1
|
Prota G, Berloffa G, Awad W, Vacchini A, Chancellor A, Schaefer V, Constantin D, Littler DR, Colombo R, Nosi V, Mori L, Rossjohn J, De Libero G. Mitochondria regulate MR1 protein expression and produce self-metabolites that activate MR1-restricted T cells. Proc Natl Acad Sci U S A 2025; 122:e2418525122. [PMID: 40354545 DOI: 10.1073/pnas.2418525122] [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: 09/11/2024] [Accepted: 03/11/2025] [Indexed: 05/14/2025] Open
Abstract
Mitochondria coordinate several metabolic pathways, producing metabolites that influence the immune response in various ways. It remains unclear whether mitochondria impact antigen presentation by the MHC-class-I-related antigen-presenting molecule, MR1, which presents small molecules to MR1-restricted T-lymphocytes. Here, we demonstrate that mitochondrial complex III and the enzyme dihydroorotate dehydrogenase are essential for the cell-surface expression of MR1 and for generating uridine- and thymidine-related compounds that bind to MR1 and are produced upon oxidation by reactive oxygen species. One mitochondria-derived immunogenic formylated metabolite we identified is 5-formyl-deoxyuridine (5-FdU). Structural studies indicate that 5-FdU binds in the A'-antigen-binding pocket of MR1, positioning the deoxyribose toward the surface of MR1 for TCR interaction. 5-FdU stimulates specific T cells and detects circulating T cells when loaded onto MR1-tetramers. 5-FdU-reactive cells resemble adaptive T cells and express the phenotypes of naïve, memory, and effector cells, indicating prior in vivo stimulation. These findings suggest that mitochondria may play a role in MR1-mediated immune surveillance.
Collapse
Affiliation(s)
- Gennaro Prota
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Giuliano Berloffa
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Wael Awad
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Alessandro Vacchini
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Andrew Chancellor
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Verena Schaefer
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Daniel Constantin
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Dene R Littler
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Rodrigo Colombo
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Vladimir Nosi
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4YS, United Kingdom
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel 4031, Switzerland
| |
Collapse
|
2
|
Paul S, Morgan P, Pernes G, Schooneveldt Y, Duong T, Mellett NA, Huynh K, Murphy AJ, Lancaster GI, Meikle PJ. Modulation of endogenous plasmalogens by genetic ablation of lysoplasmalogenase (Tmem86b) in mice. J Lipid Res 2025; 66:100808. [PMID: 40245986 DOI: 10.1016/j.jlr.2025.100808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
Plasmalogens are a distinct subclass of glycerophospholipids that exhibit unique structural features, notably possessing a vinyl ether linkage at the sn1 position of the glycerol backbone. These specialized lipids play crucial roles in various biological functions. Although the biosynthetic pathway of plasmalogens has been well-characterized, their catabolism remains less studied. In this study, we investigated the impact of global and tissue-specific loss-of-function of a plasmalogen catabolizing enzyme, lysoplasmalogenase (TMEM86B), on circulatory and tissue lipidomes. We generated both global and hepatocyte-specific Tmem86b knockout mice using cre-loxP technology. Mice with homozygous global inactivation of Tmem86b (Tmem86b KO mice) were viable and did not display any marked phenotypic abnormalities. Tmem86b KO mice demonstrated significantly elevated levels of the plasmalogens, alkenylphosphatidylethanolamine (PE(P)), and alkenylphosphatidylcholine (PC(P)), as well as lysoplasmalogens, in the plasma, liver, and natural killer cells compared to their wild-type counterparts. The endogenous alkenyl chain composition of plasmalogens remained unaltered in Tmem86b KO mice. Consistent with the global knockout findings, hepatocyte-specific Tmem86b knockout mice also exhibited increased plasmalogen levels in the plasma and liver compared to their floxed control counterparts. Overall, our findings shed light on the role of Tmem86b in plasmalogen catabolism, demonstrating how its ablation leads to elevated plasmalogen levels in select tissues and cells. This study enhances our understanding of the regulatory mechanisms governing plasmalogen metabolism and highlights the potential of targeting Tmem86b to therapeutically raise plasmalogen levels.
Collapse
Affiliation(s)
- Sudip Paul
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia.
| | - Pooranee Morgan
- Haematopoesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gerard Pernes
- Haematopoesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yvette Schooneveldt
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Thy Duong
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew J Murphy
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia; Haematopoesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Graeme I Lancaster
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Haematopoesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
3
|
Hosono Y, Tomiyasu N, Kasai H, Ishikawa E, Takahashi M, Imamura A, Ishida H, Compostella F, Kida H, Kumanogoh A, Bamba T, Izumi Y, Yamasaki S. Identification of α-galactosylceramide as an endogenous mammalian antigen for iNKT cells. J Exp Med 2025; 222:e20240728. [PMID: 39704712 DOI: 10.1084/jem.20240728] [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: 04/24/2024] [Revised: 10/08/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
Invariant natural killer T (iNKT) cells are unconventional T cells recognizing lipid antigens in a CD1d-restricted manner. Among these lipid antigens, α-galactosylceramide (α-GalCer), which was originally identified in marine sponges, is the most potent antigen. Although the presence of α-anomeric hexosylceramide and microbiota-derived branched α-GalCer is reported, antigenic α-GalCer has not been identified in mammals. Here, we developed a high-resolution separation and detection system, supercritical fluid chromatography tandem mass spectrometry (SFC/MS/MS), that can discriminate hexosylceramide diastereomers (α-GalCer, α-GlcCer, β-GalCer, or β-GlcCer). The B16 melanoma tumor cell line does not activate iNKT cells; however, ectopic expression of CD1d was sufficient to activate iNKT cells without adding antigens. B16 melanoma was unlikely to generate iNKT cell antigens; instead, antigen activity was detected in cell culture serum. Activity-based purification and SFC/MS/MS identified dihydrosphingosine-based saturated α-GalCer as an antigenic component in serum, bile, and lymphoid tissues. These results show the first evidence for the presence of potent antigenic α-GalCer in mammals.
Collapse
Affiliation(s)
- Yuki Hosono
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University , Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyuki Tomiyasu
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Hayato Kasai
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University , Suita, Japan
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University , Suita, Japan
| | - Masatomo Takahashi
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Akihiro Imamura
- Department of Applied Bioorganic Chemistry, Gifu University, Gifu, Japan
- Institute for Glyco-core Research, Gifu University , Gifu, Japan
| | - Hideharu Ishida
- Department of Applied Bioorganic Chemistry, Gifu University, Gifu, Japan
- Institute for Glyco-core Research, Gifu University , Gifu, Japan
| | - Federica Compostella
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Hiroshi Kida
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Immunopathology, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University , Suita, Japan
- Center for Advanced Modalities and DDS, Osaka University , Suita, Japan
| | - Takeshi Bamba
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Izumi
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University , Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University , Suita, Japan
- Center for Advanced Modalities and DDS, Osaka University , Suita, Japan
| |
Collapse
|
4
|
Sarkar C, Lipinski MM. Role and Function of Peroxisomes in Neuroinflammation. Cells 2024; 13:1655. [PMID: 39404418 PMCID: PMC11476013 DOI: 10.3390/cells13191655] [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: 08/22/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Peroxisomes are organelles involved in many cellular metabolic functions, including the degradation of very-long-chain fatty acids (VLCFAs; C ≥ 22), the initiation of ether-phospholipid synthesis, and the metabolism of reactive oxygen species. All of these processes are essential for the maintenance of cellular lipid and redox homeostasis, and their perturbation can trigger inflammatory response in immune cells, including in the central nervous system (CNS) resident microglia and astrocytes. Consistently, peroxisomal disorders, a group of congenital diseases caused by a block in peroxisomal biogenesis or the impairment of one of the peroxisomal enzymes, are associated with neuroinflammation. Peroxisomal function is also dysregulated in many neurodegenerative diseases and during brain aging, both of which are associated with neuroinflammation. This suggests that deciphering the role of peroxisomes in neuroinflammation may be important for understanding both congenital and age-related brain dysfunction. In this review, we discuss the current advances in understanding the role and function of peroxisomes in neuroinflammation.
Collapse
Affiliation(s)
- Chinmoy Sarkar
- Shock, Trauma and Anesthesiology Research (STAR) Center, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Marta M. Lipinski
- Shock, Trauma and Anesthesiology Research (STAR) Center, Department of Anesthesiology and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| |
Collapse
|
5
|
Cheng TY, Praveena T, Govindarajan S, Almeida CF, Pellicci DG, Arkins WC, Van Rhijn I, Venken K, Elewaut D, Godfrey DI, Rossjohn J, Moody DB. Lipidomic scanning of self-lipids identifies headless antigens for natural killer T cells. Proc Natl Acad Sci U S A 2024; 121:e2321686121. [PMID: 39141352 PMCID: PMC11348285 DOI: 10.1073/pnas.2321686121] [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: 01/08/2024] [Accepted: 06/12/2024] [Indexed: 08/15/2024] Open
Abstract
To broadly measure the spectrum of cellular self-antigens for natural killer T cells (NKT), we developed a sensitive lipidomics system to analyze lipids trapped between CD1d and NKT T cell receptors (TCRs). We captured diverse antigen complexes formed in cells from natural endogenous lipids, with or without inducing endoplasmic reticulum (ER) stress. After separating protein complexes with no, low, or high CD1d-TCR interaction, we eluted lipids to establish the spectrum of self-lipids that facilitate this interaction. Although this unbiased approach identified fifteen molecules, they clustered into only two related groups: previously known phospholipid antigens and unexpected neutral lipid antigens. Mass spectrometry studies identified the neutral lipids as ceramides, deoxyceramides, and diacylglycerols, which can be considered headless lipids because they lack polar headgroups that usually form the TCR epitope. The crystal structure of the TCR-ceramide-CD1d complex showed how the missing headgroup allowed the TCR to predominantly contact CD1d, supporting a model of CD1d autoreactivity. Ceramide and related headless antigens mediated physiological TCR binding affinity, weak NKT cell responses, and tetramer binding to polyclonal human and mouse NKT cells. Ceramide and sphingomyelin are oppositely regulated components of the "sphingomyelin cycle" that are altered during apoptosis, transformation, and ER stress. Thus, the unique molecular link of ceramide to NKT cell response, along with the recent identification of sphingomyelin blockers of NKT cell activation, provide two mutually reinforcing links for NKT cell response to sterile cellular stress conditions.
Collapse
Affiliation(s)
- Tan-Yun Cheng
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02210
| | - T. Praveena
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC3800, Australia
| | - Srinath Govindarajan
- Molecular Immunology and Inflammation Unit, Vlaams Instituut voor Biotechnologie, Center for Inflammation Research, Ghent University, 9052Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, 9000Ghent, Belgium
| | - Catarina F. Almeida
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC3010, Australia
| | - Daniel G. Pellicci
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC3010, Australia
| | - Wellington C. Arkins
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02210
| | - Ildiko Van Rhijn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02210
| | - Koen Venken
- Molecular Immunology and Inflammation Unit, Vlaams Instituut voor Biotechnologie, Center for Inflammation Research, Ghent University, 9052Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, 9000Ghent, Belgium
| | - Dirk Elewaut
- Molecular Immunology and Inflammation Unit, Vlaams Instituut voor Biotechnologie, Center for Inflammation Research, Ghent University, 9052Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, 9000Ghent, Belgium
| | - Dale I. Godfrey
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC3010, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, CardiffCF14 4XN, UK
| | - D. Branch Moody
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02210
| |
Collapse
|
6
|
Salihovic S, Nyström N, Mathisen CBW, Kruse R, Olbjørn C, Andersen S, Noble AJ, Dorn-Rasmussen M, Bazov I, Perminow G, Opheim R, Detlie TE, Huppertz-Hauss G, Hedin CRH, Carlson M, Öhman L, Magnusson MK, Keita ÅV, Söderholm JD, D'Amato M, Orešič M, Wewer V, Satsangi J, Lindqvist CM, Burisch J, Uhlig HH, Repsilber D, Hyötyläinen T, Høivik ML, Halfvarson J. Identification and validation of a blood- based diagnostic lipidomic signature of pediatric inflammatory bowel disease. Nat Commun 2024; 15:4567. [PMID: 38830848 PMCID: PMC11148148 DOI: 10.1038/s41467-024-48763-7] [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: 05/19/2023] [Accepted: 04/30/2024] [Indexed: 06/05/2024] Open
Abstract
Improved biomarkers are needed for pediatric inflammatory bowel disease. Here we identify a diagnostic lipidomic signature for pediatric inflammatory bowel disease by analyzing blood samples from a discovery cohort of incident treatment-naïve pediatric patients and validating findings in an independent inception cohort. The lipidomic signature comprising of only lactosyl ceramide (d18:1/16:0) and phosphatidylcholine (18:0p/22:6) improves the diagnostic prediction compared with high-sensitivity C-reactive protein. Adding high-sensitivity C-reactive protein to the signature does not improve its performance. In patients providing a stool sample, the diagnostic performance of the lipidomic signature and fecal calprotectin, a marker of gastrointestinal inflammation, does not substantially differ. Upon investigation in a third pediatric cohort, the findings of increased lactosyl ceramide (d18:1/16:0) and decreased phosphatidylcholine (18:0p/22:6) absolute concentrations are confirmed. Translation of the lipidomic signature into a scalable diagnostic blood test for pediatric inflammatory bowel disease has the potential to support clinical decision making.
