication processes between cell types and to enabling the development of metabolism-based innovative therapeutic strategies against CKD.
Collapse
Affiliation(s)
- Verónica Miguel
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Isaac W Shaw
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Rafael Kramann
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany.
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Demangel C, Surace L. Host-pathogen interactions from a metabolic perspective: methods of investigation. Microbes Infect 2024; 26:105267. [PMID: 38007087 DOI: 10.1016/j.micinf.2023.105267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/21/2023] [Accepted: 11/21/2023] [Indexed: 11/27/2023]
Abstract
Metabolism shapes immune homeostasis in health and disease. This review presents the range of methods that are currently available to investigate the dialog between metabolism and immunity at the systemic, tissue and cellular levels, particularly during infection.
Collapse
Affiliation(s)
- Caroline Demangel
- Institut Pasteur, Université Paris Cité, Inserm U1224, Immunobiology and Therapy Unit, Paris, France
| | - Laura Surace
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
15
|
Zheng Z, Ling X, Li Y, Qiao S, Zhang S, Wu J, Ma Z, Li M, Guo X, Li Z, Feng Y, Liu X, Goodfellow IG, Zheng H, Xiao S. Host cells reprogram lipid droplet synthesis through YY1 to resist PRRSV infection. mBio 2024; 15:e0154924. [PMID: 38953350 PMCID: PMC11323570 DOI: 10.1128/mbio.01549-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
Metabolism in host cells can be modulated after viral infection, favoring viral survival or clearance. Here, we report that lipid droplet (LD) synthesis in host cells can be modulated by yin yang 1 (YY1) after porcine reproductive and respiratory syndrome virus (PRRSV) infection, resulting in active antiviral activity. As a ubiquitously distributed transcription factor, there was increased expression of YY1 upon PRRSV infection both in vitro and in vivo. YY1 silencing promoted the replication of PRRSV, whereas YY1 overexpression inhibited PRRSV replication. PRRSV infection led to a marked increase in LDs, while YY1 knockout inhibited LD synthesis, and YY1 overexpression enhanced LD accumulation, indicating that YY1 reprograms PRRSV infection-induced intracellular LD synthesis. We also showed that the viral components do not colocalize with LDs during PRRSV infection, and the effect of exogenously induced LD synthesis on PRRSV replication is nearly lethal. Moreover, we demonstrated that YY1 affects the synthesis of LDs by regulating the expression of lipid metabolism genes. YY1 negatively regulates the expression of fatty acid synthase (FASN) to weaken the fatty acid synthesis pathway and positively regulates the expression of peroxisome proliferator-activated receptor gamma (PPARγ) to promote the synthesis of LDs, thus inhibiting PRRSV replication. These novel findings indicate that YY1 plays a crucial role in regulating PRRSV replication by reprogramming LD synthesis. Therefore, our study provides a novel mechanism of host resistance to PRRSV and suggests potential new antiviral strategies against PRRSV infection.IMPORTANCEPorcine reproductive and respiratory virus (PRRSV) has caused incalculable economic damage to the global pig industry since it was first discovered in the 1980s. However, conventional vaccines do not provide satisfactory protection. It is well known that viruses are parasitic pathogens, and the completion of their replication life cycle is highly dependent on host cells. A better understanding of host resistance to PRRSV infection is essential for developing safe and effective strategies to control PRRSV. Here, we report a crucial host antiviral molecule, yin yang 1 (YY1), which is induced to be expressed upon PRRSV infection and subsequently inhibits virus replication by reprogramming lipid droplet (LD) synthesis through transcriptional regulation. Our work provides a novel antiviral mechanism against PRRSV infection and suggests that targeting YY1 could be a new strategy for controlling PRRSV.
Collapse
Affiliation(s)
- Zifang Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xue Ling
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shuang Qiao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Shuangquan Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jie Wu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhiqian Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Mingyu Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xuyang Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhiwei Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yingtong Feng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiao Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ian G. Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shuqi Xiao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
16
|
Zimmermann JA, Lucht K, Stecher M, Badhan C, Glaser KM, Epple MW, Koch LR, Deboutte W, Manke T, Ebnet K, Brinkmann F, Fehler O, Vogl T, Schuster EM, Bremser A, Buescher JM, Rambold AS. Functional multi-organelle units control inflammatory lipid metabolism of macrophages. Nat Cell Biol 2024; 26:1261-1273. [PMID: 38969763 PMCID: PMC11321999 DOI: 10.1038/s41556-024-01457-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 06/05/2024] [Indexed: 07/07/2024]
Abstract
Eukaryotic cells contain several membrane-separated organelles to compartmentalize distinct metabolic reactions. However, it has remained unclear how these organelle systems are coordinated when cells adapt metabolic pathways to support their development, survival or effector functions. Here we present OrgaPlexing, a multi-spectral organelle imaging approach for the comprehensive mapping of six key metabolic organelles and their interactions. We use this analysis on macrophages, immune cells that undergo rapid metabolic switches upon sensing bacterial and inflammatory stimuli. Our results identify lipid droplets (LDs) as primary inflammatory responder organelle, which forms three- and four-way interactions with other organelles. While clusters with endoplasmic reticulum (ER) and mitochondria (mitochondria-ER-LD unit) help supply fatty acids for LD growth, the additional recruitment of peroxisomes (mitochondria-ER-peroxisome-LD unit) supports fatty acid efflux from LDs. Interference with individual components of these units has direct functional consequences for inflammatory lipid mediator synthesis. Together, we show that macrophages form functional multi-organellar units to support metabolic adaptation and provide an experimental strategy to identify organelle-metabolic signalling hubs.
Collapse
Affiliation(s)
- Julia A Zimmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Center of Chronic Immunodeficiency, Medical Center University of Freiburg, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Kerstin Lucht
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Manuel Stecher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Chahat Badhan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katharina M Glaser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Maximilian W Epple
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Lena R Koch
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Ward Deboutte
- Bioinformatics Core Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Thomas Manke
- Bioinformatics Core Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Munster, Munster, Germany
| | - Frauke Brinkmann
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Munster, Munster, Germany
| | - Olesja Fehler
- Institute of Immunology, University of Munster, Munster, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Munster, Munster, Germany
| | - Ev-Marie Schuster
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Anna Bremser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Joerg M Buescher
- Metabolomics Core Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Angelika S Rambold
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
- Center of Chronic Immunodeficiency, Medical Center University of Freiburg, Freiburg, Germany.
| |
Collapse
|
17
|
Zhang X, Ding G, Yang X, Lu H, Xu Y, Hu Y, Liu S, Zhang H, Huang K, Deng G, Ye T, Yu Q, Cai Y, Xie S, Wang W, Chen X. Myriocin enhances the clearance of M. tuberculosis by macrophages through the activation of PLIN2. mSphere 2024; 9:e0025724. [PMID: 38920406 PMCID: PMC11288015 DOI: 10.1128/msphere.00257-24] [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: 03/27/2024] [Accepted: 05/19/2024] [Indexed: 06/27/2024] Open
Abstract
Myriocin is an inhibitor of de novo synthesis of sphingolipids and ceramides. In this research, we showed myriocin could significantly reduce Mtb burden and histopathological inflammation in mice. However, the underlying mechanism remains unclear. RNA-seq analysis revealed a significant increase in gene expression of PLIN2/CD36/CERT1 after myriocin treatment. The reduced bactericidal burden was only reversed after silencing the lipid droplets (LDs) surface protein PLIN2. This suggests that myriocin enhances the ability of macrophages to clear Mtb depending on the PLIN2 gene, which is part of the PPARγ pathway. Indeed, we observed a significant increase in the number of LDs following myriocin treatment.IMPORTANCEMycobacterium tuberculosis has the ability to reprogram host cell lipid metabolism and alter the antimicrobial functions of infected macrophages. The sphingolipids, such as ceramides, are the primary host lipids utilized by the bacteria, making the sphingomyelinase/ceramide system critical in Mtb infections. Surprisingly, the antimicrobial effect of myriocin was found to be independent of its role in reducing ceramides, but instead, it depends on the lipid droplets surface protein PLIN2. Our findings provide a novel mechanism for how myriocin enhances Mtb clearance in macrophages.
Collapse
Affiliation(s)
- Ximeng Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Guanggui Ding
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xirui Yang
- Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Hailin Lu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yuzhong Xu
- Department of Clinical Laboratory, Shenzhen Baoan Hospital, Shenzhen, China
| | - Yunlong Hu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Song Liu
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Huihua Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Kaisong Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Guofang Deng
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Taosheng Ye
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Qing Yu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Shuixiang Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Wenfei Wang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
18
|
Du P, Liu B, Wang X, Zheng Z, Liu S, Guan S, Hou Z. Carex meyeriana Kunth Extract Is a Novel Natural Drug against Candida albicans. Int J Mol Sci 2024; 25:7288. [PMID: 39000395 PMCID: PMC11242224 DOI: 10.3390/ijms25137288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
As a widely distributed plant in Northeast China, Carex meyeriana Kunth (CMK) is generally considered to have antibacterial properties; however, there is a lack of scientific evidence for this. Therefore, we investigated the chemical composition of CMK extract and its effect against C. albicans. A total of 105 compounds were identified in the alcohol extracts of CMK by UPLC-Q-TOF-MS. Most were flavonoids, with Luteolin being the most represented. Among them, 19 compounds are found in the C. albicans lysates. After treatment with CMK ethanol extract, a significant reduction in the number of C. albicans colonies was observed in a vaginal douche solution from day 5 (p < 0.05). Furthermore, the CMK extract can reduce the number of C. albicans spores. The levels of IL-4, IL-6, IL-10, IL-1β, and TNF-α in vaginal tissues all exhibited a significant decrease (p < 0.05) compared to those in the model group as determined by ELISA. The results of HE staining showed that CMK extract can eliminate vaginal mucosa inflammation. CMK adjusts the vaginal mucosa cells by targeting twenty-six different metabolites and five specific metabolic pathways in order to effectively eliminate inflammation. Simultaneously, the CMK regulates twenty-three types of metabolites and six metabolic pathways against C. albicans infection. So, CMK strongly inhibits the growth of C. albicans and significantly reduces vaginal inflammation, making it a promising candidate for treating C. albicans infection.
