1
|
Melin EO, Thunander M, Wanby P, Holmberg S, Thulesius HO, Landin‐Olsson M, Hillman M. Low levels of soluble neuropilin-1 were associated with depression in adults with newly diagnosed type 2 diabetes. Diabetes Obes Metab 2025; 27:3299-3308. [PMID: 40150914 PMCID: PMC12046488 DOI: 10.1111/dom.16347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
AIMS To explore the association between soluble neuropilin-1 (sNRP-1) and depression in patients with newly diagnosed type 2 diabetes (T2D). MATERIALS AND METHODS Multicentre, cross-sectional study including adults with serologically confirmed newly diagnosed T2D. Included variables: sex, sNPR-1 (low sNRP-1 was defined as <226 ng/mL), psychometrically assessed depression and anxiety, antidepressants, BMI, haemoglobin A1c, C-peptide and pre-existing cardiovascular disease. Multiple regression analyses were performed with depression and low sNRP-1 as dependent variables. RESULTS The study comprised 837 participants (18-94 years, younger patients <60 years 38%). Depressed patients (n = 119) compared to non-depressed (n = 718) had a higher prevalence of anxiety (64% vs. 14%), antidepressants (36% vs. 14%), low sNRP-1 (45% vs. 22%) (all p < 0.001); physical inactivity (42% vs. 29%, p = 0.006); smoking (20% vs. 12%, p = 0.018); and higher BMI (p = 0.002). Independently associated with depression (n = 736) were anxiety (adjusted odds ratio (AOR) 11.7, p < 0.001), low sNRP-1 (AOR 3.3, p < 0.001), BMI (per kg/m2) (AOR 1.1, p = 0.016) and physical inactivity (AOR 1.8, p = 0.018). In younger patients (n = 288), independently associated with low sNRP-1 were depression (AOR 3.3, p < 0.001), myocardial infarction (AOR 3.8, p = 0.039) and younger age (per year) (AOR 0.97, p = 0.043). In older patients (n = 521), independently associated with low sNRP-1 were depression (AOR 3.1, p < 0.001), and younger age (0.97, p = 0.030). CONCLUSIONS Low sNRP-1 (<226 ng/mL) was associated with depression in all patients with newly diagnosed T2D. In younger patients (<60 years), depression, pre-existing myocardial infarction and younger age were associated with low sNRP-1. In older patients, only depression and younger age were associated with low sNRP-1.
Collapse
Affiliation(s)
- E. O. Melin
- Diabetes Research LaboratoryBiomedical Center, Lund UniversityLundSweden
- Department of Clinical Sciences, Diabetology and EndocrinologyLund UniversityLundSweden
- Department of Research and DevelopmentRegion KronobergVäxjöSweden
| | - M. Thunander
- Diabetes Research LaboratoryBiomedical Center, Lund UniversityLundSweden
- Department of Clinical Sciences, Diabetology and EndocrinologyLund UniversityLundSweden
- Department of Research and DevelopmentRegion KronobergVäxjöSweden
| | - P. Wanby
- Department of Medicine and OptometryFaculty of Health and Life Sciences, Linnaeus UniversityKalmarSweden
- Department of Medical and Health SciencesUniversity of LinköpingLinköpingSweden
- Department of Internal Medicine, Section of EndocrinologyRegion Kalmar CountySweden
| | - S. Holmberg
- Department of Research and DevelopmentRegion KronobergVäxjöSweden
- Department of Medicine and OptometryFaculty of Health and Life Sciences, Linnaeus UniversityKalmarSweden
- Division of Occupational and Environmental MedicineDepartment of Laboratory Medicine, Lund UniversityLundSweden
| | - H. O. Thulesius
- Department of Medicine and OptometryFaculty of Health and Life Sciences, Linnaeus UniversityKalmarSweden
- Department of Clinical Sciences, Division of Family MedicineLund UniversityMalmöSweden
| | - M. Landin‐Olsson
- Department of Clinical Sciences, Diabetology and EndocrinologyLund UniversityLundSweden
- Department of EndocrinologySkane University HospitalLundSweden
| | - M. Hillman
- Diabetes Research LaboratoryBiomedical Center, Lund UniversityLundSweden
| |
Collapse
|
2
|
Hui H, Yu Y, Yiwei L, Li Y, Liling X, Dongguang Z. Genetic etiology and clinical features of non-syndromic pediatric obesity in the Chinese population: a large cohort study. BMC Pediatr 2025; 25:358. [PMID: 40329189 PMCID: PMC12057247 DOI: 10.1186/s12887-025-05702-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND This study aimed to investigate the genetic etiology and clinical features of non-syndromic pediatric obesity in a large Chinese cohort, providing insights into the genetic profile and its correlation with clinical phenotypes. METHODS We enrolled 391 children, aged 7-14 years, diagnosed with non-syndromic pediatric obesity at Jiangxi Provincial Children's Hospital from January 2020 to June 2022. Whole-exome sequencing was employed to identify potential genetic