Collapse
Affiliation(s)
- Samira Salihovic
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Niklas Nyström
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Charlotte Bache-Wiig Mathisen
- Department of Gastroenterology, Oslo University Hospital, Oslo, Norway and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Robert Kruse
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Christine Olbjørn
- Department of Pediatrics and Adolescent Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Svend Andersen
- Department of Pediatrics, Vestfold Hospital Trust, Tønsberg, Norway
| | - Alexandra J Noble
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
- Biomedical Research Center, University of Oxford, Oxford, United Kingdom
| | - Maria Dorn-Rasmussen
- Department of Paediatric and Adolescence Medicine, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
| | - Igor Bazov
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Gøri Perminow
- Department of Pediatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Randi Opheim
- Department of Gastroenterology, Oslo University Hospital, Oslo, Norway and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trond Espen Detlie
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway and Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Charlotte R H Hedin
- Karolinska Institutet, Department of Medicine Solna, Stockholm, Sweden
- Karolinska University Hospital, Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Stockholm, Sweden
| | - Marie Carlson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria K Magnusson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan D Söderholm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mauro D'Amato
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Gastrointestinal Genetics Lab, CIC bioGUNE - BRTA, Derio, Spain
- Department of Medicine & Surgery, LUM University, Casamassima, Italy
| | - Matej Orešič
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Vibeke Wewer
- Department of Paediatric and Adolescence Medicine, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
- Biomedical Research Center, University of Oxford, Oxford, United Kingdom
| | - Carl Mårten Lindqvist
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Johan Burisch
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Gastrounit, medical division, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
- Biomedical Research Center, University of Oxford, Oxford, United Kingdom
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Dirk Repsilber
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | | | - Marte Lie Høivik
- Department of Gastroenterology, Oslo University Hospital, Oslo, Norway and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| |
Collapse
|
7
|
Qin X, Wang B, Lu X, Song Y, Wang W. Identification and Validation of a PEX5-Dependent Signature for Prognostic Prediction in Glioma. Biomolecules 2024; 14:314. [PMID: 38540734 PMCID: PMC10967733 DOI: 10.3390/biom14030314] [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: 02/07/2024] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Gliomas, the most prevalent and lethal form of brain cancer, are known to exhibit metabolic alterations that facilitate tumor growth, invasion, and resistance to therapies. Peroxisomes, essential organelles responsible for fatty acid oxidation and reactive oxygen species (ROS) homeostasis, rely on the receptor PEX5 for the import of metabolic enzymes into their matrix. However, the prognostic significance of peroxisomal enzymes for glioma patients remains unclear. In this study, we elucidate that PEX5 is indispensable for the cell growth, migration, and invasion of glioma cells. We establish a robust prognosis model based on the expression of peroxisomal enzymes, whose localization relies on PEX5. This PEX5-dependent signature not only serves as a robust prognosis model capable of accurately predicting outcomes for glioma patients, but also effectively distinguishes several clinicopathological features, including the grade, isocitrate dehydrogenase (IDH) mutation, and 1p19q codeletion status. Furthermore, we developed a nomogram that integrates the prognostic model with other clinicopathological factors, demonstrating highly accurate performance in estimating patient survival. Patients classified into the high-risk group based on our prognostic model exhibited an immunosuppressive microenvironment. Finally, our validation reveals that the elevated expression of GSTK1, an antioxidant enzyme within the signature, promotes the cell growth and migration of glioma cells, with this effect dependent on the peroxisomal targeting signal recognized by PEX5. These findings identify the PEX5-dependent signature as a promising prognostic tool for gliomas.
Collapse
Affiliation(s)
| | | | | | | | - Wei Wang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| |
Collapse
|
8
|
Parsons BD, Medina-Luna D, Scur M, Pinelli M, Gamage GS, Chilvers RA, Hamon Y, Ahmed IHI, Savary S, Makrigiannis AP, Braverman NE, Rodriguez-Alcazar JF, Latz E, Karakach TK, Di Cara F. Peroxisome deficiency underlies failures in hepatic immune cell development and antigen presentation in a severe Zellweger disease model. Cell Rep 2024; 43:113744. [PMID: 38329874 DOI: 10.1016/j.celrep.2024.113744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Peroxisome biogenesis disorders (PBDs) represent a group of metabolic conditions that cause severe developmental defects. Peroxisomes are essential metabolic organelles, present in virtually every eukaryotic cell and mediating key processes in immunometabolism. To date, the full spectrum of PBDs remains to be identified, and the impact PBDs have on immune function is unexplored. This study presents a characterization of the hepatic immune compartment of a neonatal PBD mouse model at single-cell resolution to establish the importance and function of peroxisomes in developmental hematopoiesis. We report that hematopoietic defects are a feature in a severe PBD murine model. Finally, we identify a role for peroxisomes in the regulation of the major histocompatibility class II expression and antigen presentation to CD4+ T cells in dendritic cells. This study adds to our understanding of the mechanisms of PBDs and expands our knowledge of the role of peroxisomes in immunometabolism.
Collapse
Affiliation(s)
- Brendon D Parsons
- University of Alberta, Department of Laboratory Medicine and Pathology, Edmonton, AB T6G 1C9, Canada
| | - Daniel Medina-Luna
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Michal Scur
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Marinella Pinelli
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Gayani S Gamage
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Rebecca A Chilvers
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Yannick Hamon
- Aix Marseille University, CNRS, INSERM au Centre d'Immunologie de Marseille Luminy, 13288 Marseille, France
| | - Ibrahim H I Ahmed
- Dalhousie University, Department of Pharmacology, Halifax, NS B3H 4R2, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Stéphane Savary
- University of Bourgogne, Laboratoire Bio-PeroxIL EA7270, Dijon, France
| | - Andrew P Makrigiannis
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Nancy E Braverman
- Research Institute of the McGill University Children's Hospital, Montreal, QC H4A 3J1, Canada
| | | | - Eicke Latz
- University of Bonn, Institute of Innate Immunity, Medical Faculty, 53127 Bonn, Germany
| | - Tobias K Karakach
- Dalhousie University, Department of Pharmacology, Halifax, NS B3H 4R2, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Francesca Di Cara
- University of Alberta, Department of Laboratory Medicine and Pathology, Edmonton, AB T6G 1C9, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada.
| |
Collapse
|
9
|
Huang S, Shahine A, Cheng TY, Chen YL, Ng SW, Balaji GR, Farquhar R, Gras S, Hardman CS, Altman JD, Tahiri N, Minnaard AJ, Ogg GS, Mayfield JA, Rossjohn J, Moody DB. CD1 lipidomes reveal lipid-binding motifs and size-based antigen-display mechanisms. Cell 2023; 186:4583-4596.e13. [PMID: 37725977 PMCID: PMC10591967 DOI: 10.1016/j.cell.2023.08.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 05/05/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023]
Abstract
The CD1 system binds lipid antigens for display to T cells. Here, we solved lipidomes for the four human CD1 antigen-presenting molecules, providing a map of self-lipid display. Answering a basic question, the detection of >2,000 CD1-lipid complexes demonstrates broad presentation of self-sphingolipids and phospholipids. Whereas peptide antigens are chemically processed, many lipids are presented in an unaltered form. However, each type of CD1 protein differentially edits the self-lipidome to show distinct capture motifs based on lipid length and chemical composition, suggesting general antigen display mechanisms. For CD1a and CD1d, lipid size matches the CD1 cleft volume. CD1c cleft size is more variable, and CD1b is the outlier, where ligands and clefts show an extreme size mismatch that is explained by uniformly seating two small lipids in one cleft. Furthermore, the list of compounds that comprise the integrated CD1 lipidome supports the ongoing discovery of lipid blockers and antigens for T cells.
Collapse
Affiliation(s)
- Shouxiong Huang
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Adam Shahine
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Tan-Yun Cheng
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yi-Ling Chen
- Human Immunology Unit, Weatherall Institute for Molecular Medicine, University of Oxford, Headington, Oxford OX3 9DS, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Soo Weei Ng
- Human Immunology Unit, Weatherall Institute for Molecular Medicine, University of Oxford, Headington, Oxford OX3 9DS, UK
| | - Gautham R. Balaji
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Rachel Farquhar
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Clare S. Hardman
- Human Immunology Unit, Weatherall Institute for Molecular Medicine, University of Oxford, Headington, Oxford OX3 9DS, UK
| | - John D. Altman
- Emory Vaccine Center, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Nabil Tahiri
- Department of Chemical Biology, Stratingh Institute for Chemistry, Groningen, Netherlands
| | - Adriaan J. Minnaard
- Department of Chemical Biology, Stratingh Institute for Chemistry, Groningen, Netherlands
| | - Graham S. Ogg
- Human Immunology Unit, Weatherall Institute for Molecular Medicine, University of Oxford, Headington, Oxford OX3 9DS, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Jacob A. Mayfield
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
- Institute of Infection and Immunity, Cardiff University, School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - D. Branch Moody
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Lead contact
| |
Collapse
|
10
|
Chaves-Filho AM, Braniff O, Angelova A, Deng Y, Tremblay MÈ. Chronic inflammation, neuroglial dysfunction, and plasmalogen deficiency as a new pathobiological hypothesis addressing the overlap between post-COVID-19 symptoms and myalgic encephalomyelitis/chronic fatigue syndrome. Brain Res Bull 2023; 201:110702. [PMID: 37423295 DOI: 10.1016/j.brainresbull.2023.110702] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/13/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
After five waves of coronavirus disease 2019 (COVID-19) outbreaks, it has been recognized that a significant portion of the affected individuals developed long-term debilitating symptoms marked by chronic fatigue, cognitive difficulties ("brain fog"), post-exertional malaise, and autonomic dysfunction. The onset, progression, and clinical presentation of this condition, generically named post-COVID-19 syndrome, overlap significantly with another enigmatic condition, referred to as myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Several pathobiological mechanisms have been proposed for ME/CFS, including redox imbalance, systemic and central nervous system inflammation, and mitochondrial dysfunction. Chronic inflammation and glial pathological reactivity are common hallmarks of several neurodegenerative and neuropsychiatric disorders and have been consistently associated with reduced central and peripheral levels of plasmalogens, one of the major phospholipid components of cell membranes with several homeostatic functions. Of great interest, recent evidence revealed a significant reduction of plasmalogen contents, biosynthesis, and metabolism in ME/CFS and acute COVID-19, with a strong association to symptom severity and other relevant clinical outcomes. These bioactive lipids have increasingly attracted attention due to their reduced levels representing a common pathophysiological manifestation between several disorders associated with aging and chronic inflammation. However, alterations in plasmalogen levels or their lipidic metabolism have not yet been examined in individuals suffering from post-COVID-19 symptoms. Here, we proposed a pathobiological model for post-COVID-19 and ME/CFS based on their common inflammation and dysfunctional glial reactivity, and highlighted the emerging implications of plasmalogen deficiency in the underlying mechanisms. Along with the promising outcomes of plasmalogen replacement therapy (PRT) for various neurodegenerative/neuropsychiatric disorders, we sought to propose PRT as a simple, effective, and safe strategy for the potential relief of the debilitating symptoms associated with ME/CFS and post-COVID-19 syndrome.