Collapse
Affiliation(s)
- Panpan Du
- School of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Bingyan Liu
- School of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Xueting Wang
- School of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Zhong Zheng
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Shu Liu
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Songlei Guan
- School of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Zong Hou
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
19
|
Nakamura A, Yoshimura T, Ichikawa T, Okuyama K. Prognostic significance of low hepatic fat content in advanced chronic liver disease: MRI-PDFF insights. Ann Hepatol 2024; 29:101507. [PMID: 38723748 DOI: 10.1016/j.aohep.2024.101507] [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: 11/08/2023] [Revised: 03/19/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION AND OBJECTIVES The mechanisms of hepatic fat loss in late-stage metabolic dysfunction-associated fatty liver disease (MASLD) are enigmatic and the prognostic significance of low hepatic fat content (LHF) in chronic liver disease (CLD) is unknown. Proton density fat fraction (PDFF), measured by magnetic resonance imaging (MRI), is considered the most accurate noninvasive method for quantifying hepatic fat content. This study aimed to address these issues by evaluating PDFF. PATIENTS AND METHODS This is a single-center, retrospective study involving 762 patients with CLD, measuring liver stiffness (LS) using MR elastography and PDFF using MRI. LHF was defined as a PDFF ≤ 2.7 % and hepatic reserve function was assessed using the albumin-bilirubin (ALBI) score. Multivariate analysis explored associations between variables. RESULTS LHF was 27 % in the entire cohort, and PDFF was significantly decreased with LS ≥ 5.5 kPa (p < 0.05). On the multivariate analysis, low body mass index and ALBI score were independently associated with LHF (p < 0.05). In advanced CLD (n = 288), ALBI score and PDFF showed a significant negative correlation regardless of etiology (MASLD/non-MASLD: r= -0.613/-0.233), and the prevalence of LHF increased with progression of ALBI grade (p < 0.01 each). In addition, lower PDFF was associated with increased liver-related and all-cause mortality (p < 0.01), and Cox proportional hazards models extracted LHF as an independent prognostic factor, along with ALBI score and hepatocellular carcinoma (p < 0.05 each). CONCLUSIONS In ACLD, hepatic reserve dysfunction contributed to hepatic fat loss independent of nutritional status, suggesting that LHF may be a poor prognostic factor in all etiologies.
Collapse
Affiliation(s)
- Atsushi Nakamura
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan.
| | - Tsubasa Yoshimura
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Takeshi Ichikawa
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Keiji Okuyama
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| |
Collapse
|
20
|
Zhou W, Tao Y, Qiao Q, Xu N, Li J, Wang G, Fang X, Chen J, Liu W, Xu Z. Cell-Impermeable Buffering Fluorogenic Probes for Live-Cell Super-Resolution Imaging of Plasma Membrane Morphology Dynamics. ACS Sens 2024; 9:3170-3177. [PMID: 38859630 DOI: 10.1021/acssensors.4c00486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Super-resolution fluorescence imaging has emerged as a potent tool for investigating the nanoscale structure and function of the plasma membrane (PM). Nevertheless, the challenge persists in achieving super-resolution imaging of PM dynamics due to limitations in probe photostability and issues with cell internalization staining. Herein, we report assembly-mediated buffering fluorogenic probes BMP-14 and BMP-16 exhibiting fast PM labeling and extended retention time (over 2 h) on PM. The incorporation of alkyl chains proves effective in promoting the aggregation of BMP-14 and BMP-16 into nonfluorescent nanoparticles to realize fluorogenicity and regulate the buffering capacity to rapidly replace photobleached probes ensuring stable long-term super-resolution imaging of PM. Utilizing these PM-buffering probes, we observed dynamic movements of PM filopodia and continuous shrinkage, leading to the formation of extracellular vesicles (EVs) using structured illumination microscopy (SIM). Furthermore, we discovered two distinct modes of EV fusion: one involving fusion through adjacent lipids and the other through filamentous lipid traction. The entire process of EV fusion outside the PM was dynamically tracked. Additionally, BMP-16 exhibited a unique capability of inducing single-molecule fluorescence blinking when used for cell membrane staining. This property makes BMP-16 suitable for the PAINT imaging of cell membranes.
Collapse
Affiliation(s)
- Wei Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Yi Tao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qinglong Qiao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Ning Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Jin Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Guangying Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Xiangning Fang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Wenjuan Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Zhaochao Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
21
|
Marissen J, Reichert L, Härtel C, Fortmann MI, Faust K, Msanga D, Harder J, Zemlin M, Gomez de Agüero M, Masjosthusmann K, Humberg A. Antimicrobial Peptides (AMPs) and the Microbiome in Preterm Infants: Consequences and Opportunities for Future Therapeutics. Int J Mol Sci 2024; 25:6684. [PMID: 38928389 PMCID: PMC11203687 DOI: 10.3390/ijms25126684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in various organisms, including humans. Beyond their direct antimicrobial effects, AMPs play essential roles in various physiological processes. They induce angiogenesis, promote wound healing, modulate immune responses, and serve as chemoattractants for immune cells. AMPs regulate the microbiome and combat microbial infections on the skin, lungs, and gastrointestinal tract. Produced in response to microbial signals, AMPs help maintain a balanced microbial community and provide a first line of defense against infection. In preterm infants, alterations in microbiome composition have been linked to various health outcomes, including sepsis, necrotizing enterocolitis, atopic dermatitis, and respiratory infections. Dysbiosis, or an imbalance in the microbiome, can alter AMP profiles and potentially lead to inflammation-mediated diseases such as chronic lung disease and obesity. In the following review, we summarize what is known about the vital role of AMPs as multifunctional peptides in protecting newborn infants against infections and modulating the microbiome and immune response. Understanding their roles in preterm infants and high-risk populations offers the potential for innovative approaches to disease prevention and treatment.
Collapse
Affiliation(s)
- Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Lilith Reichert
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Kirstin Faust
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Delfina Msanga
- Department of Pediatrics, Bugando Hospital, Catholic University of Health and Allied Sciences, Mwanza 33109, Tanzania;
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, 24105 Kiel, Germany;
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| | - Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| |
Collapse
|
22
|
Li H, Liu Y, Wang Y, Du H, Zhang G, Zhang C, Shuang S, Dong C. A specific dual-locked fluorescence probe to visualize the dynamic changes of lipid droplets and hypochlorous acid in inflammation. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 314:124182. [PMID: 38522376 DOI: 10.1016/j.saa.2024.124182] [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: 01/20/2024] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Inflammation is a key factor leading to the occurrence and development of many diseases, both lipid droplets (LDs) and hypochlorous acid (HClO/ClO-) are regarded as the important biomarkers of inflammation. Therefore, it is of great significance to develop an efficient single chemical sensor that can simultaneously detect these two biomarkers. To achieve the goal, we developed a dual-locked fluorescence probe (TPA-DNP) by fusing two targets activated reporting system, its implementation was achieved by turning-on the fluorescence of TPA-DNP through LDs and HClO/ClO- simultaneously. In simulated LDs environment, TPA-DNP displayed excellent selectivity to HClO/ClO-, high sensitivity (LOD = 0.527 μM) and strong anti-interference ability. In addition, cell and zebrafish imaging experiments showed that TPA-DNP could be utilized to visualize exogenous/endogenous HClO/ClO- in LDs environment, and could also be used to observe the impact of LDs changes on the HClO/ClO- detection. On the basis, TPA-DNP served as a favorable tool to achieve visualization of inflammatory dynamic changes.
Collapse
Affiliation(s)
- Haoyang Li
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Ying Liu
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Yuhang Wang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Huizhi Du
- Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Guomei Zhang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Caihong Zhang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Shaomin Shuang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Chuan Dong
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
23
|
Li C, Wang J, Xu JF, Pi J, Zheng B. Roles of HIF-1α signaling in Mycobacterium tuberculosis infection: New targets for anti-TB therapeutics? Biochem Biophys Res Commun 2024; 711:149920. [PMID: 38615574 DOI: 10.1016/j.bbrc.2024.149920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Tuberculosis (TB), a deadly infectious disease induced by Mycobacterium tuberculosis (Mtb), continues to be a global public health issue that kill millions of patents every year. Despite significant efforts have been paid to identify effective TB treatments, the emergence of drug-resistant strains of the disease and the presence of comorbidities in TB patients urges us to explore the detailed mechanisms involved in TB immunity and develop more effective innovative anti-TB strategies. HIF-1α, a protein involved in regulating cellular immune responses during TB infection, has been highlighted as a promising target for the development of novel strategies for TB treatment due to its critical roles in anti-TB host immunity. This review provides a summary of current research progress on the roles of HIF-1α in TB infection, highlighting its importance in regulating the host immune response upon Mtb infection and summarizing the influences and mechanisms of HIF-1α on anti-TB immunological responses of host cells. This review also discusses the various challenges associated with developing HIF-1α as a target for anti-TB therapies, including ensuring specificity and avoiding off-target effects on normal cell function, determining the regulation and expression of HIF-1α in TB patients, and developing drugs that can inhibit HIF-1α. More deep understanding of the molecular mechanisms involved in HIF-1α signaling, its impact on TB host status, and systematic animal testing and clinical trials may benefit the optimization of HIF-1α as a novel therapeutic target for TB.