causes, focusing on 79 candidate genes associated with obesity. Multivariate logistic regression analysis was performed on the clinical data of the non-syndromic obesity gene-positive group and the gene-negative group. RESULTS Among the 391 patients, 32 (8.2%) carried 18 non-syndromic obesity genes, with UCP3 and MC4R being the most common. Seven cases (1.8%) were rated as likely pathogenic by the American College of Medical Genetics and Genomics (ACMG). Clinical phenotype and genetic correlation analysis revealed that urinary microalbumin, fT4, GGT, uric acid, serum phosphorus, paternal weight, family history, impaired glucose tolerance (IGT), non-HDL cholesterol (non-HDL-C), and metabolic syndrome (MetS) showed significant statistical differences (P < 0.05). Serum phosphorus is an independent risk factor associated with genetic predispositions to obesity in children and adolescents (P < 0.05). CONCLUSION Our findings highlight the genetic heterogeneity of non-syndromic pediatric obesity and identify UCP3 and MC4R as potential hotspot genes in the Chinese population. The study underscores the potential of genetic testing for early diagnosis and personalized management of pediatric obesity.
Collapse
Affiliation(s)
- Huang Hui
- Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Child Development and Genetics, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Yang Yu
- Jiangxi Provincial Key Laboratory of Child Development and Genetics, Jiangxi Provincial Children's Hospital, Nanchang, China.
- Department of Endocrinology, Genetics and Metabolism, Jiangxi Provincial Children's Hospital Clinical Medical Research Center of Genetic Metabolic Diseases in Children, Nanchang, China.
| | - Liang Yiwei
- Department of Child Health, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Yang Li
- Department of Endocrinology, Genetics and Metabolism, Jiangxi Provincial Children's Hospital Clinical Medical Research Center of Genetic Metabolic Diseases in Children, Nanchang, China
| | - Xie Liling
- Department of Endocrinology, Genetics and Metabolism, Jiangxi Provincial Children's Hospital Clinical Medical Research Center of Genetic Metabolic Diseases in Children, Nanchang, China
| | - Zhang Dongguang
- Department of Endocrinology, Genetics and Metabolism, Jiangxi Provincial Children's Hospital Clinical Medical Research Center of Genetic Metabolic Diseases in Children, Nanchang, China
| |
Collapse
|
3
|
van Aanhold CCL, Yong Q, Landman L, Sardana S, Bouwmeester AB, Dijkstra KL, Wolterbeek R, Mei H, Tjokrodirijo RTN, de Ru AH, van Veelen PA, Bruijn JA, van Kooten C, Baelde HJ. The VEGF decoy receptor soluble Fms-like tyrosine kinase 1 binds to macrophages. Angiogenesis 2025; 28:28. [PMID: 40314836 PMCID: PMC12048422 DOI: 10.1007/s10456-025-09980-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/17/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Soluble Fms-like Tyrosine kinase-1 (sFLT1) is a native inhibitor of VEGF, best known for its antiangiogenic effects in preeclampsia. sFLT1 also reduces chronic inflammation and promotes tissue repair. In experimental diabetic nephropathy, we previously found that sFLT1 ameliorates kidney fibrosis and reduces the infiltration of macrophages. How sFLT1 regulates inflammation is still incompletely understood. Based on the direct association of sFLT1 with various cell types, we here studied whether sFLT1 interacts with macrophages to modulate inflammation. METHODS Using various macrophage cell lines, sFLT1 cell surface binding was detected with flow cytometry. Enzyme studies, mass spectrometry and RNAseq were employed to identify potential sFLT1 cell surface interactors and effects of sFLT1 on macrophage signaling. RESULTS Soluble FLT1 binds to primary macrophages, THP-1 and RAW264.7 macrophages in vitro. Alternative activation with IL-4 increases sFLT1 binding in THP-1 macrophages, whereas proinflammatory activation with IFN-γ and LPS decreases binding. Binding of sFLT1 depends on heparan sulphates, and colocalizes with the membrane heparin sulfate proteoglycan neuropilin-1. Incubation with sFLT1 reduces the gene expression of chemokine receptors. CONCLUSION Our results show that sFLT1, while typically associated with angiogenesis, also directly interacts with macrophages. Alternative activation of macrophages by IL-4 strongly increases binding of sFLT1 to the cell surface membrane, possibly via the VEGF co-receptor neuropilin-1. Considering sFLT1's anti-inflammatory effects in animal studies, our findings indicate a novel function for sFLT1 to directly control anti-inflammatory macrophage function.