Collapse
Affiliation(s)
| | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, F-91400 Orsay, France
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Department of Molecular Medicine, Université Laval, Québec City, Québec, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, Québec, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Advanced Materials and Related Technology (CAMTEC) and Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
11
|
Cedillo L, Ahsan FM, Li S, Stuhr NL, Zhou Y, Zhang Y, Adedoja A, Murphy LM, Yerevanian A, Emans S, Dao K, Li Z, Peterson ND, Watrous J, Jain M, Das S, Pukkila-Worley R, Curran SP, Soukas AA. Ether lipid biosynthesis promotes lifespan extension and enables diverse pro-longevity paradigms in Caenorhabditis elegans. eLife 2023; 12:e82210. [PMID: 37606250 PMCID: PMC10444025 DOI: 10.7554/elife.82210] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/13/2023] [Indexed: 08/23/2023] Open
Abstract
Biguanides, including the world's most prescribed drug for type 2 diabetes, metformin, not only lower blood sugar, but also promote longevity in preclinical models. Epidemiologic studies in humans parallel these findings, indicating favorable effects of metformin on longevity and on reducing the incidence and morbidity associated with aging-related diseases. Despite this promise, the full spectrum of molecular effectors responsible for these health benefits remains elusive. Through unbiased screening in Caenorhabditis elegans, we uncovered a role for genes necessary for ether lipid biosynthesis in the favorable effects of biguanides. We demonstrate that biguanides prompt lifespan extension by stimulating ether lipid biogenesis. Loss of the ether lipid biosynthetic machinery also mitigates lifespan extension attributable to dietary restriction, target of rapamycin (TOR) inhibition, and mitochondrial electron transport chain inhibition. A possible mechanistic explanation for this finding is that ether lipids are required for activation of longevity-promoting, metabolic stress defenses downstream of the conserved transcription factor skn-1/Nrf. In alignment with these findings, overexpression of a single, key, ether lipid biosynthetic enzyme, fard-1/FAR1, is sufficient to promote lifespan extension. These findings illuminate the ether lipid biosynthetic machinery as a novel therapeutic target to promote healthy aging.
Collapse
Affiliation(s)
- Lucydalila Cedillo
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard Medical SchoolBostonUnited States
| | - Fasih M Ahsan
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard Medical SchoolBostonUnited States
| | - Sainan Li
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| | - Nicole L Stuhr
- Leonard Davis School of Gerontology, University of Southern CaliforniaLos AngelesUnited States
| | - Yifei Zhou
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| | - Yuyao Zhang
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| | - Adebanjo Adedoja
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard Medical SchoolBostonUnited States
| | - Luke M Murphy
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard Medical SchoolBostonUnited States
| | - Armen Yerevanian
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| | - Sinclair Emans
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| | - Khoi Dao
- Department of Medicine and Pharmacology, University of California San DiegoSan DiegoUnited States
| | - Zhaozhi Li
- Biomedical Informatics Core, Massachusetts General Hospital and Harvard Medical SchooCambridgeUnited States
| | - Nicholas D Peterson
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Jeramie Watrous
- Department of Medicine and Pharmacology, University of California San DiegoSan DiegoUnited States
| | - Mohit Jain
- Department of Medicine and Pharmacology, University of California San DiegoSan DiegoUnited States
| | - Sudeshna Das
- Biomedical Informatics Core, Massachusetts General Hospital and Harvard Medical SchooCambridgeUnited States
| | - Read Pukkila-Worley
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Sean P Curran
- Leonard Davis School of Gerontology, University of Southern CaliforniaLos AngelesUnited States
| | - Alexander A Soukas
- Center for Genomic Medicine and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| |
Collapse
|
12
|
Lattanzi G, Strati F, Díaz-Basabe A, Perillo F, Amoroso C, Protti G, Rita Giuffrè M, Iachini L, Baeri A, Baldari L, Cassinotti E, Ghidini M, Galassi B, Lopez G, Noviello D, Porretti L, Trombetta E, Messuti E, Mazzarella L, Iezzi G, Nicassio F, Granucci F, Vecchi M, Caprioli F, Facciotti F. iNKT cell-neutrophil crosstalk promotes colorectal cancer pathogenesis. Mucosal Immunol 2023; 16:326-340. [PMID: 37004750 DOI: 10.1016/j.mucimm.2023.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023]
Abstract
iNKT cells account for a relevant fraction of effector T-cells in the intestine and are considered an attractive platform for cancer immunotherapy. Although iNKT cells are cytotoxic lymphocytes, their functional role in colorectal cancer (CRC) is still controversial, limiting their therapeutic use. Thus, we examined the immune cell composition and iNKT cell phenotype of CRC lesions in patients (n = 118) and different murine models. High-dimensional single-cell flow-cytometry, metagenomics, and RNA sequencing experiments revealed that iNKT cells are enriched in tumor lesions. The tumor-associated pathobiont Fusobacterium nucleatum induces IL-17 and Granulocyte-macrophage colony-stimulating factor (GM-CSF) expression in iNKT cells without affecting their cytotoxic capability but promoting iNKT-mediated recruitment of neutrophils with polymorphonuclear myeloid-derived suppressor cells-like phenotype and functions. The lack of iNKT cells reduced the tumor burden and recruitment of immune suppressive neutrophils. iNKT cells in-vivo activation with α-galactosylceramide restored their anti-tumor function, suggesting that iNKT cells can be modulated to overcome CRC-associated immune evasion. Tumor co-infiltration by iNKT cells and neutrophils correlates with negative clinical outcomes, highlighting the importance of iNKT cells in the pathophysiology of CRC. Our results reveal a functional plasticity of iNKT cells in CRC, suggesting a pivotal role of iNKT cells in shaping the tumor microenvironment, with relevant implications for treatment.
Collapse
Affiliation(s)
- Georgia Lattanzi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-oncology, Università degli Studi di Milano, Milan, Italy
| | - Francesco Strati
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy; Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Angélica Díaz-Basabe
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-oncology, Università degli Studi di Milano, Milan, Italy
| | - Federica Perillo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-oncology, Università degli Studi di Milano, Milan, Italy
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Protti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Maria Rita Giuffrè
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Luca Iachini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Alberto Baeri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Ludovica Baldari
- General and Emergency Surgery Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Cassinotti
- General and Emergency Surgery Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Michele Ghidini
- Medical Oncology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Galassi
- Medical Oncology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianluca Lopez
- Pathology Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniele Noviello
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Laura Porretti
- Clinical Chemistry and Microbiology Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Trombetta
- Clinical Chemistry and Microbiology Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Eleonora Messuti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Luca Mazzarella
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giandomenica Iezzi
- Department of Visceral Surgery, EOC Translational Research Laboratory, Bellinzona, Switzerland
| | - Francesco Nicassio
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Francesca Granucci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy; Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.
| |
Collapse
|
13
|
Sultanov RM, Poleshchuk TS, Ermolenko EV, Kasyanov SP. Protective Properties of Marine Alkyl Glycerol Ethers in Chronic Stress. Mar Drugs 2023; 21:md21040202. [PMID: 37103343 PMCID: PMC10145234 DOI: 10.3390/md21040202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 04/28/2023] Open
Abstract
In this paper we discuss the effect of alkyl glycerol ethers (AGs) from the squid Berryteuthis magister on a chronic stress model in rats. The study was performed on 32 male Wistar rats. Animals received AGs at a dose of 200 mg/kg through a gavage for six weeks (1.5 months), and were divided into four groups: group 1 (control), group 2 (animals received AGs), group 3 (stress control), group 4 (animals received AGs and were subjected to stress). Chronic immobilization stress was induced by placing each rat into an individual plexiglass cages for 2 h daily for 15 days. The serum lipid spectrum was evaluated by the content of total cholesterol, triglycerides, high-density lipoprotein cholesterol, low lipoprotein cholesterol and very low-density lipoprotein cholesterol. The atherogenic coefficient was calculated. The hematological parameters of peripheral blood were evaluated. The neutrophil-lymphocyte ratio was counted. The levels of cortisol and testosterone in blood plasma were determined. AGs at the selected dose did not have a significant effect on the body weight of rats in the preliminary period of the experiment. Under stress, the body weight gain, the concentrations of very low-density lipoprotein cholesterol and blood triglycerides decreased significantly. The neutrophil-lymphocyte ratio in animals treated with AGs shifted towards lymphocytes. A favorable increase in the percentage of lymphocytes was found in the stressed group of animals treated with AGs. So, for the first time, it was found that AGs prevent stress-induced suppression of the immune system. This confirms the benefit of AGs for the immune system under chronic stress. Our results prove the efficiency of the use of AGs for treating chronic stress, a serious social problem in modern society.
Collapse
Affiliation(s)
- Ruslan M Sultanov
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 17 Palchevskogo Str., Vladivostok 690041, Russia
| | - Tatiana S Poleshchuk
- Faculty of Pediatrics and Pharmacy, Pacific State Medical University, 2 Ostryakova Ave., Vladivostok 690002, Russia
| | - Ekaterina V Ermolenko
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 17 Palchevskogo Str., Vladivostok 690041, Russia
| | - Sergey P Kasyanov
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 17 Palchevskogo Str., Vladivostok 690041, Russia
| |
Collapse
|
14
|
Qin Y, Bao X, Zheng M. CD8 + T-cell immunity orchestrated by iNKT cells. Front Immunol 2023; 13:1109347. [PMID: 36741397 PMCID: PMC9889858 DOI: 10.3389/fimmu.2022.1109347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
CD8+ T cells belonging to the adaptive immune system play key roles in defending against viral infections and cancers. The current CD8+ T cell-based immunotherapy has emerged as a superior therapeutic avenue for the eradication of tumor cells and long-term prevention of their recurrence in hematologic malignancies. It is believed that an effective adaptive immune response critically relies on the help of the innate compartment. Invariant natural killer T (iNKT) cells are innate-like T lymphocytes that have been considered some of the first cells to respond to infections and can secrete a large amount of diverse cytokines and chemokines to widely modulate the innate and adaptive immune responders. Like CD8+ T cells, iNKT cells also play an important role in defense against intracellular pathogenic infections and cancers. In this review, we will discuss the CD8+ T-cell immunity contributed by iNKT cells, including iNKT cell-mediated cross-priming and memory formation, and discuss recent advances in our understanding of the mechanisms underlying memory CD8+ T-cell differentiation, as well as aging-induced impairment of T-cell immunity.
Collapse
|
15
|
Ueki K, Sueyoshi K, Inuki S, Fujimoto Y. Chemical Synthesis and Molecular Interaction Analysis of α-Galactosyl Ceramide Derivatives as CD1d Ligands. Methods Mol Biol 2023; 2613:13-22. [PMID: 36587067 DOI: 10.1007/978-1-0716-2910-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CD1d is a non-classical major histocompatibility complex (MHC) protein, responsible for lipid antigen presentation, which presents lipids to natural killer T (NKT) cells. Various CD1d lipid ligands have been reported, including microbial and endogenous glycolipids/phospholipids. Among them, an α-galactosylceramide (α-GalCer), a representative CD1d ligand, is one of the most potent ligands and its derivatives have been developed. In this chapter, the chemistry of α-GalCer and its derivatives are described with an emphasis on their chemical syntheses and molecular interaction analysis with CD1d are described.