Collapse
Affiliation(s)
- Chaowei Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jiajun Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Biying Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| |
Collapse
|
24
|
Su Y, Qu Q, Li J, Han Z, Fang Y, Flavorta BL, Jia Z, Yu Q, Zhang Y, Qian P, Tang X. Perilipin1 inhibits Nosema bombycis proliferation by promoting Domeless- and Hop-mediated JAK-STAT pathway activation in Bombyx mori. Microbiol Spectr 2024; 12:e0367123. [PMID: 38690912 PMCID: PMC11237581 DOI: 10.1128/spectrum.03671-23] [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: 10/16/2023] [Accepted: 03/22/2024] [Indexed: 05/03/2024] Open
Abstract
Lipid droplets (LDs) are dynamic organelles that participate in the regulation of lipid metabolism and cellular homeostasis inside of cells. LD-associated proteins, also known as perilipins (PLINs), are a family of proteins found on the surface of LDs that regulate lipid metabolism, immunity, and other functions. In silkworms, pébrine disease caused by infection by the microsporidian Nosema bombycis (Nb) is a severe threat to the sericultural industry. Although we found that Nb relies on lipids from silkworms to facilitate its proliferation, the relationship between PLINs and Nb proliferation remains unknown. Here, we found Nb infection caused the accumulation of LDs in the fat bodies of silkworm larvae. The characterized perilipin1 gene (plin1) promotes the accumulation of intracellular LDs and is involved in Nb proliferation. plin1 is similar to perilipin1 in humans and is conserved in all insects. The expression of plin1 was mostly enriched in the fat body rather than in other tissues. Knockdown of plin1 enhanced Nb proliferation, whereas overexpression of plin1 inhibited its proliferation. Furthermore, we confirmed that plin1 increased the expression of the Domeless and Hop in the JAK-STAT immune pathway and inhibited Nb proliferation. Taken together, our current findings demonstrate that plin1 inhibits Nb proliferation by promoting the JAK-STAT pathway through increased expression of Domeless and Hop. This study provides new insights into the complicated connections among microsporidia pathogens, LD surface proteins, and insect immunity.IMPORTANCELipid droplets (LDs) are lipid storage sites in cells and are present in almost all animals. Many studies have found that LDs may play a role in host resistance to pathogens and are closely related to innate immunity. The present study found that a surface protein of insect lipid droplets could not only regulate the morphological changes of lipid droplets but also inhibit the proliferation of a microsporidian pathogen Nosema bombycis (Nb) by activating the JAK-STAT signaling pathway. This is the first discovery of the relationship between microsporidian pathogen and insect lipid surface protein perilipin and insect immunity.
Collapse
Affiliation(s)
- Yaping Su
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Qingsheng Qu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Junling Li
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Zhenghao Han
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Yujia Fang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Billong Laura Flavorta
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Zhenwei Jia
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Qiong Yu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Yiling Zhang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Ping Qian
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Xudong Tang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| |
Collapse
|
25
|
Kim EJ, Jeon HB, Kang MJ, Lee J. Dynamic Imaging of Lipid Droplets in Cells and Tissues by Using Dioxaborine Barbiturate-Based Fluorogenic Probes. Anal Chem 2024; 96:8356-8364. [PMID: 38753674 DOI: 10.1021/acs.analchem.3c05368] [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: 05/18/2024]
Abstract
Lipids are essential for various cellular functions, including energy storage, membrane flexibility, and signaling molecule production. Maintaining proper lipid levels is important to prevent health problems such as cancer, neurodegenerative disorders, cardiovascular diseases, obesity, and diabetes. Monitoring cellular lipid droplets (LDs) in real-time with high resolution can provide insights into LD-related pathways and diseases owing to the dynamic nature of LDs. Fluorescence-based imaging is widely used for tracking LDs in live cells and animal models. However, the current fluorophores have limitations such as poor photostability and high background staining. Herein, we developed a novel fluorogenic probe based on a push-pull interaction combined with aggregation-induced emission enhancement (AIEE) for dynamic imaging of LDs. Probe 1 exhibits favorable membrane permeability and spectroscopic characteristics, allowing specific imaging of cellular LDs and time-lapse imaging of LD accumulation. This probe can also be used to examine LDs in fruit fly tissues in various metabolic states, serving as a highly versatile and specific tool for dynamic LD imaging in cellular and tissue environments.
Collapse
Affiliation(s)
- Eun-Ji Kim
- Department of Next-Generation Applied Science and School of Biopharmaceutical and Medical Sciences, Sungshin University, Seoul 01133, Republic of Korea
| | - Hye-Bin Jeon
- Department of Next-Generation Applied Science and School of Biopharmaceutical and Medical Sciences, Sungshin University, Seoul 01133, Republic of Korea
| | - Min-Ji Kang
- Department of Pharmacology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Jiyoun Lee
- Department of Next-Generation Applied Science and School of Biopharmaceutical and Medical Sciences, Sungshin University, Seoul 01133, Republic of Korea
| |
Collapse
|
26
|
Macalester W, Boussahel A, Moreno-Tortolero RO, Shannon MR, West N, Hill D, Perriman A. A 3D In-vitro model of the human dentine interface shows long-range osteoinduction from the dentine surface. Int J Oral Sci 2024; 16:37. [PMID: 38734663 PMCID: PMC11088668 DOI: 10.1038/s41368-024-00298-9] [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: 10/12/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 05/13/2024] Open
Abstract
Emerging regenerative cell therapies for alveolar bone loss have begun to explore the use of cell laden hydrogels for minimally invasive surgery to treat small and spatially complex maxilla-oral defects. However, the oral cavity presents a unique and challenging environment for in vivo bone tissue engineering, exhibiting both hard and soft periodontal tissue as well as acting as key biocenosis for many distinct microbial communities that interact with both the external environment and internal body systems, which will impact on cell fate and subsequent treatment efficacy. Herein, we design and bioprint a facile 3D in vitro model of a human dentine interface to probe the effect of the dentine surface on human mesenchymal stem cells (hMSCs) encapsulated in a microporous hydrogel bioink. We demonstrate that the dentine substrate induces osteogenic differentiation of encapsulated hMSCs, and that both dentine and β-tricalcium phosphate substrates stimulate extracellular matrix production and maturation at the gel-media interface, which is distal to the gel-substrate interface. Our findings demonstrate the potential for long-range effects on stem cells by mineralized surfaces during bone tissue engineering and provide a framework for the rapid development of 3D dentine-bone interface models.
Collapse
Affiliation(s)
- William Macalester
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom
- Bristol Centre for Functional Nanomaterials, HH Wills Physics Laboratory, University of Bristol, Tyndall Avenue, Bristol, United Kingdom
| | - Asme Boussahel
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom.
| | - Rafael O Moreno-Tortolero
- Bristol Centre for Functional Nanomaterials, HH Wills Physics Laboratory, University of Bristol, Tyndall Avenue, Bristol, United Kingdom
- Centre for Protolife Research, School of Chemistry, University of Bristol, Cantocks Close, Bristol, United Kingdom
- Max Planck-Bristol Centre for Minimal Biology, School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Mark R Shannon
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom
| | - Nicola West
- Periodontology, Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol, United Kingdom
| | - Darryl Hill
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom
| | - Adam Perriman
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom.
| |
Collapse
|
27
|
Safi R, Menéndez P, Pol A. Lipid droplets provide metabolic flexibility for cancer progression. FEBS Lett 2024; 598:1301-1327. [PMID: 38325881 DOI: 10.1002/1873-3468.14820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
A hallmark of cancer cells is their remarkable ability to efficiently adapt to favorable and hostile environments. Due to a unique metabolic flexibility, tumor cells can grow even in the absence of extracellular nutrients or in stressful scenarios. To achieve this, cancer cells need large amounts of lipids to build membranes, synthesize lipid-derived molecules, and generate metabolic energy in the absence of other nutrients. Tumor cells potentiate strategies to obtain lipids from other cells, metabolic pathways to synthesize new lipids, and mechanisms for efficient storage, mobilization, and utilization of these lipids. Lipid droplets (LDs) are the organelles that collect and supply lipids in eukaryotes and it is increasingly recognized that the accumulation of LDs is a new hallmark of cancer cells. Furthermore, an active role of LD proteins in processes underlying tumorigenesis has been proposed. Here, by focusing on three major classes of LD-resident proteins (perilipins, lipases, and acyl-CoA synthetases), we provide an overview of the contribution of LDs to cancer progression and discuss the role of LD proteins during the proliferation, invasion, metastasis, apoptosis, and stemness of cancer cells.
Collapse
Affiliation(s)
- Rémi Safi
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
| |
Collapse
|
28
|
Enkler L, Spang A. Functional interplay of lipid droplets and mitochondria. FEBS Lett 2024; 598:1235-1251. [PMID: 38268392 DOI: 10.1002/1873-3468.14809] [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: 10/12/2023] [Revised: 12/12/2023] [Accepted: 01/04/2024] [Indexed: 01/26/2024]
Abstract
Our body stores energy mostly in form of fatty acids (FAs) in lipid droplets (LDs). From there the FAs can be mobilized and transferred to peroxisomes and mitochondria. This transfer is dependent on close opposition of LDs and mitochondria and peroxisomes and happens at membrane contact sites. However, the composition and the dynamics of these contact sites is not well understood, which is in part due to the dependence on the metabolic state of the cell and on the cell- and tissue-type. Here, we summarize the current knowledge on the contacts between lipid droplets and mitochondria both in mammals and in the yeast Saccharomyces cerevisiae, in which various contact sites are well studied. We discuss possible functions of the contact site and their implication in disease.