Collapse
Affiliation(s)
- Cleo C L van Aanhold
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Qing Yong
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa Landman
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Samiksha Sardana
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Anouk B Bouwmeester
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kyra L Dijkstra
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ron Wolterbeek
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Rayman T N Tjokrodirijo
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnoud H de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan A Bruijn
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cees van Kooten
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
4
|
de Melo BP, da Silva JAM, Rodrigues MA, Palmeira JDF, Amato AA, Argañaraz GA, Argañaraz ER. SARS-CoV-2 Spike Protein and Long COVID-Part 2: Understanding the Impact of Spike Protein and Cellular Receptor Interactions on the Pathophysiology of Long COVID Syndrome. Viruses 2025; 17:619. [PMID: 40431631 PMCID: PMC12115913 DOI: 10.3390/v17050619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/06/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
SARS-CoV-2 infection has had a significant impact on global health through both acute illness, referred to as coronavirus disease 2019 (COVID-19), and chronic conditions (long COVID or post-acute sequelae of COVID-19, PASC). Despite substantial advancements in preventing severe COVID-19 cases through vaccination, the rise in the prevalence of long COVID syndrome and a notable degree of genomic mutation, primarily in the S protein, underscores the necessity for a deeper understanding of the underlying pathophysiological mechanisms related to the S protein of SARS-CoV-2. In this review, the latest part of this series, we investigate the potential pathophysiological molecular mechanisms triggered by the interaction between the spike protein and cellular receptors. Therefore, this review aims to provide a differential and focused view on the mechanisms potentially activated by the binding of the spike protein to canonical and non-canonical receptors for SARS-CoV-2, together with their possible interactions and effects on the pathogenesis of long COVID.
Collapse
Affiliation(s)
- Bruno Pereira de Melo
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Jhéssica Adriane Mello da Silva
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Mariana Alves Rodrigues
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Julys da Fonseca Palmeira
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Angélica Amorim Amato
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Gustavo Adolfo Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Enrique Roberto Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| |
Collapse
|
5
|
Sur S, Stewart C, Liddle TA, Monteiro AM, Denizli I, Majumdar G, Stevenson TJ. Molecular basis of photoinduced seasonal energy rheostasis in Japanese quail (Coturnix japonica). Mol Cell Endocrinol 2025; 595:112415. [PMID: 39561917 DOI: 10.1016/j.mce.2024.112415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Seasonal rhythms in photoperiod are a predictive cue used by many temperate-zone animals to time cycles of lipid accumulation. The neuroendocrine regulation of seasonal energy homeostasis and rheostasis are widely studied. However, the molecular pathways underlying tissue-specific adaptations remain poorly described. We conducted two experiments to examine long-term rheostatic changes in energy stability using the well-characterized photoperiodic response of the Japanese quail. In experiment 1, we exposed quails to photoperiodic transitions simulating the annual photic cycle and examined the morphology and fat deposition in liver, muscle, and adipose tissue. To identify changes in gene expression and molecular pathways during the vernal transition in lipid accumulation, we conducted transcriptomic analyses of adipose and liver tissues. Experiment 2 assessed whether the changes observed in Experiment 1 reflected constitutive levels or were due to time-of-day sampling. We identified increased expression of transcripts involved in adipocyte growth, such as Cysteine Rich Angiogenic Inducer 61 and Very Low-Density Lipoprotein Receptor, and in obesity-linked disease resistance, such as Insulin-Like Growth Factor Binding Protein 2 and Apolipoprotein D, in anticipation of body mass gain. Under long photoperiods, hepatic transcripts involved in fatty acid (FA) synthesis (FA Synthase, FA Desaturase 2) were down-regulated. Parallel upregulation of hepatic FA Translocase and Pyruvate Dehydrogenase Kinase 4 expression suggests increased FA uptake and inhibition of the pyruvate dehydrogenase complex. Our findings demonstrate tissue-specific biochemical and molecular changes that drive photoperiod-induced adipogenesis. These findings can be used to determine conserved pathways that enable animals to accumulate fat without developing metabolic diseases.