Collapse
Affiliation(s)
- Kazunari Ueki
- Department of Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Kodai Sueyoshi
- Department of Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yukari Fujimoto
- Department of Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan.
| |
Collapse
|
16
|
Kocherlakota S, Swinkels D, Van Veldhoven PP, Baes M. Mouse Models to Study Peroxisomal Functions and Disorders: Overview, Caveats, and Recommendations. Methods Mol Biol 2023; 2643:469-500. [PMID: 36952207 DOI: 10.1007/978-1-0716-3048-8_34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
During the last three decades many mouse lines were created or identified that are deficient in one or more peroxisomal functions. Different methodologies were applied to obtain global, hypomorph, cell type selective, inducible, and knockin mice. Whereas some models closely mimic pathologies in patients, others strongly deviate or no human counterpart has been reported. Often, mice, apparently endowed with a stronger transcriptional adaptation, have to be challenged with dietary additions or restrictions in order to trigger phenotypic changes. Depending on the inactivated peroxisomal protein, several approaches can be taken to validate the loss-of-function. Here, an overview is given of the available mouse models and their most important characteristics.
Collapse
Affiliation(s)
- Sai Kocherlakota
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniëlle Swinkels
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Paul P Van Veldhoven
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
17
|
Di Cara F, Savary S, Kovacs WJ, Kim P, Rachubinski RA. The peroxisome: an up-and-coming organelle in immunometabolism. Trends Cell Biol 2023; 33:70-86. [PMID: 35788297 DOI: 10.1016/j.tcb.2022.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 12/27/2022]
Abstract
Peroxisomes are essential metabolic organelles, well known for their roles in the metabolism of complex lipids and reactive ionic species. In the past 10 years, peroxisomes have also been cast as central regulators of immunity. Lipid metabolites of peroxisomes, such as polyunsaturated fatty acids (PUFAs), are precursors for important immune mediators, including leukotrienes (LTs) and resolvins. Peroxisomal redox metabolism modulates cellular immune signaling such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. Additionally, peroxisomal β-oxidation and ether lipid synthesis control the development and aspects of the activation of both innate and adaptive immune cells. Finally, peroxisome number and metabolic activity have been linked to inflammatory diseases. These discoveries have opened avenues of investigation aimed at targeting peroxisomes for therapeutic intervention in immune disorders, inflammation, and cancer.
Collapse
Affiliation(s)
- Francesca Di Cara
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada.
| | - Stéphane Savary
- Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Werner J Kovacs
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology in Zurich (ETH Zürich), Zurich, Switzerland
| | - Peter Kim
- Cell Biology Program, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | | |
Collapse
|
18
|
Wanders RJA, Baes M, Ribeiro D, Ferdinandusse S, Waterham HR. The physiological functions of human peroxisomes. Physiol Rev 2023; 103:957-1024. [PMID: 35951481 DOI: 10.1152/physrev.00051.2021] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peroxisomes are subcellular organelles that play a central role in human physiology by catalyzing a range of unique metabolic functions. The importance of peroxisomes for human health is exemplified by the existence of a group of usually severe diseases caused by an impairment in one or more peroxisomal functions. Among others these include the Zellweger spectrum disorders, X-linked adrenoleukodystrophy, and Refsum disease. To fulfill their role in metabolism, peroxisomes require continued interaction with other subcellular organelles including lipid droplets, lysosomes, the endoplasmic reticulum, and mitochondria. In recent years it has become clear that the metabolic alliance between peroxisomes and other organelles requires the active participation of tethering proteins to bring the organelles physically closer together, thereby achieving efficient transfer of metabolites. This review intends to describe the current state of knowledge about the metabolic role of peroxisomes in humans, with particular emphasis on the metabolic partnership between peroxisomes and other organelles and the consequences of genetic defects in these processes. We also describe the biogenesis of peroxisomes and the consequences of the multiple genetic defects therein. In addition, we discuss the functional role of peroxisomes in different organs and tissues and include relevant information derived from model systems, notably peroxisomal mouse models. Finally, we pay particular attention to a hitherto underrated role of peroxisomes in viral infections.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniela Ribeiro
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Alkyl Glycerol Ethers as Adaptogens. Mar Drugs 2022; 21:md21010004. [PMID: 36662177 PMCID: PMC9862039 DOI: 10.3390/md21010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Τhis mini-review summarizes the hematopoietic and immunostimulating properties of alkyl glycerol ethers (AGs) reported earlier in the literature available to us. The role of AGs in the nervous system and aging of the body are also briefly described. We made an attempt to consider the data in terms of adaptation. The hematopoietic, immunostimulating and antioxidant properties of AGs in a variety of experimental situations, including stress, as well as the protective action of AGs against some adaptation diseases, allow us to consider them as substances that prevent some negative effects of stress and promote adaptation. The new approach to AGs as adaptogens seems promising and opens good opportunities for their new application.
Collapse
|
20
|
Tremblay MÈ, Almsherqi ZA, Deng Y. Plasmalogens and platelet-activating factor roles in chronic inflammatory diseases. Biofactors 2022; 48:1203-1216. [PMID: 36370412 DOI: 10.1002/biof.1916] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022]
Abstract
Fatty acids and phospholipid molecules are essential for determining the structure and function of cell membranes, and they hence participate in many biological processes. Platelet activating factor (PAF) and its precursor plasmalogen, which represent two subclasses of ether phospholipids, have attracted increasing research attention recently due to their association with multiple chronic inflammatory, neurodegenerative, and metabolic disorders. These pathophysiological conditions commonly involve inflammatory processes linked to an excess presence of PAF and/or decreased levels of plasmalogens. However, the molecular mechanisms underlying the roles of plasmalogens in inflammation have remained largely elusive. While anti-inflammatory responses most likely involve the plasmalogen signal pathway; pro-inflammatory responses recruit arachidonic acid, a precursor of pro-inflammatory lipid mediators which is released from membrane phospholipids, notably derived from the hydrolysis of plasmalogens. Plasmalogens per se are vital membrane phospholipids in humans. Changes in their homeostatic levels may alter cell membrane properties, thus affecting key signaling pathways that mediate inflammatory cascades and immune responses. The plasmalogen analogs of PAF are also potentially important, considering that anti-PAF activity has strong anti-inflammatory effects. Plasmalogen replacement therapy was further identified as a promising anti-inflammatory strategy allowing for the relief of pathological hallmarks in patients affected by chronic diseases with an inflammatory component. The aim of this Short Review is to highlight the emerging roles and implications of plasmalogens in chronic inflammatory disorders, along with the promising outcomes of plasmalogen replacement therapy for the treatment of various PAF-related chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, Canada
- Department of Molecular Medicine, Université de Laval, Québec City, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
| | - Zakaria A Almsherqi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| |
Collapse
|
21
|
Intermittent calorie restriction alters T cell subsets and metabolic markers in people with multiple sclerosis. EBioMedicine 2022; 82:104124. [PMID: 35816900 PMCID: PMC9283513 DOI: 10.1016/j.ebiom.2022.104124] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 11/22/2022] Open
|
22
|
Oh SF, Jung DJ, Choi E. Gut Microbiota-Derived Unconventional T Cell Ligands: Contribution to Host Immune Modulation. Immunohorizons 2022; 6:476-487. [PMID: 35868838 PMCID: PMC9924074 DOI: 10.4049/immunohorizons.2200006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/28/2022] [Indexed: 01/26/2023] Open
Abstract
Besides the prototypic innate and adaptive pathways, immune responses by innate-like lymphocytes have gained significant attention due to their unique roles. Among innate-like lymphocytes, unconventional T cells such as NKT cells and mucosal-associated invariant T (MAIT) cells recognize small nonpeptide molecules of specific chemical classes. Endogenous or microbial ligands are loaded to MHC class I-like molecule CD1d or MR1, and inducing immediate effector T cell and ligand structure is one of the key determinants of NKT/MAIT cell functions. Unconventional T cells are in close, constant contact with symbiotic microbes at the mucosal layer, and CD1d/MR1 can accommodate diverse metabolites produced by gut microbiota. There is a strong interest to identify novel immunoactive molecules of endobiotic (symbiont-produced) origin as new NKT/MAIT cell ligands, as well as new cognate Ags for previously uncharacterized unconventional T cell subsets. Further studies will open an possibility to explore basic biology as well as therapeutic potential.
Collapse
Affiliation(s)
- Sungwhan F. Oh
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Da-Jung Jung
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Eungyo Choi
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
23
|
Hossain MS, Mawatari S, Fujino T. Plasmalogen-Mediated Activation of GPCR21 Regulates Cytolytic Activity of NK Cells against the Target Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:310-325. [PMID: 35777853 DOI: 10.4049/jimmunol.2200183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
It is widely known that the immune system becomes slower to respond among elderly people, making them more susceptible to viral infection and cancer. The mechanism of aging-related immune deficiency remained mostly elusive. In this article, we report that plasmalogens (Pls), special phospholipids found to be reduced among the elderly population, critically control cytolytic activity of human NK cells, which is associated with activation of a cell surface receptor, G protein-coupled receptor 21 (GPCR21). We found the extracellular glycosylation site of GPCR21, which is conserved among the mammalian species, to be critically important for the activation of NK cells by Pls. The Pls-GPCR21 signaling cascade induces the expression of Perforin-1, a cytolytic pore-forming protein, via activation of STAT5 transcription factor. Inhibition of STAT5 abrogates GPCR21-mediated cytolytic activation of NK cells against the target cancer cells. In addition, oral ingestion of Pls inhibited cancer growth in SCID mice and inhibited the systemic spread of murine CMV in adult C57BL/6J mice. These findings advocate that Pls-GPCR21 signaling could be critical in maintaining NK cell function, and that the age-related reduction of this signaling cascade could be one of the factors behind immune deficiency in mammals, including humans.
Collapse
Affiliation(s)
- Md Shamim Hossain
- Institute of Rheological Functions of Food, Kasuya-gun, Fukuoka, Japan
| | - Shiro Mawatari
- Institute of Rheological Functions of Food, Kasuya-gun, Fukuoka, Japan
| | - Takehiko Fujino
- Institute of Rheological Functions of Food, Kasuya-gun, Fukuoka, Japan
| |
Collapse
|
24
|
Ding R, Ning S, Yang X, Shi J, Zhao S, Zhang A, Gao X, Tian J, Zhang B, Qin X. Brain and testicular metabonomics revealed the protective effects of Guilingji on senile sexual dysfunction rats. JOURNAL OF ETHNOPHARMACOLOGY 2022; 290:115047. [PMID: 35122976 DOI: 10.1016/j.jep.2022.115047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Guilingji (GLJ), which has been used to treat male diseases in China for centuries, contains 28 Chinese herbs and was previously established as an effective treatment for male sexual dysfunction. However, its mechanism of action remains unclear. AIM OF THE STUDY To explore the efficacy and mechanism of action of GLJ in improving senile sexual dysfunction (SSD) in aging rats. MATERIALS AND METHODS An aging rat model of SSD was induced by the subcutaneous injection of d-galactose (300 mg⋅kg-1) and used to analyse the effects of GLJ (different concentrations of 37.5, 75, and 150 mg⋅kg-1) on the mating of aging rats. At the end of the 8th week, histopathological analysis of testicular tissues, assessment of the hypothalamic-pituitary-gonadal (HPG) axis hormone levels in serum or brain, and metabonomics analysis of the brain and testicular tissue with liquid chromatography-mass spectrometry was performed to explore the mechanism of action of GLJ. RESULT After treatment with GLJ, the mount and ejaculation latency levels were increased in the treatment group than those in model group (P < 0.05), moreover, the testicular morphology was improved. Gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) levels in rats were also improved significant (P < 0.05) compared with those in the model group. Furthermore, the metabonomics results in the testicular and brain tissue showed that GLJ improved SSD by adjusting amino acid and lipid metabolism. CONCLUSION This study integrated the complementary metabolic profiles of the target tissues. GLJ might affect SSD rats by regulating amino acid and lipid metabolism and may modulate sensitivity to the signaling pathway in the HPG axis. This study provides an essential basis for the broad clinical application of GLJ.