Collapse
Affiliation(s)
| | - Anne Spang
- Biozentrum, University of Basel, Switzerland
| |
Collapse
|
29
|
Dudka W, Salo VT, Mahamid J. Zooming into lipid droplet biology through the lens of electron microscopy. FEBS Lett 2024; 598:1127-1142. [PMID: 38726814 DOI: 10.1002/1873-3468.14899] [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: 01/26/2024] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Electron microscopy (EM), in its various flavors, has significantly contributed to our understanding of lipid droplets (LD) as central organelles in cellular metabolism. For example, EM has illuminated that LDs, in contrast to all other cellular organelles, are uniquely enclosed by a single phospholipid monolayer, revealed the architecture of LD contact sites with different organelles, and provided near-atomic resolution maps of key enzymes that regulate neutral lipid biosynthesis and LD biogenesis. In this review, we first provide a brief history of pivotal findings in LD biology unveiled through the lens of an electron microscope. We describe the main EM techniques used in the context of LD research and discuss their current capabilities and limitations, thereby providing a foundation for utilizing suitable EM methodology to address LD-related questions with sufficient level of structural preservation, detail, and resolution. Finally, we highlight examples where EM has recently been and is expected to be instrumental in expanding the frontiers of LD biology.
Collapse
Affiliation(s)
- Wioleta Dudka
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Veijo T Salo
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Julia Mahamid
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Cell Biology and Biophysics Unit, EMBL, Heidelberg, Germany
| |
Collapse
|
30
|
Griseti E, Bello AA, Bieth E, Sabbagh B, Iacovoni JS, Bigay J, Laurell H, Čopič A. Molecular mechanisms of perilipin protein function in lipid droplet metabolism. FEBS Lett 2024; 598:1170-1198. [PMID: 38140813 DOI: 10.1002/1873-3468.14792] [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: 10/21/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Perilipins are abundant lipid droplet (LD) proteins present in all metazoans and also in Amoebozoa and fungi. Humans express five perilipins, which share a similar domain organization: an amino-terminal PAT domain and an 11-mer repeat region, which can fold into amphipathic helices that interact with LDs, followed by a structured carboxy-terminal domain. Variations of this organization that arose during vertebrate evolution allow for functional specialization between perilipins in relation to the metabolic needs of different tissues. We discuss how different features of perilipins influence their interaction with LDs and their cellular targeting. PLIN1 and PLIN5 play a direct role in lipolysis by regulating the recruitment of lipases to LDs and LD interaction with mitochondria. Other perilipins, particularly PLIN2, appear to protect LDs from lipolysis, but the molecular mechanism is not clear. PLIN4 stands out with its long repetitive region, whereas PLIN3 is most widely expressed and is used as a nascent LD marker. Finally, we discuss the genetic variability in perilipins in connection with metabolic disease, prominent for PLIN1 and PLIN4, underlying the importance of understanding the molecular function of perilipins.
Collapse
Affiliation(s)
- Elena Griseti
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Abdoul Akim Bello
- Institut de Pharmacologie Moléculaire et Cellulaire - IPMC, Université Côte d'Azur, CNRS, Valbonne, France
| | - Eric Bieth
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
- Departement de Génétique Médicale, Centre Hospitalier Universitaire de Toulouse, France
| | - Bayane Sabbagh
- Centre de Recherche en Biologie Cellulaire de Montpellier - CRBM, Université de Montpellier, CNRS, France
| | - Jason S Iacovoni
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Joëlle Bigay
- Institut de Pharmacologie Moléculaire et Cellulaire - IPMC, Université Côte d'Azur, CNRS, Valbonne, France
| | - Henrik Laurell
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Alenka Čopič
- Centre de Recherche en Biologie Cellulaire de Montpellier - CRBM, Université de Montpellier, CNRS, France
| |
Collapse
|
31
|
Morito D. Molecular structure and function of mysterin/RNF213. J Biochem 2024; 175:495-505. [PMID: 38378744 DOI: 10.1093/jb/mvae020] [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: 12/05/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
Mysterin is a large intracellular protein harboring a RING finger ubiquitin ligase domain and is also referred to as RING finger protein 213 (RNF213). The author performed the first molecular cloning of the mysterin gene as the final step in genetic exploration of cerebrovascular moyamoya disease (MMD) and initiated the next round of exploration to understand its molecular and cellular functions. Although much remains unknown, accumulating findings suggest that mysterin functions in cells by targeting massive intracellular structures, such as lipid droplets (LDs) and various invasive pathogens. In the latter case, mysterin appears to directly surround and ubiquitylate the surface of pathogens and stimulate cell-autonomous antimicrobial reactions, such as xenophagy and inflammatory response. To date, multiple mutations causing MMD have been identified within and near the RING finger domain of mysterin; however, their functional relevance remains largely unknown. Besides the RING finger, mysterin harbors a dynein-like ATPase core and an RZ finger, another ubiquitin ligase domain unique to mysterin, while functional exploration of these domains has also just commenced. In this review, the author attempts to summarize the core findings regarding the molecular structure and function of the mysterin protein, with an emphasis on the perspective of MMD research.
Collapse
Affiliation(s)
- Daisuke Morito
- Department of Biochemistry, Showa University School of Medicine, Hatanodai 1-5-8, Shinagawa, Tokyo 142-0064, Japan
| |
Collapse
|
32
|
Haney MS, Pálovics R, Munson CN, Long C, Johansson PK, Yip O, Dong W, Rawat E, West E, Schlachetzki JCM, Tsai A, Guldner IH, Lamichhane BS, Smith A, Schaum N, Calcuttawala K, Shin A, Wang YH, Wang C, Koutsodendris N, Serrano GE, Beach TG, Reiman EM, Glass CK, Abu-Remaileh M, Enejder A, Huang Y, Wyss-Coray T. APOE4/4 is linked to damaging lipid droplets in Alzheimer's disease microglia. Nature 2024; 628:154-161. [PMID: 38480892 PMCID: PMC10990924 DOI: 10.1038/s41586-024-07185-7] [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: 01/02/2022] [Accepted: 02/09/2024] [Indexed: 03/26/2024]
Abstract
Several genetic risk factors for Alzheimer's disease implicate genes involved in lipid metabolism and many of these lipid genes are highly expressed in glial cells1. However, the relationship between lipid metabolism in glia and Alzheimer's disease pathology remains poorly understood. Through single-nucleus RNA sequencing of brain tissue in Alzheimer's disease, we have identified a microglial state defined by the expression of the lipid droplet-associated enzyme ACSL1 with ACSL1-positive microglia being most abundant in patients with Alzheimer's disease having the APOE4/4 genotype. In human induced pluripotent stem cell-derived microglia, fibrillar Aβ induces ACSL1 expression, triglyceride synthesis and lipid droplet accumulation in an APOE-dependent manner. Additionally, conditioned media from lipid droplet-containing microglia lead to Tau phosphorylation and neurotoxicity in an APOE-dependent manner. Our findings suggest a link between genetic risk factors for Alzheimer's disease with microglial lipid droplet accumulation and neurotoxic microglia-derived factors, potentially providing therapeutic strategies for Alzheimer's disease.
Collapse
Affiliation(s)
- Michael S Haney
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Róbert Pálovics
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Christy Nicole Munson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Chris Long
- Department of Materials Science & Engineering Department, Stanford University School of Medicine, Stanford, CA, USA
| | - Patrik K Johansson
- Department of Materials Science & Engineering Department, Stanford University School of Medicine, Stanford, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Wentao Dong
- The Institute for Chemistry, Engineering & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Eshaan Rawat
- The Institute for Chemistry, Engineering & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Elizabeth West
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA
| | - Andy Tsai
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Ian Hunter Guldner
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Bhawika S Lamichhane
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Amanda Smith
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Nicholas Schaum
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Kruti Calcuttawala
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Andrew Shin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Yung-Hua Wang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Chengzhong Wang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Development and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
| | - Geidy E Serrano
- Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas G Beach
- Banner Alzheimer's Institute and Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute and Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Christopher K Glass
- Development and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
| | - Monther Abu-Remaileh
- The Institute for Chemistry, Engineering & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Annika Enejder
- Department of Materials Science & Engineering Department, Stanford University School of Medicine, Stanford, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Development and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA.
| |
Collapse
|
33
|
Cabodevilla AG, Son N, Goldberg IJ. Intracellular lipase and regulation of the lipid droplet. Curr Opin Lipidol 2024; 35:85-92. [PMID: 38447014 PMCID: PMC10919935 DOI: 10.1097/mol.0000000000000918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
PURPOSE OF REVIEW Lipid droplets are increasingly recognized as distinct intracellular organelles that have functions exclusive to the storage of energetic lipids. Lipid droplets modulate macrophage inflammatory phenotype, control the availability of energy for muscle function, store excess lipid, sequester toxic lipids, modulate mitochondrial activity, and allow transfer of fatty acids between tissues. RECENT FINDINGS There have been several major advances in our understanding of the formation, dissolution, and function of this organelle during the past two years. These include new information on movement and partition of amphipathic proteins between the cytosol and lipid droplet surface, molecular determinants of lipid droplet formation, and pathways leading to lipid droplet hydrophobic lipid formation. Rapid advances in mitochondrial biology have also begun to define differences in their function and partnering with lipid droplets to modulate lipid storage versus oxidation. SUMMARY This relationship of lipid droplets biology and cellular function provides new understanding of an important cellular organelle that influences muscle function, adipose lipid storage, and diseases of lipotoxicity.