Collapse
Affiliation(s)
- Sayantan Sur
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom.
| | - Calum Stewart
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom.
| | - Timothy A Liddle
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom.
| | - Ana Maria Monteiro
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom.
| | - Irem Denizli
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom.
| | - Gaurav Majumdar
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom; Department of Zoology, University of Allahabad, Uttar Pradesh, 211002, India.
| | - Tyler J Stevenson
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
6
|
Takaoka M, Zhao X, Lim HY, Magnussen CG, Ang O, Suffee N, Schrank PR, Ong WS, Tsiantoulas D, Sommer F, Mohanta SK, Harrison J, Meng Y, Laurans L, Wu F, Lu Y, Masters L, Newland SA, Denti L, Hong M, Chajadine M, Juonala M, Koskinen JS, Kähönen M, Pahkala K, Rovio SP, Mykkänen J, Thomson R, Kaisho T, Habenicht AJR, Clement M, Tedgui A, Ait-Oufella H, Zhao TX, Nus M, Ruhrberg C, Taleb S, Williams JW, Raitakari OT, Angeli V, Mallat Z. Early intermittent hyperlipidaemia alters tissue macrophages to fuel atherosclerosis. Nature 2024; 634:457-465. [PMID: 39231480 PMCID: PMC11464399 DOI: 10.1038/s41586-024-07993-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Hyperlipidaemia is a major risk factor of atherosclerotic cardiovascular disease (ASCVD). Risk of cardiovascular events depends on cumulative lifetime exposure to low-density lipoprotein cholesterol (LDL-C) and, independently, on the time course of exposure to LDL-C, with early exposure being associated with a higher risk1. Furthermore, LDL-C fluctuations are associated with ASCVD outcomes2-4. However, the precise mechanisms behind this increased ASCVD risk are not understood. Here we find that early intermittent feeding of mice on a high-cholesterol Western-type diet (WD) accelerates atherosclerosis compared with late continuous exposure to the WD, despite similar cumulative circulating LDL-C levels. We find that early intermittent hyperlipidaemia alters the number and homeostatic phenotype of resident-like arterial macrophages. Macrophage genes with altered expression are enriched for genes linked to human ASCVD in genome-wide association studies. We show that LYVE1+ resident macrophages are atheroprotective, and identify biological pathways related to actin filament organization, of which alteration accelerates atherosclerosis. Using the Young Finns Study, we show that exposure to cholesterol early in life is significantly associated with the incidence and size of carotid atherosclerotic plaques in mid-adulthood. In summary, our results identify early intermittent exposure to cholesterol as a strong determinant of accelerated atherosclerosis, highlighting the importance of optimal control of hyperlipidaemia early in life, and providing insights into the underlying biological mechanisms. This knowledge will be essential to designing effective therapeutic strategies to combat ASCVD.