Collapse
Affiliation(s)
- Renjie Ding
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Suyun Ning
- Shanxi Pharmaceutical Vocational College, No.16 Minhang South Road, Taiyuan, 030031, Shanxi, PR China
| | - Xiaoling Yang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Jingchao Shi
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; School of Traditional Chinese Materia Medica and Food Engineering, Shanxi University of Chinese Medicine, 030619, Jinzhong, PR China
| | - Sijun Zhao
- Inspection and Testing Center of Shanxi Province, No.106 Changzhi Road, Taiyuan, 030006, Shanxi, PR China
| | - Airong Zhang
- Shanxi Guangyuyuan Traditional Chinese Medicine Co., Ltd, No.1, Guangyuyuan Road, Jinzhong, 030800, Shanxi, PR China
| | - Xiaoxia Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| | - Junsheng Tian
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Bin Zhang
- Shanxi Guangyuyuan Traditional Chinese Medicine Co., Ltd, No.1, Guangyuyuan Road, Jinzhong, 030800, Shanxi, PR China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| |
Collapse
|
25
|
Meghnem D, Leong E, Pinelli M, Marshall JS, Di Cara F. Peroxisomes Regulate Cellular Free Fatty Acids to Modulate Mast Cell TLR2, TLR4, and IgE-Mediated Activation. Front Cell Dev Biol 2022; 10:856243. [PMID: 35756999 PMCID: PMC9215104 DOI: 10.3389/fcell.2022.856243] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Mast cells are specialized, tissue resident, immune effector cells able to respond to a wide range of stimuli. MCs are involved in the regulation of a variety of physiological functions, including vasodilation, angiogenesis and pathogen elimination. In addition, MCs recruit and regulate the functions of many immune cells such as dendritic cells, macrophages, T cells, B cells and eosinophils through their selective production of multiple cytokines and chemokines. MCs generate and release multi-potent molecules, such as histamine, proteases, prostanoids, leukotrienes, heparin, and many cytokines, chemokines, and growth factors through both degranulation dependent and independent pathways. Recent studies suggested that metabolic shifts dictate the activation and granule content secretion by MCs, however the metabolic signaling promoting these events is at its infancy. Lipid metabolism is recognized as a pivotal immunometabolic regulator during immune cell activation. Peroxisomes are organelles found across all eukaryotes, with a pivotal role in lipid metabolism and the detoxification of reactive oxygen species. Peroxisomes are one of the emerging axes in immunometabolism. Here we identified the peroxisome as an essential player in MCs activation. We determined that lack of functional peroxisomes in murine MCs causes a significant reduction of interleukin-6, Tumor necrosis factor and InterleukinL-13 following immunoglobulin IgE-mediated and Toll like receptor 2 and 4 activation compared to the Wild type (WT) BMMCs. We linked these defects in cytokine release to defects in free fatty acids homeostasis. In conclusion, our study identified the importance of peroxisomal fatty acids homeostasis in regulating mast cell-mediated immune functions.
Collapse
Affiliation(s)
- Dihia Meghnem
- Dalhousie Human Immunology and Inflammation Group, Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pediatrics, Nova Scotia Health Authority IWK, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Edwin Leong
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Marinella Pinelli
- Department of Pediatrics, Nova Scotia Health Authority IWK, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jean S. Marshall
- Dalhousie Human Immunology and Inflammation Group, Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- *Correspondence: Jean S. Marshall, ; Francesca Di Cara,
| | - Francesca Di Cara
- Department of Pediatrics, Nova Scotia Health Authority IWK, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- *Correspondence: Jean S. Marshall, ; Francesca Di Cara,
| |
Collapse
|
26
|
Burrello C, Strati F, Lattanzi G, Diaz-Basabe A, Mileti E, Giuffrè MR, Lopez G, Cribiù FM, Trombetta E, Kallikourdis M, Cremonesi M, Conforti F, Botti F, Porretti L, Rescigno M, Vecchi M, Fantini MC, Caprioli F, Facciotti F. IL10 Secretion Endows Intestinal Human iNKT Cells with Regulatory Functions Towards Pathogenic T Lymphocytes. J Crohns Colitis 2022; 16:1461-1474. [PMID: 35358301 PMCID: PMC9455792 DOI: 10.1093/ecco-jcc/jjac049] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Invariant natural killer T [iNKT] cells perform pleiotropic functions in different tissues by secreting a vast array of pro-inflammatory and cytotoxic molecules. However, the presence and function of human intestinal iNKT cells capable of secreting immunomodulatory molecules such as IL-10 has never been reported so far. Here we describe for the first time the presence of IL10-producing iNKT cells [NKT10 cells] in the intestinal lamina propria of healthy individuals and of Crohn's disease [CD] patients. METHODS Frequency and phenotype of NKT10 cells were analysed ex vivo from intestinal specimens of Crohn's disease [n = 17] and controls [n = 7]. Stable CD-derived intestinal NKT10 cell lines were used to perform in vitro suppression assays and co-cultures with patient-derived mucosa-associated microbiota. Experimental colitis models were performed by adoptive cell transfer of splenic naïve CD4+ T cells in the presence or absence of IL10-sufficient or -deficient iNKT cells. In vivo induction of NKT10 cells was performed by administration of short chain fatty acids [SCFA] by oral gavage. RESULTS Patient-derived intestinal NKT10 cells demonstrated suppressive capabilities towards pathogenic CD4+ T cells. The presence of increased proportions of mucosal NKT10 cells associated with better clinical outcomes in CD patients. Moreover, an intestinal microbial community enriched in SCFA-producing bacteria sustained the production of IL10 by iNKT cells. Finally, IL10-deficient iNKT cells failed to control the pathogenic activity of adoptively transferred CD4+ T cells in an experimental colitis model. CONCLUSIONS These results describe an unprecedentd IL10-mediated immunoregulatory role of intestinal iNKT cells in controlling the pathogenic functions of mucosal T helper subsets and in maintaining the intestinal immune homeostasis.
Collapse
Affiliation(s)
- Claudia Burrello
- Current address: Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | - Erika Mileti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Rita Giuffrè
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Gianluca Lopez
- Pathology Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Fulvia Milena Cribiù
- Pathology Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Trombetta
- Clinical Chemistry and Microbiology Laboratory Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Laboratory of Adaptive Immunity, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Marco Cremonesi
- Laboratory of Adaptive Immunity, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Francesco Conforti
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Fiorenzo Botti
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- General and Emergency Surgery Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Porretti
- Clinical Chemistry and Microbiology Laboratory Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Rescigno
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Massimo C Fantini
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Federica Facciotti
- Corresponding author: Dr Federica Facciotti, Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20135, Milan, Italy.
| |
Collapse
|
27
|
Nath AS, Parsons BD, Makdissi S, Chilvers RL, Mu Y, Weaver CM, Euodia I, Fitze KA, Long J, Scur M, Mackenzie DP, Makrigiannis AP, Pichaud N, Boudreau LH, Simmonds AJ, Webber CA, Derfalvi B, Hammon Y, Rachubinski RA, Di Cara F. Modulation of the cell membrane lipid milieu by peroxisomal β-oxidation induces Rho1 signaling to trigger inflammatory responses. Cell Rep 2022; 38:110433. [PMID: 35235794 DOI: 10.1016/j.celrep.2022.110433] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/21/2021] [Accepted: 02/01/2022] [Indexed: 12/11/2022] Open
Abstract
Phagocytosis, signal transduction, and inflammatory responses require changes in lipid metabolism. Peroxisomes have key roles in fatty acid homeostasis and in regulating immune function. We find that Drosophila macrophages lacking peroxisomes have perturbed lipid profiles, which reduce host survival after infection. Using lipidomic, transcriptomic, and genetic screens, we determine that peroxisomes contribute to the cell membrane glycerophospholipid composition necessary to induce Rho1-dependent signals, which drive cytoskeletal remodeling during macrophage activation. Loss of peroxisome function increases membrane phosphatidic acid (PA) and recruits RhoGAPp190 during infection, inhibiting Rho1-mediated responses. Peroxisome-glycerophospholipid-Rho1 signaling also controls cytoskeleton remodeling in mouse immune cells. While high levels of PA in cells without peroxisomes inhibit inflammatory phenotypes, large numbers of peroxisomes and low amounts of cell membrane PA are features of immune cells from patients with inflammatory Kawasaki disease and juvenile idiopathic arthritis. Our findings reveal potential metabolic markers and therapeutic targets for immune diseases and metabolic disorders.
Collapse
Affiliation(s)
- Anu S Nath
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Brendon D Parsons
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Stephanie Makdissi
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Rebecca L Chilvers
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Yizhu Mu
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Ceileigh M Weaver
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Irene Euodia
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Katherine A Fitze
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Juyang Long
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Michal Scur
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Duncan P Mackenzie
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Andrew P Makrigiannis
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Nicolas Pichaud
- Université de Moncton, Department of Chemistry and Biochemistry, Moncton, NB E1A 3E9, Canada; New Brunswick Centre for Precision Medicine (NBCPM), Moncton, NB E1A 3E9, Canada
| | - Luc H Boudreau
- Université de Moncton, Department of Chemistry and Biochemistry, Moncton, NB E1A 3E9, Canada; New Brunswick Centre for Precision Medicine (NBCPM), Moncton, NB E1A 3E9, Canada
| | - Andrew J Simmonds
- University of Alberta, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Christine A Webber
- University of Alberta, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Beata Derfalvi
- Dalhousie University, Department of Pediatrics, Halifax, NS B3K 6R8, Canada
| | - Yannick Hammon
- INSERM au Centre d'Immunologie de Marseille Luminy, Marseille 13288, France
| | | | - Francesca Di Cara
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada; Dalhousie University, Department of Pediatrics, Halifax, NS B3K 6R8, Canada.
| |
Collapse
|
28
|
Mice with a deficiency in Peroxisomal Membrane Protein 4 (PXMP4) display mild changes in hepatic lipid metabolism. Sci Rep 2022; 12:2512. [PMID: 35169201 PMCID: PMC8847483 DOI: 10.1038/s41598-022-06479-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/31/2022] [Indexed: 11/08/2022] Open
Abstract
Peroxisomes play an important role in the metabolism of a variety of biomolecules, including lipids and bile acids. Peroxisomal Membrane Protein 4 (PXMP4) is a ubiquitously expressed peroxisomal membrane protein that is transcriptionally regulated by peroxisome proliferator-activated receptor α (PPARα), but its function is still unknown. To investigate the physiological function of PXMP4, we generated a Pxmp4 knockout (Pxmp4-/-) mouse model using CRISPR/Cas9-mediated gene editing. Peroxisome function was studied under standard chow-fed conditions and after stimulation of peroxisomal activity using the PPARα ligand fenofibrate or by using phytol, a metabolite of chlorophyll that undergoes peroxisomal oxidation. Pxmp4-/- mice were viable, fertile, and displayed no changes in peroxisome numbers or morphology under standard conditions. Also, no differences were observed in the plasma levels of products from major peroxisomal pathways, including very long-chain fatty acids (VLCFAs), bile acids (BAs), and BA intermediates di- and trihydroxycholestanoic acid. Although elevated levels of the phytol metabolites phytanic and pristanic acid in Pxmp4-/- mice pointed towards an impairment in peroxisomal α-oxidation capacity, treatment of Pxmp4-/- mice with a phytol-enriched diet did not further increase phytanic/pristanic acid levels. Finally, lipidomic analysis revealed that loss of Pxmp4 decreased hepatic levels of the alkyldiacylglycerol class of neutral ether lipids, particularly those containing polyunsaturated fatty acids. Together, our data show that while PXMP4 is not critical for overall peroxisome function under the conditions tested, it may have a role in the metabolism of (ether)lipids.