Collapse
Affiliation(s)
- Ainara G Cabodevilla
- Division of Endocrinology, New York University Grossman School of Medicine, New York, New York, USA
| | | | | |
Collapse
|
34
|
Windham IA, Cohen S. The cell biology of APOE in the brain. Trends Cell Biol 2024; 34:338-348. [PMID: 37805344 PMCID: PMC10995109 DOI: 10.1016/j.tcb.2023.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/23/2023] [Accepted: 09/08/2023] [Indexed: 10/09/2023]
Abstract
Apolipoprotein E (APOE) traffics lipids in the central nervous system. The E4 variant of APOE is a major genetic risk factor for Alzheimer's disease (AD) and a multitude of other neurodegenerative diseases, yet the molecular mechanisms by which APOE4 drives disease are still unclear. A growing collection of studies in iPSC models, knock-in mice, and human postmortem brain tissue have demonstrated that APOE4 expression in astrocytes and microglia is associated with the accumulation of cytoplasmic lipid droplets, defects in endolysosomal trafficking, impaired mitochondrial metabolism, upregulation of innate immune pathways, and a transition into a reactive state. In this review, we collate these developments and suggest testable mechanistic hypotheses that could explain common APOE4 phenotypes.
Collapse
Affiliation(s)
- Ian A Windham
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC, USA
| | - Sarah Cohen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
35
|
Windham IA, Powers AE, Ragusa JV, Wallace ED, Zanellati MC, Williams VH, Wagner CH, White KK, Cohen S. APOE traffics to astrocyte lipid droplets and modulates triglyceride saturation and droplet size. J Cell Biol 2024; 223:e202305003. [PMID: 38334983 PMCID: PMC10857907 DOI: 10.1083/jcb.202305003] [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: 05/02/2023] [Revised: 12/01/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024] Open
Abstract
The E4 variant of APOE strongly predisposes individuals to late-onset Alzheimer's disease. We demonstrate that in response to lipogenesis, apolipoprotein E (APOE) in astrocytes can avoid translocation into the endoplasmic reticulum (ER) lumen and traffic to lipid droplets (LDs) via membrane bridges at ER-LD contacts. APOE knockdown promotes fewer, larger LDs after a fatty acid pulse, which contain more unsaturated triglyceride after fatty acid pulse-chase. This LD size phenotype was rescued by chimeric APOE that targets only LDs. Like APOE depletion, APOE4-expressing astrocytes form a small number of large LDs enriched in unsaturated triglyceride. Additionally, the LDs in APOE4 cells exhibit impaired turnover and increased sensitivity to lipid peroxidation. Our data indicate that APOE plays a previously unrecognized role as an LD surface protein that regulates LD size and composition. APOE4 causes aberrant LD composition and morphology. Our study contributes to accumulating evidence that APOE4 astrocytes with large, unsaturated LDs are sensitized to lipid peroxidation, which could contribute to Alzheimer's disease risk.
Collapse
Affiliation(s)
- Ian A. Windham
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alex E. Powers
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joey V. Ragusa
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - E. Diane Wallace
- Mass Spectrometry Core Laboratory, Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maria Clara Zanellati
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria H. Williams
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Colby H. Wagner
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristen K. White
- Microscopy Services Laboratory, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah Cohen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
36
|
Ravel-Godreuil C, Roy ER, Puttapaka SN, Li S, Wang Y, Yuan X, Eltzschig HK, Cao W. Transcriptional Responses of Different Brain Cell Types to Oxygen Decline. Brain Sci 2024; 14:341. [PMID: 38671993 PMCID: PMC11048388 DOI: 10.3390/brainsci14040341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Brain hypoxia is associated with a wide range of physiological and clinical conditions. Although oxygen is an essential constituent of maintaining brain functions, our understanding of how specific brain cell types globally respond and adapt to decreasing oxygen conditions is incomplete. In this study, we exposed mouse primary neurons, astrocytes, and microglia to normoxia and two hypoxic conditions and obtained genome-wide transcriptional profiles of the treated cells. Analysis of differentially expressed genes under conditions of reduced oxygen revealed a canonical hypoxic response shared among different brain cell types. In addition, we observed a higher sensitivity of neurons to oxygen decline, and dissected cell type-specific biological processes affected by hypoxia. Importantly, this study establishes novel gene modules associated with brain cells responding to oxygen deprivation and reveals a state of profound stress incurred by hypoxia.
Collapse
Affiliation(s)
- Camille Ravel-Godreuil
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (C.R.-G.); (E.R.R.); (S.N.P.); (S.L.); (Y.W.); (X.Y.); (H.K.E.)
| | - Ethan R. Roy
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (C.R.-G.); (E.R.R.); (S.N.P.); (S.L.); (Y.W.); (X.Y.); (H.K.E.)
| | - Srinivas N. Puttapaka
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (C.R.-G.); (E.R.R.); (S.N.P.); (S.L.); (Y.W.); (X.Y.); (H.K.E.)
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Sanming Li
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (C.R.-G.); (E.R.R.); (S.N.P.); (S.L.); (Y.W.); (X.Y.); (H.K.E.)
| | - Yanyu Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (C.R.-G.); (E.R.R.); (S.N.P.); (S.L.); (Y.W.); (X.Y.); (H.K.E.)
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (C.R.-G.); (E.R.R.); (S.N.P.); (S.L.); (Y.W.); (X.Y.); (H.K.E.)
| | - Holger K. Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (C.R.-G.); (E.R.R.); (S.N.P.); (S.L.); (Y.W.); (X.Y.); (H.K.E.)
| | - Wei Cao
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (C.R.-G.); (E.R.R.); (S.N.P.); (S.L.); (Y.W.); (X.Y.); (H.K.E.)
| |
Collapse
|
37
|
He Y, Liu L, Dong Y, Zhang X, Song Y, Jing Y, Ni Y, Wang Y, Wang Z, Ding L. Lipid droplets-related Perilipin-3: potential immune checkpoint and oncogene in oral squamous cell carcinoma. Cancer Immunol Immunother 2024; 73:78. [PMID: 38554152 PMCID: PMC10981595 DOI: 10.1007/s00262-024-03659-9] [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: 01/09/2024] [Accepted: 02/20/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND Lipid droplets (LDs) as major lipid storage organelles are recently reported to be innate immune hubs. Perilipin-3 (PLIN3) is indispensable for the formation and accumulation of LDs. Since cancer patients show dysregulated lipid metabolism, we aimed to elaborate the role of LDs-related PLIN3 in oral squamous cell carcinoma (OSCC). METHODS PLIN3 expression patterns (n = 87), its immune-related landscape (n = 74) and association with B7-H2 (n = 51) were assessed by immunohistochemistry and flow cytometry. Real-time PCR, Western blot, Oil Red O assay, immunofluorescence, migration assay, spheroid-forming assay and flow cytometry were performed for function analysis. RESULTS Spotted LDs-like PLIN3 staining was dominantly enriched in tumor cells than other cell types. PLIN3high tumor showed high proliferation index with metastasis potential, accompanied with less CD3+CD8+ T cells in peripheral blood and in situ tissue, conferring immunosuppressive microenvironment and shorter postoperative survival. Consistently, PLIN3 knockdown in tumor cells not only reduced LD deposits and tumor migration, but benefited for CD8+ T cells activation in co-culture system with decreased B7-H2. An OSCC subpopulation harbored PLIN3highB7-H2high tumor showed more T cells exhaustion, rendering higher risk of cancer-related death (95% CI 1.285-6.851). CONCLUSIONS LDs marker PLIN3 may be a novel immunotherapeutic target in OSCC.
Collapse
Affiliation(s)
- Yijia He
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lingyun Liu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuexin Dong
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoxin Zhang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuxian Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yue Jing
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yanhong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yi Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Zhiyong Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Liang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
38
|
Nakahara H, Hiranita T, Shibata O. A Sigma 1 Receptor Agonist Alters Fluidity and Stability of Lipid Monolayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:6484-6492. [PMID: 38470245 PMCID: PMC11554242 DOI: 10.1021/acs.langmuir.4c00053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Interactions between the sigma1 receptor agonist PRE-084 and various lipid monolayers, including dipalmitoylphosphatidylcholine (DPPC), DPP-ethanolamine (DPPE), DPP-glycerol (DPPG), DPP-serine (DPPS), palmitoylsphingomyelin (PSM), and cholesterol (Ch), were investigated to elucidate the effects of PRE-084 on membrane fluidity and stability. Their interactions with sigma1 receptor agonists have potential implications for neuroprotection, antidepressant, analgesic, and cognitive enhancement effects. In this study, we observed that the presence of PRE-084 in the subphase led to increased fluidity in DPPC and DPPE monolayers, whereas decreasing fluidity was observed in DPPG, DPPS, and PSM monolayers. The interaction of PRE-084 with Ch monolayers was found to be distinct from its interaction with other lipids. Fluorescence microscopy images revealed changes in the size and shape of liquid-condensed domains in the presence of PRE-084, supporting the notion of altered membrane fluidity. Our findings provide new insights into the interaction of PRE-084 with lipid monolayers and its potential implications for biological and membrane science.