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Child
- Child, Preschool
- Female
- Humans
- Male
- Mice
- Middle Aged
- Young Adult
- Atherosclerosis/epidemiology
- Atherosclerosis/etiology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cholesterol, LDL/blood
- Cholesterol, LDL/metabolism
- Diet, Western/adverse effects
- Diet, Western/statistics & numerical data
- Finland/epidemiology
- Genome-Wide Association Study
- Hyperlipidemias/complications
- Hyperlipidemias/epidemiology
- Hyperlipidemias/genetics
- Hyperlipidemias/metabolism
- Hyperlipidemias/pathology
- Incidence
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Inbred C57BL
- Phenotype
- Plaque, Atherosclerotic/epidemiology
- Plaque, Atherosclerotic/etiology
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Time Factors
Collapse
Affiliation(s)
- Minoru Takaoka
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Xiaohui Zhao
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Costan G Magnussen
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Owen Ang
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Nadine Suffee
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Patricia R Schrank
- Department of Integrative Biology & Physiology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Wei Siong Ong
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Dimitrios Tsiantoulas
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Felix Sommer
- Institute of Clinical Molecular Biology, University of Kiel and University Hospital Schleswig Holstein (UKSH), Kiel, Germany
| | - Sarajo K Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - James Harrison
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Yaxing Meng
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Ludivine Laurans
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Feitong Wu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yuning Lu
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Leanne Masters
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Stephen A Newland
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Laura Denti
- Institute of Ophthalmology, University College London, London, UK
| | - Mingyang Hong
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Mouna Chajadine
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Markus Juonala
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Juhani S Koskinen
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Medicine, Satakunta Central Hospital, Pori, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere, Tampere, Finland
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, University of Tampere, Tampere, Finland
| | - Katja Pahkala
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Suvi P Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Juha Mykkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Russell Thomson
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Analytical Edge, Hobart, Tasmania, Australia
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Marc Clement
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Alain Tedgui
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Hafid Ait-Oufella
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Tian X Zhao
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | - Meritxell Nus
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK
| | | | - Soraya Taleb
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France
| | - Jesse W Williams
- Department of Integrative Biology & Physiology, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Véronique Angeli
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Ziad Mallat
- Department of Medicine, Section of CardioRespiratory Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, UK.
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, U970, PARCC, Paris, France.
| |
Collapse
|
7
|
Liu J, Chen Y. Cell-cell crosstalk between fat cells and immune cells. Am J Physiol Endocrinol Metab 2024; 327:E371-E383. [PMID: 39082899 DOI: 10.1152/ajpendo.00024.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/06/2024]
Abstract
Obesity is a metabolic disorder with pandemic-like implications, lacking viable pharmaceutical treatments currently. Thermogenic adipose tissues, including brown and beige adipose tissues, play an essential role in regulating systemic energy homeostasis and have emerged as appealing therapeutic targets for the treatment of obesity and obesity-related diseases. The function of adipocytes is subject to complex regulation by a cellular network of immune signaling pathways in response to environmental signals. However, the specific regulatory roles of immune cells in thermogenesis and relevant involving mechanisms are still not well understood. Here, we concentrate on our present knowledge of the interaction between thermogenic adipocytes and immune cells and present an overview of cellular and molecular mechanisms underlying immunometabolism in adipose tissues. We discuss cytokines, especially interleukins, which originate from widely variable sources, and their impacts on the development and function of thermogenic adipocytes. Moreover, we summarize the neuroimmune regulation in heat production and expand a new mode of intercellular communication mediated by mitochondrial transfer. The crosstalk between immune cells and adipocytes achieves adipose tissue homeostasis and systemic energy balance. A deep understanding of this intricate interaction would provide evidence for improving thermogenic efficiency by remodeling the immune microenvironment. Interventions based on these factors show a high potential to prevent adverse metabolic outcomes in patients with obesity.