Collapse
|
29
|
Yin J, Wang H, Hong Y, Ren A, Wang H, Liu L, Zhao Q. Identification of an at-risk subpopulation with high immune infiltration based on the peroxisome pathway and TIM3 in colorectal cancer. BMC Cancer 2022; 22:44. [PMID: 34996408 PMCID: PMC8739708 DOI: 10.1186/s12885-021-09085-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 11/29/2021] [Indexed: 12/17/2022] Open
Abstract
Background Peroxisomes are pivotal metabolic organelles that exist in almost all eukaryote cells. A reduction in numbers and enzymatic activities of peroxisomes was found in colon adenocarcinomas. However, the role of peroxisomes or the peroxisome pathway in colorectal cancer (CRC) is not defined. Methods In the current study, a peroxisome score was calculated to indicate the activity of the peroxisome pathway using gene set variant analysis based on transcriptomic datasets. CIBERSORTx was chosen to infer enriched immune cells for tumors among subgroups. The SubMap algorithm was applied to predict its sensitivity to immunotherapy. Results The patients with a relatively low peroxisome score and high level of T-cell immunoglobulin and mucin domain 3 (TIM-3) presented the worse overall survival than others. Moreover, low peroxisome scores were associated with high infiltration of lymphocytes and poor prognosis in those CRC patients. Thus, a PERLowTIM3High CRC risk subpopulation was identified and characterized by high immune infiltration. The results also showed that CD8 T cells and macrophages highly infiltrated tumors of the PERLowTIM3High group, regardless of consortium molecular subtype and microsatellite instability status. This subgroup had the highest tumor mutational burden and overexpression of immune checkpoint genes. Further, the PERLowTIM3High group showed a higher probability of responding to programmed cell death protein-1-based immunotherapy. In addition, genes involved in peroxisomal metabolic processes in CRC were also investigated since peroxisome is a rather pleiotropic and highly metabolic organelle in cell. The results indicated that only those genes involved in fatty acid alpha oxidation could be used to stratify CRC patients as similar as peroxisome pathway genes. Conclusions We revealed the favorable prognostic value of the peroxisome pathway in CRC and provided a new CRC stratification based on peroxisomes and TIM3, which might be helpful for CRC diagnostics and personalized treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09085-9.
Collapse
Affiliation(s)
- Jinwen Yin
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000, China
| | - Hao Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000, China
| | - Yuntian Hong
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, China.,Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430000, China
| | - Anli Ren
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000, China
| | - Haizhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, China. .,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000, China.
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, China. .,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430000, China.
| |
Collapse
|
30
|
Bharadwaj NS, Gumperz JE. Harnessing invariant natural killer T cells to control pathological inflammation. Front Immunol 2022; 13:998378. [PMID: 36189224 PMCID: PMC9519390 DOI: 10.3389/fimmu.2022.998378] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are innate T cells that are recognized for their potent immune modulatory functions. Over the last three decades, research in murine models and human observational studies have revealed that iNKT cells can act to limit inflammatory pathology in a variety of settings. Since iNKT cells are multi-functional and can promote inflammation in some contexts, understanding the mechanistic basis for their anti-inflammatory effects is critical for effectively harnessing them for clinical use. Two contrasting mechanisms have emerged to explain the anti-inflammatory activity of iNKT cells: that they drive suppressive pathways mediated by other regulatory cells, and that they may cytolytically eliminate antigen presenting cells that promote excessive inflammatory responses. How these activities are controlled and separated from their pro-inflammatory functions remains a central question. Murine iNKT cells can be divided into four functional lineages that have either pro-inflammatory (NKT1, NKT17) or anti-inflammatory (NKT2, NKT10) cytokine profiles. However, in humans these subsets are not clearly evident, and instead most iNKT cells that are CD4+ appear oriented towards polyfunctional (TH0) cytokine production, while CD4- iNKT cells appear more predisposed towards cytolytic activity. Additionally, structurally distinct antigens have been shown to induce TH1- or TH2-biased responses by iNKT cells in murine models, but human iNKT cells may respond to differing levels of TCR stimulation in a way that does not neatly separate TH1 and TH2 cytokine production. We discuss the implications of these differences for translational efforts focused on the anti-inflammatory activity of iNKT cells.
Collapse
Affiliation(s)
- Nikhila S Bharadwaj
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Jenny E Gumperz
- Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
31
|
Development of αβ T Cells with Innate Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:149-160. [DOI: 10.1007/978-981-16-8387-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
32
|
Díaz‐Basabe A, Burrello C, Lattanzi G, Botti F, Carrara A, Cassinotti E, Caprioli F, Facciotti F. Human intestinal and circulating invariant natural killer T cells are cytotoxic against colorectal cancer cells via the perforin-granzyme pathway. Mol Oncol 2021; 15:3385-3403. [PMID: 34535957 PMCID: PMC8637555 DOI: 10.1002/1878-0261.13104] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/01/2021] [Accepted: 09/16/2021] [Indexed: 11/05/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are lipid-specific T lymphocytes endowed with cytotoxic activities and are thus considered important in antitumor immunity. While several studies have demonstrated iNKT cell cytotoxicity against different tumors, very little is known about their cell-killing activities in human colorectal cancer (CRC). Our aim was to assess whether human iNKT cells are cytotoxic against colon cancer cells and the mechanisms underlying this activity. For this purpose, we generated stable iNKT cell lines from peripheral blood and colon specimens and used NK-92 and peripheral blood natural killer cells as cell-mediated cytotoxicity controls. In vitro cytotoxicity was assessed using a panel of well-characterized human CRC cell lines, and the cellular requirements for iNKT cell cytotoxic functions were evaluated. We demonstrated that both intestinal and circulating iNKT cells were cytotoxic against the entire panel of CRC lines, as well as against freshly isolated patient-derived colonic epithelial cancer cells. Perforin and/or granzyme inhibition impaired iNKT cell cytotoxicity, whereas T-cell receptor (TCR) signaling was a less stringent requirement for efficient killing. This study is the first evidence of tissue-derived iNKT cell cytotoxic activity in humans, as it shows that iNKT cells depend on the perforin-granzyme pathway and both adaptive and innate signal recognition for proper elimination of colon cancer cells.
Collapse
Affiliation(s)
- Angélica Díaz‐Basabe
- Department of Experimental OncologyIEO European Institute of Oncology IRCCSMilanItaly
- Department of Oncology and Hemato‐oncologyUniversità degli Studi di MilanoMilanItaly
| | - Claudia Burrello
- Department of Experimental OncologyIEO European Institute of Oncology IRCCSMilanItaly
| | - Georgia Lattanzi
- Department of Experimental OncologyIEO European Institute of Oncology IRCCSMilanItaly
- Department of Oncology and Hemato‐oncologyUniversità degli Studi di MilanoMilanItaly
| | - Fiorenzo Botti
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
- Department of SurgeryFondazione IRCCS Cà GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Alberto Carrara
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
- Department of SurgeryFondazione IRCCS Cà GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Elisa Cassinotti
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Flavio Caprioli
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
- Gastroenterology and Endoscopy UnitFondazione IRCCS Cà GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Federica Facciotti
- Department of Experimental OncologyIEO European Institute of Oncology IRCCSMilanItaly
| |
Collapse
|
33
|
Suarez-Trujillo A, Luecke SM, Logan L, Bradshaw C, Stewart KR, Minor RC, Ramires Ferreira C, Casey TM. Changes in sow milk lipidome across lactation occur in fatty acyl residues of triacylglycerol and phosphatidylglycerol lipids, but not in plasma membrane phospholipids. Animal 2021; 15:100280. [PMID: 34252722 DOI: 10.1016/j.animal.2021.100280] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/26/2022] Open
Abstract
Milk fats are vital to neonate survival and development, but vary highly by diet, maternal metabolic state and stage of lactation. To gain a better understanding of changes in lipid composition of sow milk across lactation, milk was collected from nine multiparous sows on days 0, 3, 7, and 14, relative to birth and lipids were profiled using multiple reaction monitoring (MRM) profiling. Percent fat was determined by creamatocrit, and found to be different (P < 0.05) between day 0 (12.36 ± 5.90%) and day 3 (16.22 ± 3.65%) but not between day 7 (13.13 ± 2.19%) and day 14 (12.13 ± 2.45%). Fat was extracted from milk (n = 6/day) using the Bligh-Dyer method and profiled using tandem mass spectrometry MRM to determine the abundance of lipids defined by class and fatty acyl residue composition. Lipid species relative concentration was calculated from internal standards, and data analysis was performed using Metaboanalyst 4.0. Concentration of phosphatidyl-choline, -serine, -ethanolamine, -inositol, cholesteryl ester and sphingomyelin did not vary across lactation days, nor did the distribution of associated fatty acyl residues. The total abundance of triacylglycerides (TGs) and phosphatidylglycerols (PGs) increased (P < 0.05) from colostrum (day 0) to transitional (days 3 and 7) and mature milk (day 14). As lactation days increased from day 0 to day 14, the number of carbons and unsaturation within fatty acyl residues decreased (P < 0.05) in both TGs and PGs. The proportion of TGs and PGs increased (P < 0.05) relative to other lipid classes. Changes in composition of milk triglycerides and phosphatidylglycerols likely reflect the metabolic activity of the mammary gland and developmental needs of neonates.
Collapse
Affiliation(s)
- A Suarez-Trujillo
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - S M Luecke
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - L Logan
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - C Bradshaw
- Department of Animal Sciences, North Carolina A&T University, Greensboro, NC 27411, United States
| | - K R Stewart
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - R C Minor
- Department of Animal Sciences, North Carolina A&T University, Greensboro, NC 27411, United States
| | - C Ramires Ferreira
- Metabolomics Core, Bindley Science Center, Purdue University, West Lafayette, IN 47907, United States
| | - T M Casey
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
34
|
Paul S, Smith AAT, Culham K, Gunawan KA, Weir JM, Cinel MA, Jayawardana KS, Mellett NA, Lee MKS, Murphy AJ, Lancaster GI, Nestel PJ, Kingwell BA, Meikle PJ. Shark liver oil supplementation enriches endogenous plasmalogens and reduces markers of dyslipidemia and inflammation. J Lipid Res 2021; 62:100092. [PMID: 34146594 PMCID: PMC8281607 DOI: 10.1016/j.jlr.2021.100092] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/27/2021] [Accepted: 06/06/2021] [Indexed: 01/09/2023] Open
Abstract
Plasmalogens are membrane glycerophospholipids with diverse biological functions. Reduced plasmalogen levels have been observed in metabolic diseases; hence, increasing their levels might be beneficial in ameliorating these conditions. Shark liver oil (SLO) is a rich source of alkylglycerols that can be metabolized into plasmalogens. This study was designed to evaluate the impact of SLO supplementation on endogenous plasmalogen levels in individuals with features of metabolic disease. In this randomized, double-blind, placebo-controlled cross-over study, the participants (10 overweight or obese males) received 4-g Alkyrol® (purified SLO) or placebo (methylcellulose) per day for 3 weeks followed by a 3-week washout phase and were then crossed over to 3 weeks of the alternate placebo/Alkyrol® treatment. SLO supplementation led to significant changes in plasma and circulatory white blood cell lipidomes, notably increased levels of plasmalogens and other ether lipids. In addition, SLO supplementation significantly decreased the plasma levels of total free cholesterol, triglycerides, and C-reactive protein. These findings suggest that SLO supplementation can enrich plasma and cellular plasmalogens and this enrichment may provide protection against obesity-related dyslipidemia and inflammation.