Collapse
Affiliation(s)
- Hiromichi Nakahara
- Department of Industrial Pharmacy, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan
| | - Takato Hiranita
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
- Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Osamu Shibata
- Department of Biophysical Chemistry, Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki 859-3298, Japan
| |
Collapse
|
39
|
徐 思, 魏 洁, 谢 静. [Research Progress of Cellular Lipid Droplets in Oral Diseases]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:475-481. [PMID: 38645850 PMCID: PMC11026902 DOI: 10.12182/20240160102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Indexed: 04/23/2024]
Abstract
Lipid droplets are dynamic multifunctional organelles composed of a neutral lipid core and a phospholipid monolayer membrane modified by a specific set of proteins. PAT family proteins are the most characteristic lipid droplet proteins, playing an important role in regulating lipid droplet structure, function, and metabolism. The biogenesis of lipid droplets involves neutral lipid synthesis and the nucleation, budding, and growth of the lipid droplets. Lipid droplets not only serve as the energy metabolism reserve of cells but also participate in intracellular signal transduction and the development of inflammation and tumor. Lipid droplets are closely connected to and interact with various organelles, regulating the division, the transportation, and the genetics of organelles. The complexity of lipid droplets biogenesis and the diversity of their functions may have provided a physiological basis for the pathogenesis and development of diseases, but further research is needed in order to better understand the relevant processes. Published findings have helped elucidate the association between lipid droplets and diseases, such as obesity, non-alcoholic fatty liver disease, neurodegenerative disease, cancer, and cardiovascular disease, but the relationship between lipid droplets and oral diseases has not been fully studied. Topics that warrant further research include the role and mechanisms of lipid droplets in the pathogenesis and development of oral diseases, the relationship between oral diseases and systemic diseases, and translation of the effect of lipid droplets on oral diseases into valuable clinical diagnostic and treatment methods. Herein, we reviewed the biogenesis and functions of lipid droplets and the progress in research concerning lipid droplets in oral diseases, including mouth neoplasms, periodontitis, and dental caries.
Collapse
Affiliation(s)
- 思群 徐
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 洁雅 魏
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 静 谢
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
40
|
Deng B, Kong W, Shen X, Han C, Zhao Z, Chen S, Zhou C, Bae-Jump V. The role of DGAT1 and DGAT2 in regulating tumor cell growth and their potential clinical implications. J Transl Med 2024; 22:290. [PMID: 38500157 PMCID: PMC10946154 DOI: 10.1186/s12967-024-05084-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: 12/20/2023] [Accepted: 03/10/2024] [Indexed: 03/20/2024] Open
Abstract
Lipid metabolism is widely reprogrammed in tumor cells. Lipid droplet is a common organelle existing in most mammal cells, and its complex and dynamic functions in maintaining redox and metabolic balance, regulating endoplasmic reticulum stress, modulating chemoresistance, and providing essential biomolecules and ATP have been well established in tumor cells. The balance between lipid droplet formation and catabolism is critical to maintaining energy metabolism in tumor cells, while the process of energy metabolism affects various functions essential for tumor growth. The imbalance of synthesis and catabolism of fatty acids in tumor cells leads to the alteration of lipid droplet content in tumor cells. Diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2, the enzymes that catalyze the final step of triglyceride synthesis, participate in the formation of lipid droplets in tumor cells and in the regulation of cell proliferation, migration and invasion, chemoresistance, and prognosis in tumor. Several diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2 inhibitors have been developed over the past decade and have shown anti-tumor effects in preclinical tumor models and improvement of metabolism in clinical trials. In this review, we highlight key features of fatty acid metabolism and different paradigms of diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2 activities on cell proliferation, migration, chemoresistance, and prognosis in tumor, with the hope that these scientific findings will have potential clinical implications.
Collapse
Affiliation(s)
- Boer Deng
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiaochang Shen
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chao Han
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
| | - Ziyi Zhao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shuning Chen
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
41
|
Lu X, Li G, Liu Y, Luo G, Ding S, Zhang T, Li N, Geng Q. The role of fatty acid metabolism in acute lung injury: a special focus on immunometabolism. Cell Mol Life Sci 2024; 81:120. [PMID: 38456906 PMCID: PMC10923746 DOI: 10.1007/s00018-024-05131-4] [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: 11/09/2023] [Revised: 01/06/2024] [Accepted: 01/17/2024] [Indexed: 03/09/2024]
Abstract
Reputable evidence from multiple studies suggests that excessive and uncontrolled inflammation plays an indispensable role in mediating, amplifying, and protracting acute lung injury (ALI). Traditionally, immunity and energy metabolism are regarded as separate functions regulated by distinct mechanisms, but recently, more and more evidence show that immunity and energy metabolism exhibit a strong interaction which has given rise to an emerging field of immunometabolism. Mammalian lungs are organs with active fatty acid metabolism, however, during ALI, inflammation and oxidative stress lead to a series metabolic reprogramming such as impaired fatty acid oxidation, increased expression of proteins involved in fatty acid uptake and transport, enhanced synthesis of fatty acids, and accumulation of lipid droplets. In addition, obesity represents a significant risk factor for ALI/ARDS. Thus, we have further elucidated the mechanisms of obesity exacerbating ALI from the perspective of fatty acid metabolism. To sum up, this paper presents a systematical review of the relationship between extensive fatty acid metabolic pathways and acute lung injury and summarizes recent advances in understanding the involvement of fatty acid metabolism-related pathways in ALI. We hold an optimistic believe that targeting fatty acid metabolism pathway is a promising lung protection strategy, but the specific regulatory mechanisms are way too complex, necessitating further extensive and in-depth investigations in future studies.
Collapse
Affiliation(s)
- Xiao Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guoqing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Tianyu Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
42
|
He D, Yan M, Sun Q, Zhang M, Xia Y, Sun Y, Li Z. Ketocyanine-Based Fluorescent Probe Revealing the Polarity Heterogeneity of Lipid Droplets and Enabling Accurate Diagnosis of Hepatocellular Carcinoma. Adv Healthc Mater 2024; 13:e2303212. [PMID: 38241604 DOI: 10.1002/adhm.202303212] [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: 09/23/2023] [Revised: 01/02/2024] [Indexed: 01/21/2024]
Abstract
Hepatocellular carcinoma (HCC) has gradually become a pronoun for terrifying death owing to its high mortality rate. With the progression of HCC, lipid droplets (LDs) in HCC cells exhibit specific variations such as increased LDs number and decreased polarity, which can serve as the diagnostic target. However, developing an effective method to achieve HCC diagnosis and reveal LDs polarity heterogeneity is still a crucial challenge. Herein, the first high-performance LDs-targeting probe (1) is reported based on ketocyanine strategy with ultrasensitive polarity-responding ability and near-infrared emission. Probe 1 shows excellent sensitivity to polarity parameter Δf (0.027-0.290) with 808-fold fluorescence enhancement and the emission wavelength red-shifts 91 nm. In HCC cells, probe 1 shows a 2.5- to 5.9-fold fluorescence enhancement compared with normal and other cancer cells which exceeds clinical threshold of 2.0, indicating probe 1 can distinguish HCC cells. The LDs polarity heterogeneity is revealed and it displays a sequence, HCC cells < other cancer cells < normal cells, which may provide useful insight to engineer LDs-targeting probes for HCC cell discrimination. Finally, probe 1 realizes accurate HCC diagnosis on the cellular, organ, and in vivo levels, providing a satisfying tool for clinical HCC diagnosis and surgical navigation.
Collapse
Affiliation(s)
- Deming He
- College of Chemistry, Institute of Analytical Chemistry for Life Science, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou, 450001, China
| | - Minmin Yan
- College of Chemistry, Institute of Analytical Chemistry for Life Science, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou, 450001, China
| | - Qiuling Sun
- College of Chemistry, Institute of Analytical Chemistry for Life Science, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou, 450001, China
| | - Mingwei Zhang
- College of Chemistry, Institute of Analytical Chemistry for Life Science, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou, 450001, China
| | - Yu Xia
- College of Chemistry, Institute of Analytical Chemistry for Life Science, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuanqiang Sun
- College of Chemistry, Institute of Analytical Chemistry for Life Science, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhaohui Li
- College of Chemistry, Institute of Analytical Chemistry for Life Science, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
43
|
Tan YJ, Jin Y, Zhou J, Yang YF. Lipid droplets in pathogen infection and host immunity. Acta Pharmacol Sin 2024; 45:449-464. [PMID: 37993536 PMCID: PMC10834987 DOI: 10.1038/s41401-023-01189-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023] Open
Abstract
As the hub of cellular lipid metabolism, lipid droplets (LDs) have been linked to a variety of biological processes. During pathogen infection, the biogenesis, composition, and functions of LDs are tightly regulated. The accumulation of LDs has been described as a hallmark of pathogen infection and is thought to be driven by pathogens for their own benefit. Recent studies have revealed that LDs and their subsequent lipid mediators contribute to effective immunological responses to pathogen infection by promoting host stress tolerance and reducing toxicity. In this comprehensive review, we delve into the intricate roles of LDs in governing the replication and assembly of a wide spectrum of pathogens within host cells. We also discuss the regulatory function of LDs in host immunity and highlight the potential for targeting LDs for the diagnosis and treatment of infectious diseases.