Collapse
Affiliation(s)
- Jiadai Liu
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Laboratory of Endocrinology and Metabolism, Ministry of Education Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Laboratory of Endocrinology and Metabolism, Ministry of Education Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, People's Republic of China
| |
Collapse
|
8
|
Lechner K, Kia S, von Korn P, Dinges SM, Mueller S, Tjønna AE, Wisløff U, Van Craenenbroeck EM, Pieske B, Adams V, Pressler A, Landmesser U, Halle M, Kränkel N. Cardiometabolic and immune response to exercise training in patients with metabolic syndrome: retrospective analysis of two randomized clinical trials. Front Cardiovasc Med 2024; 11:1329633. [PMID: 38638882 PMCID: PMC11025358 DOI: 10.3389/fcvm.2024.1329633] [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: 10/29/2023] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
Background Metabolic syndrome (MetS) is defined by the presence of central obesity plus ≥two metabolic/cardiovascular risk factors (RF), with inflammation being a major disease-driving mechanism. Structured endurance exercise training (ET) may positively affect these traits, as well as cardiorespiratory fitness (V̇O2peak). Aims We explore individual ET-mediated improvements of MetS-associated RF in relation to improvements in V̇O2peak and inflammatory profile. Methods MetS patients from two randomized controlled trials, ExMET (n = 24) and OptimEx (n = 34), had performed 4- or 3-months supervised ET programs according to the respective trial protocol. V̇O2peak, MetS-defining RFs (both RCTs), broad blood leukocyte profile, cytokines and plasma proteins (ExMET only) were assessed at baseline and follow-up. Intra-individual changes in RFs were analysed for both trials separately using non-parametric approaches. Associations between changes in each RF over the exercise period (n-fold of baseline values) were correlated using a non-parametrical approach (Spearman). RF clustering was explored by uniform manifold approximation and projection (UMAP) and changes in RF depending on other RF or exercise parameters were explored by recursive partitioning. Results Four months of ET reduced circulating leukocyte counts (63.5% of baseline, P = 8.0e-6), especially effector subtypes. ET response of MetS-associated RFs differed depending on patients' individual RF constellation, but was not associated with individual change in V̇O2peak. Blood pressure lowering depended on cumulative exercise duration (ExMET: ≥102 min per week; OptimEx-MetS: ≥38 min per session) and baseline triglyceride levels (ExMET: <150 mg/dl; OptimEx-MetS: <174.8 mg/dl). Neuropilin-1 plasma levels were inversely associated with fasting plasma triglycerides (R: -0.4, P = 0.004) and changes of both parameters during the ET phase were inversely correlated (R: -0.7, P = 0.0001). Conclusions ET significantly lowered effector leukocyte blood counts. The improvement of MetS-associated cardiovascular RFs depended on individual basal RF profile and exercise duration but was not associated with exercise-mediated increase in V̇O2peak. Neuropilin-1 may be linked to exercise-mediated triglyceride lowering.
Collapse
Affiliation(s)
- Katharina Lechner
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Sylvia Kia
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
| | - Pia von Korn
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Sophia M. Dinges
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Stephan Mueller
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Arnt-Erik Tjønna
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisløff
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Emeline M. Van Craenenbroeck
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Volker Adams
- Department of Cardiology and Internal Medicine, Heart Center Dresden-University Hospital, TU Dresden, Dresden, Germany
| | - Axel Pressler
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Private Center for Sports and Exercise Cardiology, Munich, Germany
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
- Friede Springer—Centre of Cardiovascular Prevention at Charité, Charité University Medicine Berlin, Berlin, Germany
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nicolle Kränkel
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie und Intensivmedizin, Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site, Berlin, Germany
- Friede Springer—Centre of Cardiovascular Prevention at Charité, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
9
|
Patel NA, Lui A, Trujillo AN, Motawe ZY, Bader D, Schuster J, Burgess A, Alves NG, Jo M, Breslin JW. Female and male obese Zucker rats display differential inflammatory mediator and long non-coding RNA profiles. Life Sci 2023; 335:122285. [PMID: 37995934 PMCID: PMC10760426 DOI: 10.1016/j.lfs.2023.122285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023]
Abstract
AIMS The goal of this study was to identify mediators in peri-lymphatic adipose tissue (PLAT) that are altered in obese versus lean Zucker rats, with focus on potential sex differences MAIN METHODS: Mesenteric PLAT was analyzed with protein and lncRNA arrays. Additional RT-PCR confirmation was performed with epididymal/ovarian fat. KEY FINDINGS MCP-1, TCK-1, Galectin-1, Galectin-3, and neuropilin-1 were elevated in PLAT from obese rats of both sexes. However, 11 additional proteins were elevated only in obese males while 24 different proteins were elevated in obese females. Profiling of lncRNAs revealed lean males have elevated levels of NEAT1, MALAT1 and GAS5 compared to lean females. NEAT1, MALAT1, and GAS5 were significantly reduced with obesity in males but not in females. Another lncRNA, HOTAIR, was higher in lean females compared to males, and its levels in females were reduced with obesity. Obese rats of both sexes had similar histologic findings of mesenteric macrophage crown-like structures and hepatocyte fat accumulation. SIGNIFICANCE While obese male and female Zucker rats both have increased inflammation, they have distinct signals. Future studies of the proteome and lncRNA landscape of obese males vs. females in various animal models and in human subjects are warranted to better guide development of therapeutics for obesity-induced inflammation.