Collapse
Affiliation(s)
- Sudip Paul
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Adam Alexander T Smith
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kevin Culham
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kevin A Gunawan
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jacqueline M Weir
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Michelle A Cinel
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kaushala S Jayawardana
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Man K S Lee
- Haematopoiesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andrew J Murphy
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Haematopoiesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Graeme I Lancaster
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia; Haematopoiesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Paul J Nestel
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Bronwyn A Kingwell
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
35
|
Abstract
BACKGROUND Dengue virus causes dengue fever (DF)disease, transmitted by the mosquito Aedes aegypti. The symptoms could be severe and disable the affected individuals for weeks. The severe form, dengue hemorrhagic fever (DHF), can lead to death if not adequately attended to. Due to global warming, the vector mosquito will advance over new areas and expose more people to this disease over the next decades. Despite the severity, there are no treatments nor efficient vaccines available. Metabolomic studies have shown a new perspective to understand this disease better at a new molecular level. AIM OF REVIEW Many published works rely on samples obtained from animal studies. This review will mainly focus on human samples and cell culture experiments to view how the dengue virus affects the metabolomic profile. KEY SCIENTIFIC CONCEPTS OF REVIEW The review compiles the sample sources, metabolomic techniques used, the detected compounds, and how they behave in different DF stages. This disease causes a significant change in many metabolites, but some results are still conflicting between studies. The results gathered here show that metabolomic approaches prove to be an excellent and viable way to expand knowledge about DF.
Collapse
Affiliation(s)
| | - Karina Inacio Carvalho
- Hospital Israelita Albert Einstein, São Paulo, Brazil.
- Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
36
|
Deng Y, Angelova A. Coronavirus-Induced Host Cubic Membranes and Lipid-Related Antiviral Therapies: A Focus on Bioactive Plasmalogens. Front Cell Dev Biol 2021; 9:630242. [PMID: 33791293 PMCID: PMC8006408 DOI: 10.3389/fcell.2021.630242] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
Coronaviruses have lipid envelopes required for their activity. The fact that coronavirus infection provokes the formation of cubic membranes (CM) (denoted also as convoluted membranes) in host cells has not been rationalized in the development of antiviral therapies yet. In this context, the role of bioactive plasmalogens (vinyl ether glycerophospholipids) is not completely understood. These lipid species display a propensity for non-lamellar phase formation, facilitating membrane fusion, and modulate the activity of membrane-bound proteins such as enzymes and receptors. At the organism level, plasmalogen deficiency is associated with cardiometabolic disorders including obesity and type 2 diabetes in humans. A straight link is perceived with the susceptibility of such patients to SARS-CoV-2 (severe acute respiratory syndrome-coronavirus-2) infection, the severity of illness, and the related difficulty in treatment. Based on correlations between the coronavirus-induced modifications of lipid metabolism in host cells, plasmalogen deficiency in the lung surfactant of COVID-19 patients, and the alterations of lipid membrane structural organization and composition including the induction of CM, we emphasize the key role of plasmalogens in the coronavirus (SARS-CoV-2, SARS-CoV, or MERS-CoV) entry and replication in host cells. Considering that plasmalogen-enriched lung surfactant formulations may improve the respiratory process in severe infected individuals, plasmalogens can be suggested as an anti-viral prophylactic, a lipid biomarker in SARS-CoV and SARS-CoV-2 infections, and a potential anti-viral therapeutic component of lung surfactant development for COVID-19 patients.
Collapse
Affiliation(s)
- Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay UMR 8612, Châtenay-Malabry, France
| |
Collapse
|
37
|
Chornyi S, IJlst L, van Roermund CWT, Wanders RJA, Waterham HR. Peroxisomal Metabolite and Cofactor Transport in Humans. Front Cell Dev Biol 2021; 8:613892. [PMID: 33505966 PMCID: PMC7829553 DOI: 10.3389/fcell.2020.613892] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
Peroxisomes are membrane-bound organelles involved in many metabolic pathways and essential for human health. They harbor a large number of enzymes involved in the different pathways, thus requiring transport of substrates, products and cofactors involved across the peroxisomal membrane. Although much progress has been made in understanding the permeability properties of peroxisomes, there are still important gaps in our knowledge about the peroxisomal transport of metabolites and cofactors. In this review, we discuss the different modes of transport of metabolites and essential cofactors, including CoA, NAD+, NADP+, FAD, FMN, ATP, heme, pyridoxal phosphate, and thiamine pyrophosphate across the peroxisomal membrane. This transport can be mediated by non-selective pore-forming proteins, selective transport proteins, membrane contact sites between organelles, and co-import of cofactors with proteins. We also discuss modes of transport mediated by shuttle systems described for NAD+/NADH and NADP+/NADPH. We mainly focus on current knowledge on human peroxisomal metabolite and cofactor transport, but also include knowledge from studies in plants, yeast, fruit fly, zebrafish, and mice, which has been exemplary in understanding peroxisomal transport mechanisms in general.
Collapse
Affiliation(s)
- Serhii Chornyi
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Lodewijk IJlst
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Carlo W T van Roermund
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
38
|
Bioactive Ether Lipids: Primordial Modulators of Cellular Signaling. Metabolites 2021; 11:metabo11010041. [PMID: 33430006 PMCID: PMC7827237 DOI: 10.3390/metabo11010041] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 12/14/2022] Open
Abstract
The primacy of lipids as essential components of cellular membranes is conserved across taxonomic domains. In addition to this crucial role as a semi-permeable barrier, lipids are also increasingly recognized as important signaling molecules with diverse functional mechanisms ranging from cell surface receptor binding to the intracellular regulation of enzymatic cascades. In this review, we focus on ether lipids, an ancient family of lipids having ether-linked structures that chemically differ from their more prevalent acyl relatives. In particular, we examine ether lipid biosynthesis in the peroxisome of mammalian cells, the roles of selected glycerolipids and glycerophospholipids in signal transduction in both prokaryotes and eukaryotes, and finally, the potential therapeutic contributions of synthetic ether lipids to the treatment of cancer.
Collapse
|
39
|
Wang H, Li L, Li Y, Li Y, Sha Y, Wen S, You Q, Liu L, Shi M, Zhou H. Intravital imaging of interactions between iNKT and kupffer cells to clear free lipids during steatohepatitis. Theranostics 2021; 11:2149-2169. [PMID: 33500717 PMCID: PMC7797696 DOI: 10.7150/thno.51369] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/29/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Invariant natural killer T (iNKT) cells and Kupffer cells represent major hepatic populations of innate immune cells. However, their roles in steatohepatitis remain poorly understood. To elucidate their functions in steatohepatitis development, real-time, in vivo analysis is necessary to understand the pathophysiological events in the dynamic interactions between them during diet-induced steatohepatitis. Methods: We used a steatohepatitis animal model induced by a methionine-choline-deficient (MCD) diet. Multi-photon confocal live imaging and conventional experimental techniques were employed to investigate the hepatic pathological microenvironment of iNKT and Kupffer cells, interactions between them, and the biological effects of these interactions in steatohepatitis. Results: We found that iNKT cells were recruited and aggregated into small clusters and interacted dynamically with Kupffer cells in the early stage of steatohepatitis. Most significantly, the iNKT cells in the cluster cleared free lipids released by necrotic hepatocytes and presented a non-classical activation state with high IFN-γ expression. Furthermore, the Kupffer cells in the cell cluster were polarized to type M1. The transcriptome sequencing of iNKT cells showed upregulation of genes related to phagocytosis and lipid processing. Adoptive transfer of iNKT cells to Jα18-/- mice showed that iNKT and Kupffer cell clusters were essential for balancing the liver and peripheral lipid levels and inhibiting liver fibrosis development. Conclusions: Our study identified an essential role for dynamic interactions between iNKT cells and Kupffer cells in promoting lipid phagocytosis and clearance by iNKT cells during early liver steatohepatitis. Therefore, modulating iNKT cells is a potential therapeutic strategy for early steatohepatitis.
Collapse
|
40
|
Dorninger F, Forss-Petter S, Wimmer I, Berger J. Plasmalogens, platelet-activating factor and beyond - Ether lipids in signaling and neurodegeneration. Neurobiol Dis 2020; 145:105061. [PMID: 32861763 PMCID: PMC7116601 DOI: 10.1016/j.nbd.2020.105061] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Glycerol-based ether lipids including ether phospholipids form a specialized branch of lipids that in mammals require peroxisomes for their biosynthesis. They are major components of biological membranes and one particular subgroup, the plasmalogens, is widely regarded as a cellular antioxidant. Their vast potential to influence signal transduction pathways is less well known. Here, we summarize the literature showing associations with essential signaling cascades for a wide variety of ether lipids, including platelet-activating factor, alkylglycerols, ether-linked lysophosphatidic acid and plasmalogen-derived polyunsaturated fatty acids. The available experimental evidence demonstrates links to several common players like protein kinase C, peroxisome proliferator-activated receptors or mitogen-activated protein kinases. Furthermore, ether lipid levels have repeatedly been connected to some of the most abundant neurological diseases, particularly Alzheimer's disease and more recently also neurodevelopmental disorders like autism. Thus, we critically discuss the potential role of these compounds in the etiology and pathophysiology of these diseases with an emphasis on signaling processes. Finally, we review the emerging interest in plasmalogens as treatment target in neurological diseases, assessing available data and highlighting future perspectives. Although many aspects of ether lipid involvement in cellular signaling identified in vitro still have to be confirmed in vivo, the compiled data show many intriguing properties and contributions of these lipids to health and disease that will trigger further research.
Collapse
Affiliation(s)
- Fabian Dorninger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna 1090, Austria.
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna 1090, Austria
| | - Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna 1090, Austria.
| |
Collapse
|
41
|
Sinisalu L, Sen P, Salihović S, Virtanen SM, Hyöty H, Ilonen J, Toppari J, Veijola R, Orešič M, Knip M, Hyötyläinen T. Early-life exposure to perfluorinated alkyl substances modulates lipid metabolism in progression to celiac disease. ENVIRONMENTAL RESEARCH 2020; 188:109864. [PMID: 32846648 DOI: 10.1016/j.envres.2020.109864] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 06/11/2023]
Abstract
Celiac disease (CD) is a systemic immune-mediated disorder with increased frequency in the developed countries over the last decades implicating the potential causal role of various environmental triggers in addition to gluten. Herein, we apply determination of perfluorinated alkyl substances (PFAS) and combine the results with the determination of bile acids (BAs) and molecular lipids, with the aim to elucidate the impact of prenatal exposure on risk of progression to CD in a prospective series of children prior the first exposure to gluten (at birth and at 3 months of age). Here we analyzed PFAS, BAs and lipidomic profiles in 66 plasma samples at birth and at 3 months of age in the Type 1 Diabetes Prediction and Prevention (DIPP) study (n = 17 progressors to CD, n = 16 healthy controls, HCs). Plasma PFAS levels showed a significant inverse association with the age of CD diagnosis in infants who later progressed to the disease. Associations between BAs and triacylglycerols (TGs) showed different patterns already at birth in CD progressors, indicative of different absorption of lipids in these infants. In conclusion, PFAS exposure may modulate lipid and BA metabolism, and the impact is different in the infants who develop CD later in life, in comparison to HCs. The results indicate more efficient uptake of PFAS in such infants. Higher PFAS exposure during prenatal and early life may accelerate the progression to CD in the genetically predisposed children.