Collapse
Affiliation(s)
- Yan-Jie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Yi Jin
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yun-Fan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
44
|
Bórquez JC, Díaz-Castro F, La Fuente FPD, Espinoza K, Figueroa AM, Martínez-Ruíz I, Hernández V, López-Soldado I, Ventura R, Domingo JC, Bosch M, Fajardo A, Sebastián D, Espinosa A, Pol A, Zorzano A, Cortés V, Hernández-Alvarez MI, Troncoso R. Mitofusin-2 induced by exercise modifies lipid droplet-mitochondria communication, promoting fatty acid oxidation in male mice with NAFLD. Metabolism 2024; 152:155765. [PMID: 38142958 DOI: 10.1016/j.metabol.2023.155765] [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: 08/29/2023] [Revised: 12/09/2023] [Accepted: 12/16/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND AND AIM The excessive accumulation of lipid droplets (LDs) is a defining characteristic of nonalcoholic fatty liver disease (NAFLD). The interaction between LDs and mitochondria is functionally important for lipid metabolism homeostasis. Exercise improves NAFLD, but it is not known if it has an effect on hepatic LD-mitochondria interactions. Here, we investigated the influence of exercise on LD-mitochondria interactions and its significance in the context of NAFLD. APPROACH AND RESULTS Mice were fed high-fat diet (HFD) or HFD-0.1 % methionine and choline-deficient diet (MCD) to emulate simple hepatic steatosis or non-alcoholic steatohepatitis, respectively. In both models, aerobic exercise decreased the size of LDs bound to mitochondria and the number of LD-mitochondria contacts. Analysis showed that the effects of exercise on HOMA-IR and liver triglyceride levels were independent of changes in body weight, and a positive correlation was observed between the number of LD-mitochondria contacts and NAFLD severity and with the lipid droplet size bound to mitochondria. Cellular fractionation studies revealed that ATP-coupled respiration and fatty acid oxidation (FAO) were greater in hepatic peridroplet mitochondria (PDM) from HFD-fed exercised mice than from equivalent sedentary mice. Finally, exercise increased FAO and mitofusin-2 abundance exclusively in PDM through a mechanism involving the curvature of mitochondrial membranes and the abundance of saturated lipids. Accordingly, hepatic mitofusin-2 ablation prevented exercise-induced FAO in PDM. CONCLUSIONS This study demonstrates that aerobic exercise has beneficial effects in murine NAFLD models by lessening the interactions between hepatic LDs and mitochondria, and by decreasing LD size, correlating with a reduced severity of NAFLD. Additionally, aerobic exercise increases FAO in PDM and this process is reliant on Mfn-2 enrichment, which modifies LD-mitochondria communication.
Collapse
Affiliation(s)
- Juan Carlos Bórquez
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Chile
| | - Francisco Díaz-Castro
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Chile
| | - Francisco Pino-de La Fuente
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Chile
| | - Karla Espinoza
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Chile
| | - Ana María Figueroa
- Department of Nutrition, Diabetes and Metabolism, Pontificia Universidad Católica de Chile, Chile
| | - Inma Martínez-Ruíz
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona IBUB, Barcelona, Spain
| | - Vanessa Hernández
- Institute for Research in Biomedicine (IRB Barcelona). The Barcelona Institute of Science and Technology, Barcelona (BIST), Spain
| | - Iliana López-Soldado
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona IBUB, Barcelona, Spain
| | - Raúl Ventura
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona IBUB, Barcelona, Spain
| | - Joan Carles Domingo
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Marta Bosch
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alba Fajardo
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - David Sebastián
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Alejandra Espinosa
- Escuela de Medicina, Campus San Felipe, Universidad de Valparaíso, Chile; Department of Medical Technology, Faculty of Medicine, University of Chile, Chile
| | - Albert Pol
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Antonio Zorzano
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona). The Barcelona Institute of Science and Technology, Barcelona (BIST), Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Víctor Cortés
- Department of Nutrition, Diabetes and Metabolism, Pontificia Universidad Católica de Chile, Chile.
| | - María Isabel Hernández-Alvarez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona IBUB, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain.
| | - Rodrigo Troncoso
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Chile; Obesity-induced Accelerated Aging (ObAGE), Universidad de Chile, Chile.
| |
Collapse
|
45
|
Alemany M. The Metabolic Syndrome, a Human Disease. Int J Mol Sci 2024; 25:2251. [PMID: 38396928 PMCID: PMC10888680 DOI: 10.3390/ijms25042251] [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: 12/01/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
This review focuses on the question of metabolic syndrome (MS) being a complex, but essentially monophyletic, galaxy of associated diseases/disorders, or just a syndrome of related but rather independent pathologies. The human nature of MS (its exceptionality in Nature and its close interdependence with human action and evolution) is presented and discussed. The text also describes the close interdependence of its components, with special emphasis on the description of their interrelations (including their syndromic development and recruitment), as well as their consequences upon energy handling and partition. The main theories on MS's origin and development are presented in relation to hepatic steatosis, type 2 diabetes, and obesity, but encompass most of the MS components described so far. The differential effects of sex and its biological consequences are considered under the light of human social needs and evolution, which are also directly related to MS epidemiology, severity, and relations with senescence. The triggering and maintenance factors of MS are discussed, with especial emphasis on inflammation, a complex process affecting different levels of organization and which is a critical element for MS development. Inflammation is also related to the operation of connective tissue (including the adipose organ) and the widely studied and acknowledged influence of diet. The role of diet composition, including the transcendence of the anaplerotic maintenance of the Krebs cycle from dietary amino acid supply (and its timing), is developed in the context of testosterone and β-estradiol control of the insulin-glycaemia hepatic core system of carbohydrate-triacylglycerol energy handling. The high probability of MS acting as a unique complex biological control system (essentially monophyletic) is presented, together with additional perspectives/considerations on the treatment of this 'very' human disease.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
46
|
Cinato M, Andersson L, Miljanovic A, Laudette M, Kunduzova O, Borén J, Levin MC. Role of Perilipins in Oxidative Stress-Implications for Cardiovascular Disease. Antioxidants (Basel) 2024; 13:209. [PMID: 38397807 PMCID: PMC10886189 DOI: 10.3390/antiox13020209] [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: 12/09/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Oxidative stress is the imbalance between the production of reactive oxygen species (ROS) and antioxidants in a cell. In the heart, oxidative stress may deteriorate calcium handling, cause arrhythmia, and enhance maladaptive cardiac remodeling by the induction of hypertrophic and apoptotic signaling pathways. Consequently, dysregulated ROS production and oxidative stress have been implicated in numerous cardiac diseases, including heart failure, cardiac ischemia-reperfusion injury, cardiac hypertrophy, and diabetic cardiomyopathy. Lipid droplets (LDs) are conserved intracellular organelles that enable the safe and stable storage of neutral lipids within the cytosol. LDs are coated with proteins, perilipins (Plins) being one of the most abundant. In this review, we will discuss the interplay between oxidative stress and Plins. Indeed, LDs and Plins are increasingly being recognized for playing a critical role beyond energy metabolism and lipid handling. Numerous reports suggest that an essential purpose of LD biogenesis is to alleviate cellular stress, such as oxidative stress. Given the yet unmet suitability of ROS as targets for the intervention of cardiovascular disease, the endogenous antioxidant capacity of Plins may be beneficial.
Collapse
Affiliation(s)
- Mathieu Cinato
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Linda Andersson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Azra Miljanovic
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Marion Laudette
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University—Paul Sabatier, 31432 Toulouse, France;
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Malin C. Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| |
Collapse
|
47
|
Gao X, Jian L, Zhang L, Xu Y, Zhao Y, Yang Y, Yuan Y, Wang Y, Xu S, Ren B, Li Z, Wang C, Li J, Gu Y, Ye J. Perilipin 5 protects the mitochondrial oxidative functions and improves the alcoholic liver injury in mice. Liver Int 2024; 44:357-369. [PMID: 37933091 DOI: 10.1111/liv.15775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/20/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND AND AIMS Alcohol consumption is a well-established risk factor for the onset and progression of hepatic steatosis. Perilipin 5 (Plin5), a lipid droplet protein, is an important protective factor against hepatic lipotoxicity induced by excessive lipolysis, but its role and molecular mechanism in alcoholic liver disease (ALD) are not fully elucidated. METHODS The optimized National Institute on Alcohol Abuse and Alcoholism model was used to construct ALD model mice. Automatic biochemical analyser was used for Biochemical Parameters. The primary hepatocytes and Plin5-overexpressed HepG2 cells (including full-length Plin5 and Plin5 deleting 444-464 aa) were used for in vitro experiment. Haematoxylin and Eosin staining, Oil Red O staining, Bodipy 493/503 staining, Periodic Acid-Schiff staining, immunohistochemistry and JC-1 staining were used to evaluate cell morphology, lipids, glycogen, inflammation and membrane potential. Commercially kits are used to detect glycolipid metabolites, such as triglycerides, glycogen, glucose, reactive oxygen species, lactic acids, ketone bodies. Fluorescently labelled deoxyglucose, NBDG, was used for glucose intake. An XF96 extracellular flux analyser was used to determinate oxygen consumption rate in hepatocytes. The morphological and structural damage of mitochondria was evaluated by electron microscopy. Classical ultracentrifugation is used to separate the subcellular organelles of tissues and cells. Immunoblotting and qPCR were used to detect changes in mRNA and protein levels of related genes. RESULTS Our results showed that the expression of Plin5 in mouse livers was enhanced by alcohol intake, and Plin5 deficiency aggravated the alcohol-induced liver injury. To clarify the mechanism, we found that Plin5 deficiency significantly elevated the hepatic NADH levels and ketone body production in the alcohol-treated mice. As NADH elevation could promote the reduction of pyruvate into lactate and then inhibit the gluconeogenesis, alcohol-treated Plin5-deficient mice exhibited more lactate production and severer hypoglycemia. These results implied that Plin5 deficiency impaired the mitochondrial oxidative functions in the presence of alcohol. In addition, we demonstrated that Plin5 could be recruited onto mitochondria by alcohol, while Plin5 without mitochondrial targeting sequences lost its mitochondrial protection functions. CONCLUSION Collectively, this study demonstrated that the mitochondrial Plin5 could protect the alcohol-induced mitochondrial injury, which provides an important new insight on the roles of Plin5 in highly oxidative tissues.