Collapse
Affiliation(s)
- Lisanna Sinisalu
- School of Science and Technology, Örebro University, Örebro, Sweden
| | - Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Samira Salihović
- School of Science and Technology, Örebro University, Örebro, Sweden; School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Suvi M Virtanen
- Finnish Institute for Health and Welfare, Public Health Promotion Unit, Helsinki, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland; Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Heikki Hyöty
- Faculty of Medicine Health Technology, Tampere University, Tampere, Finland; Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland; Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Jorma Toppari
- Institute of Biomedicine, Centre for Integrative Physiology and Pharmacology, And Centre for Population Health Research, University of Turku, Turku, Finland; Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Riitta Veijola
- Department of Paediatrics, PEDEGO Research Unit, Medical Research Centre, University of Oulu, Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland; School of Medical Sciences, Örebro University, Örebro, Sweden.
| | - Mikael Knip
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Center for Child Health Research, Tampere University Hospital, Tampere, Finland.
| | | |
Collapse
|
42
|
Affiliation(s)
- Francesca Di Cara
- Department of Microbiology and Immunology-IWK Health Centre- Dalhousie University, Halifax (NS), Canada
| |
Collapse
|
43
|
Thymic development of unconventional T cells: how NKT cells, MAIT cells and γδ T cells emerge. Nat Rev Immunol 2020; 20:756-770. [DOI: 10.1038/s41577-020-0345-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2020] [Indexed: 12/11/2022]
|
44
|
Du K, Gao XX, Feng Y, Li J, Wang H, Lv SL, Wang PY, Zhang B, Qin XM. Integrated adrenal and testicular metabolomics revealed the protective effects of Guilingji on the Kidney-Yang deficiency syndrome rats. JOURNAL OF ETHNOPHARMACOLOGY 2020; 255:112734. [PMID: 32151756 DOI: 10.1016/j.jep.2020.112734] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/08/2020] [Accepted: 03/01/2020] [Indexed: 05/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Guilingji (GLJ) is a well-known traditional Chinese medicine (TCM) prescription for the treatment of Kidney-Yang deficiency syndrome (KYDS). AIM OF THE STUDY This study aimed to address the protective effects of GLJ against KYDS in rats with pharmacodynamic indicators and target tissues (adrenal gland and testis) metabolomics. MATERIALS AND METHODS The rats were injected intraperitoneally (i.p) hydrocortisone to simulate KYDS and administered orally of GLJ for 30 days. Traditional pharmacodynamic indicators (body weight, behavioral indicators, biochemical parameters and histological examination) were performed to evaluate the efficacy of GLJ. Furthermore, adrenal gland and testis metabolic profiles obtained by UHPLC-Q Exactive Orbitrap-MS coupled with multivariate analysis were conducted to explore the metabolic regulation mechanism of GLJ. RESULTS After administration of GLJ, the weight, levels of behavioral indicators and biochemical parameters of rats were increased compared with those of the model group, and the abnormalities of morphology in adrenal and testicular tissues were improved. Furthermore, GLJ had recovering effects via the adjustment of vitamins metabolism, which was accompanied by lipids metabolism, amino acid metabolism and nucleotides metabolism. CONCLUSIONS The study firstly integrated the target tissues metabolic profiles, which were complementary, and GLJ had protective effects on KYDS rats via the regulation of steroid hormone biosynthesis, oxidant-antioxidant balance and energy acquisition.
Collapse
Affiliation(s)
- Ke Du
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, 030006, PR China; College of Chemistry and Chemical Engineering of Shanxi University, Taiyuan, 030006, PR China
| | - Xiao-Xia Gao
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, 030006, PR China.
| | - Yan Feng
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, 030006, PR China; College of Chemistry and Chemical Engineering of Shanxi University, Taiyuan, 030006, PR China
| | - Jing Li
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, 030006, PR China
| | - Hui Wang
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, 030006, PR China
| | - Si-Lin Lv
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, 030006, PR China
| | - Pei-Yi Wang
- Shanxi Guangyuyuan Chinese Medicine Co., Ltd, Jinzhong, 030800, PR China
| | - Bin Zhang
- Shanxi Guangyuyuan Chinese Medicine Co., Ltd, Jinzhong, 030800, PR China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan, 030006, PR China.
| |
Collapse
|
45
|
License to Kill: When iNKT Cells Are Granted the Use of Lethal Cytotoxicity. Int J Mol Sci 2020; 21:ijms21113909. [PMID: 32486268 PMCID: PMC7312231 DOI: 10.3390/ijms21113909] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Invariant Natural Killer T (iNKT) cells are a non-conventional, innate-like, T cell population that recognize lipid antigens presented by the cluster of differentiation (CD)1d molecule. Although iNKT cells are mostly known for mediating several immune responses due to their massive and diverse cytokine release, these cells also work as effectors in various contexts thanks to their cytotoxic potential. In this Review, we focused on iNKT cell cytotoxicity; we provide an overview of iNKT cell subsets, their activation cues, the mechanisms of iNKT cell cytotoxicity, the specific roles and outcomes of this activity in various contexts, and how iNKT killing functions are currently activated in cancer immunotherapies. Finally, we discuss the future perspectives for the better understanding and potential uses of iNKT cell killing functions in tumor immunosurveillance.
Collapse
|
46
|
Shimizu K, Iyoda T, Yamasaki S, Kadowaki N, Tojo A, Fujii SI. NK and NKT Cell-Mediated Immune Surveillance against Hematological Malignancies. Cancers (Basel) 2020; 12:cancers12040817. [PMID: 32231116 PMCID: PMC7226455 DOI: 10.3390/cancers12040817] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Recent cancer treatment modalities have been intensively focused on immunotherapy. The success of chimeric antigen receptor T cell therapy for treatment of refractory B cell acute lymphoblastic leukemia has pushed forward research on hematological malignancies. Among the effector types of innate lymphocytes, natural killer (NK) cells show great importance in immune surveillance against infectious and tumor diseases. Particularly, the role of NK cells has been argued in either elimination of target tumor cells or escape of tumor cells from immune surveillance. Therefore, an NK cell activation approach has been explored. Recent findings demonstrate that invariant natural killer T (iNKT) cells capable of producing IFN-γ when optimally activated can promptly trigger NK cells. Here, we review the role of NKT and/or NK cells and their interaction in anti-tumor responses by highlighting how innate immune cells recognize tumors, exert effector functions, and amplify adaptive immune responses. In addition, we discuss these innate lymphocytes in hematological disorders, particularly multiple myeloma and acute myeloid leukemia. The immune balance at different stages of both diseases is explored in light of disease progression. Various types of innate immunity-mediated therapeutic approaches, recent advances in clinical immunotherapies, and iNKT-mediated cancer immunotherapy as next-generation immunotherapy are then discussed.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
- Correspondence: (K.S.); (S.-i.F.); Tel.: +81-45-503-7062 (K.S. & S.-i.F.); Fax: +81-45-503-7061 (K.S. & S.-i.F.)
| | - Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
| | - Norimitsu Kadowaki
- Department of Internal Medicine, Hematology, Rheumatology and Respiratory Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan;
| | - Arinobu Tojo
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Minato, Tokyo 108-8639, Japan;
| | - Shin-ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
- Correspondence: (K.S.); (S.-i.F.); Tel.: +81-45-503-7062 (K.S. & S.-i.F.); Fax: +81-45-503-7061 (K.S. & S.-i.F.)
| |
Collapse
|
47
|
Gu L, Zhu Y, Lin X, Tan X, Lu B, Li Y. Stabilization of FASN by ACAT1-mediated GNPAT acetylation promotes lipid metabolism and hepatocarcinogenesis. Oncogene 2020; 39:2437-2449. [PMID: 31974474 DOI: 10.1038/s41388-020-1156-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 12/24/2019] [Accepted: 01/10/2020] [Indexed: 11/09/2022]
Abstract
Metabolic alteration for adaptation of the local environment has been recognized as a hallmark of cancer. GNPAT dysregulation has been implicated in hepatocellular carcinoma (HCC). However, the precise posttranslational regulation of GNPAT is still undiscovered. Here we show that ACAT1 is upregulated in response to extra palmitic acid (PA). ACAT1 acetylates GNPAT at K128, which represses TRIM21-mediated GNPAT ubiquitination and degradation. Conversely, GNPAT deacetylation by SIRT4 antagonizes ACAT1's function. GNPAT represses TRIM21-mediated FASN degradation and promotes lipid metabolism. Furthermore, shRNA-mediated ACAT1 ablation and acetylation deficiency of GNPAT repress lipid metabolism and tumor progression in xenograft and DEN/CCl4-induced HCC. Otherwise, ACAT1 inhibitor combination with sorafenib enormously retards tumor formation in mice. Collectively, we demonstrate that stabilization of FASN by ACAT1-mediated GNPAT acetylation plays a critical role in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Li Gu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China. .,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China.
| | - Yahui Zhu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Xi Lin
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Xingyu Tan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Bingjun Lu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Youjun Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China. .,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
48
|
Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in Immunity and Sterile Inflammation. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2019; 15:493-518. [PMID: 31675482 DOI: 10.1146/annurev-pathmechdis-012419-032847] [Citation(s) in RCA: 507] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recognizing the importance of leukocyte trafficking in inflammation led to some therapeutic breakthroughs. However, many inflammatory pathologies remain without specific therapy. This review discusses leukocytes in the context of sterile inflammation, a process caused by sterile (non-microbial) molecules, comprising damage-associated molecular patterns (DAMPs). DAMPs bind specific receptors to activate inflammation and start a highly optimized sequence of immune cell recruitment of neutrophils and monocytes to initiate effective tissue repair. When DAMPs are cleared, the recruited leukocytes change from a proinflammatory to a reparative program, a switch that is locally supervised by invariant natural killer T cells. In addition, neutrophils exit the inflammatory site and reverse transmigrate back to the bloodstream. Inflammation persists when the program switch or reverse transmigration fails, or when the coordinated leukocyte effort cannot clear the immunostimulatory molecules. The latter causes inappropriate leukocyte activation, a driver of many pathologies associated with poor lifestyle choices. We discuss lifestyle-associated inflammatory diseases and their corresponding immunostimulatory lifestyle-associated molecular patterns (LAMPs) and distinguish them from DAMPs.
Collapse
Affiliation(s)
- Joel Zindel
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; .,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Visceral Surgery and Medicine, Department for BioMedical Research, University of Bern, CH-3008 Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; .,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
49
|
Invariant NKT cells facilitate cytotoxic T-cell activation via direct recognition of CD1d on T cells. Exp Mol Med 2019; 51:1-9. [PMID: 31653827 PMCID: PMC6814837 DOI: 10.1038/s12276-019-0329-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/02/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a major subset of NKT cells that recognize foreign and endogenous lipid antigens presented by CD1d. Although iNKT cells are characteristically autoreactive to self-antigens, the role of iNKT cells in the regulation of cytotoxic T lymphocytes (CTL) has been elucidated using α-galactosylceramide (α-GalCer), a strong synthetic glycolipid that is presented by professional antigen presenting cells (APCs), such as dendritic cells. Despite the well-known effects of α-GalCer and dendritic cells on lipid antigen presentation, the physiological role of endogenous antigens presented by CTLs during crosstalk with iNKT cells has not yet been addressed. In this study, we found that antigen-primed CTLs with transient CD1d upregulation could present lipid self-antigens to activate the iNKT cell production of IFN-γ. CTL-mediated iNKT cell activation in turn enhanced IFN-γ production and the proliferation and cytotoxicity of CTLs. We also found that the direct interaction of iNKT cells and CTLs enhanced the antitumor immune responses of CTLs. This partially explains the functional role of iNKT cells in CTL-mediated antitumor immunity. Our findings suggest that in the absence of exogenous iNKT cell ligands, iNKT cells enhanced the CTL production of IFN-γ and CTL proliferation and cytotoxicity via direct interaction with CD1d expressed on T cells without interacting with APCs. Cancer-killing T cells engage in a form of molecular crosstalk with other specialized immune cells to enhance anti-tumor immune responses in mice. Se-Ho Park and colleagues from Korea University in Seoul, South Korea, studied a specialized population of immune cells known as invariant natural killer T (iNKT) cells, which serve as important mediators of tumor surveillance. They showed that iNKT cells directly interact with cancer-killing cytotoxic T cells through surface molecules and not through other immune cells. The resulting activation of iNKT cells leads to the production of a pro-inflammatory signaling molecule, which in turn enhances the proliferation and killing potential of the cytotoxic T cells, ultimately producing more potent tumor control in a mouse model of lymphoma. The findings could aid in the development of iNKT-based cancer immunotherapies.
Collapse
|
50
|
Paul S, Lancaster GI, Meikle PJ. WITHDRAWN: Plasmalogens: A potential therapeutic target for neurodegenerative and cardiometabolic disease. Prog Lipid Res 2019:100993. [PMID: 31442528 DOI: 10.1016/j.plipres.2019.100993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/06/2019] [Accepted: 04/07/2019] [Indexed: 01/30/2023]
Affiliation(s)
- Sudip Paul
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, VIC 3800, Australia
| | - Graeme I Lancaster
- Haematopoiesis and Leukocyte Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, VIC 3800, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, VIC 3800, Australia
| |
Collapse
|