Collapse
Affiliation(s)
- Xing Gao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lele Jian
- State Key Laboratory of Cancer Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- Shaanxi Provincial Corps, Chinese People's Armed Police Force, Xi'an, China
| | - Lijun Zhang
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuqiao Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuanlin Zhao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ying Yang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuan Yuan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuying Wang
- State Key Laboratory of Cancer Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shenhui Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bincheng Ren
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zimeng Li
- State Key Laboratory of Cancer Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chao Wang
- Department of Pathology, The General Hospital of Western Theater Command, Chengdu, China
| | - Jing Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yu Gu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jing Ye
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
48
|
Ghosh S, Bandyopadhyay S, Smith DM, Adak S, Semenkovich CF, Nagy L, Wolfgang MJ, O’Connor TJ. Legionella pneumophila usurps host cell lipids for vacuole expansion and bacterial growth. PLoS Pathog 2024; 20:e1011996. [PMID: 38386622 PMCID: PMC10883544 DOI: 10.1371/journal.ppat.1011996] [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: 07/27/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Vacuolar pathogens reside in membrane-bound compartments within host cells. Maintaining the integrity of this compartment is paramount to bacterial survival and replication as it protects against certain host surveillance mechanisms that function to eradicate invading pathogens. Preserving this compartment during bacterial replication requires expansion of the vacuole membrane to accommodate the increasing number of bacteria, and yet, how this is accomplished remains largely unknown. Here, we show that the vacuolar pathogen Legionella pneumophila exploits multiple sources of host cell fatty acids, including inducing host cell fatty acid scavenging pathways, in order to promote expansion of the replication vacuole and bacteria growth. Conversely, when exogenous lipids are limited, the decrease in host lipid availability restricts expansion of the replication vacuole membrane, resulting in a higher density of bacteria within the vacuole. Modifying the architecture of the vacuole prioritizes bacterial growth by allowing the greatest number of bacteria to remain protected by the vacuole membrane despite limited resources for its expansion. However, this trade-off is not without risk, as it can lead to vacuole destabilization, which is detrimental to the pathogen. However, when host lipid resources become extremely scarce, for example by inhibiting host lipid scavenging, de novo biosynthetic pathways, and/or diverting host fatty acids to storage compartments, bacterial replication becomes severely impaired, indicating that host cell fatty acid availability also directly regulates L. pneumophila growth. Collectively, these data demonstrate dual roles for host cell fatty acids in replication vacuole expansion and bacterial proliferation, revealing the central functions for these molecules and their metabolic pathways in L. pneumophila pathogenesis.
Collapse
Affiliation(s)
- Soma Ghosh
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Saumya Bandyopadhyay
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Danielle M. Smith
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sangeeta Adak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Clay F. Semenkovich
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Laszlo Nagy
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, United States of America
| | - Michael J. Wolfgang
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Tamara J. O’Connor
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
49
|
Teixeira L, Pereira-Dutra FS, Reis PA, Cunha-Fernandes T, Yoshinaga MY, Souza-Moreira L, Souza EK, Barreto EA, Silva TP, Espinheira-Silva H, Igreja T, Antunes MM, Bombaça ACS, Gonçalves-de-Albuquerque CF, Menezes GB, Hottz ED, Menna-Barreto RF, Maya-Monteiro CM, Bozza FA, Miyamoto S, Melo RC, Bozza PT. Prevention of lipid droplet accumulation by DGAT1 inhibition ameliorates sepsis-induced liver injury and inflammation. JHEP Rep 2024; 6:100984. [PMID: 38293685 PMCID: PMC10827501 DOI: 10.1016/j.jhepr.2023.100984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 11/11/2023] [Accepted: 11/21/2023] [Indexed: 02/01/2024] Open
Abstract
Background & Aims Lipid droplet (LD) accumulation in cells and tissues is understood to be an evolutionarily conserved tissue tolerance mechanism to prevent lipotoxicity caused by excess lipids; however, the presence of excess LDs has been associated with numerous diseases. Sepsis triggers the reprogramming of lipid metabolism and LD accumulation in cells and tissues, including the liver. The functions and consequences of sepsis-triggered liver LD accumulation are not well known. Methods Experimental sepsis was induced by CLP (caecal ligation and puncture) in mice. Markers of hepatic steatosis, liver injury, hepatic oxidative stress, and inflammation were analysed using a combination of functional, imaging, lipidomic, protein expression and immune-enzymatic assays. To prevent LD formation, mice were treated orally with A922500, a pharmacological inhibitor of DGAT1. Results We identified that liver LD overload correlates with liver injury and sepsis severity. Moreover, the progression of steatosis from 24 h to 48 h post-CLP occurs in parallel with increased cytokine expression, inflammatory cell recruitment and oxidative stress. Lipidomic analysis of purified LDs demonstrated that sepsis leads LDs to harbour increased amounts of unsaturated fatty acids, mostly 18:1 and 18:2. An increased content of lipoperoxides within LDs was also observed. Conversely, the impairment of LD formation by inhibition of the DGAT1 enzyme reduces levels of hepatic inflammation and lipid peroxidation markers and ameliorates sepsis-induced liver injury. Conclusions Our results indicate that sepsis triggers lipid metabolism alterations that culminate in increased liver LD accumulation. Increased LDs are associated with disease severity and liver injury. Moreover, inhibition of LD accumulation decreased the production of inflammatory mediators and lipid peroxidation while improving tissue function, suggesting that LDs contribute to the pathogenesis of liver injury triggered by sepsis. Impact and Implications Sepsis is a complex life-threatening syndrome caused by dysregulated inflammatory and metabolic host responses to infection. The observation that lipid droplets may contribute to sepsis-associated organ injury by amplifying lipid peroxidation and inflammation provides a rationale for therapeutically targeting lipid droplets and lipid metabolism in sepsis.
Collapse
Affiliation(s)
- Lívia Teixeira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Filipe S. Pereira-Dutra
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, FIOCRUZ, Rio de Janeiro, Brazil
| | - Patrícia A. Reis
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Biochemistry Department, Roberto Alcântara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tamires Cunha-Fernandes
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, FIOCRUZ, Rio de Janeiro, Brazil
| | - Marcos Y. Yoshinaga
- Laboratory of Modified Lipids, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Luciana Souza-Moreira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Ellen K. Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Ester A. Barreto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Thiago P. Silva
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences (ICB), Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Hugo Espinheira-Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, FIOCRUZ, Rio de Janeiro, Brazil
| | - Tathiany Igreja
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Maísa M. Antunes
- Center for Gastrointestinal Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina S. Bombaça
- Laboratory of Cellular Biology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratory of Parasitic Disease, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Cassiano F. Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo B. Menezes
- Center for Gastrointestinal Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Eugênio D. Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | | | - Clarissa M. Maya-Monteiro
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, FIOCRUZ, Rio de Janeiro, Brazil
| | - Fernando A. Bozza
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, FIOCRUZ, Rio de Janeiro, Brazil
- Intensive Care Medicine Laboratory, INI, FIOCRUZ, Rio de Janeiro, Brazil
- D'Or Institute Research and Education (IDOr), Rio de Janeiro, Brazil
| | - Sayuri Miyamoto
- Laboratory of Modified Lipids, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Rossana C.N. Melo
- Laboratory of Cellular Biology, Department of Biology, Institute of Biological Sciences (ICB), Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Patrícia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
50
|
Landowski M, Gogoi P, Ikeda S, Ikeda A. Roles of transmembrane protein 135 in mitochondrial and peroxisomal functions - implications for age-related retinal disease. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1355379. [PMID: 38576540 PMCID: PMC10993500 DOI: 10.3389/fopht.2024.1355379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Aging is the most significant risk factor for age-related diseases in general, which is true for age-related diseases in the eye including age-related macular degeneration (AMD). Therefore, in order to identify potential therapeutic targets for these diseases, it is crucial to understand the normal aging process and how its mis-regulation could cause age-related diseases at the molecular level. Recently, abnormal lipid metabolism has emerged as one major aspect of age-related symptoms in the retina. Animal models provide excellent means to identify and study factors that regulate lipid metabolism in relation to age-related symptoms. Central to this review is the role of transmembrane protein 135 (TMEM135) in the retina. TMEM135 was identified through the characterization of a mutant mouse strain exhibiting accelerated retinal aging and positional cloning of the responsible mutation within the gene, indicating the crucial role of TMEM135 in regulating the normal aging process in the retina. Over the past decade, the molecular functions of TMEM135 have been explored in various models and tissues, providing insights into the regulation of metabolism, particularly lipid metabolism, through its action in multiple organelles. Studies indicated that TMEM135 is a significant regulator of peroxisomes, mitochondria, and their interaction. Here, we provide an overview of the molecular functions of TMEM135 which is crucial for regulating mitochondria, peroxisomes, and lipids. The review also discusses the age-dependent phenotypes in mice with TMEM135 perturbations, emphasizing the importance of a balanced TMEM135 function for the health of the retina and other tissues including the heart, liver, and adipose tissue. Finally, we explore the potential roles of TMEM135 in human age-related retinal diseases, connecting its functions to the pathobiology of AMD.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Purnima Gogoi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
| | - Sakae Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|