Collapse
Affiliation(s)
- Niketa A Patel
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, United States of America; James A. Haley Veteran's Hospital, United States of America
| | - Ashley Lui
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, United States of America; James A. Haley Veteran's Hospital, United States of America
| | - Andrea N Trujillo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Zeinab Y Motawe
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Deena Bader
- James A. Haley Veteran's Hospital, United States of America
| | - Jane Schuster
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Andrea Burgess
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Natascha G Alves
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America
| | - Michiko Jo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America; Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Japan
| | - Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, United States of America.
| |
Collapse
|
10
|
Saleki K, Alijanizadeh P, Azadmehr A. Is neuropilin-1 the neuroimmune initiator of multi-system hyperinflammation in COVID-19? Biomed Pharmacother 2023; 167:115558. [PMID: 37748412 DOI: 10.1016/j.biopha.2023.115558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023] Open
Abstract
A major immunopathological feature of Coronavirus disease-2019 (COVID-19) is excessive inflammation in the form of "cytokine storm". The storm is characterized by injurious levels of cytokines which form a complicated network damaging different organs, including the lungs and the brain. The main starter of "cytokine network" hyperactivation in COVID-19 has not been discovered yet. Neuropilins (NRPs) are transmembrane proteins that act as neuronal guidance and angiogenesis modulators. The crucial function of NRPs in forming the nervous and vascular systems has been well-studied. NRP1 and NRP2 are the two identified homologs of NRP. NRP1 has been shown as a viral entry pathway for SARS-CoV2, which facilitates neuroinvasion by the virus within the central or peripheral nervous systems. These molecules directly interact with various COVID-19-related molecules, such as specific regions of the spike protein (major immune element of SARS-CoV2), vascular endothelial growth factor (VEGF) receptors, VEGFR1/2, and ANGPTL4 (regulator of vessel permeability and integrity). NRPs mainly play a role in hyperinflammatory injury of the CNS and lungs, and also the liver, kidney, pancreas, and heart in COVID-19 patients. New findings have suggested NRPs good candidates for pharmacotherapy of COVID-19. However, therapeutic targeting of NRP1 in COVID-19 is still in the preclinical phase. This review presents the implications of NRP1 in multi-organ inflammation-induced injury by SARS-CoV2 and provides insights for NRP1-targeting treatments for COVID-19 patients.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences(SBMU), Tehran, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
11
|
Mela V, Agüera Z, Alvarez-Bermudez MD, Martín-Reyes F, Granero R, Sánchez-García A, Oliva-Olivera W, Tomé M, Moreno-Ruiz FJ, Soler-Humanes R, Fernández-Serrano JL, Sánchez-Gallegos P, Martínez-Moreno JM, Sancho-Marín R, Fernández-Aranda F, García-Fuentes E, Tinahones FJ, Garrido-Sánchez L. The Relationship between Depressive Symptoms, Quality of Life and miRNAs 8 Years after Bariatric Surgery. Nutrients 2023; 15:4109. [PMID: 37836393 PMCID: PMC10574314 DOI: 10.3390/nu15194109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
(1) Background: There are conflicting results on whether weight loss after bariatric surgery (BS) might be associated with quality of life (QoL)/depressive symptomatology. We aim to determine whether BS outcomes are associated with QoL/depressive symptomatology in studied patients at the 8-year follow-up after BS, as well as their relationship with different serum proteins and miRNAs. (2) Methods: A total of 53 patients with class III obesity who underwent BS, and then classified into "good responders" and "non-responders" depending on the percentage of excess weight lost (%EWL) 8 years after BS (%EWL ≥ 50% and %EWL < 50%, respectively), were included. Basal serum miRNAs and different proteins were analysed, and patients completed tests to evaluate QoL/depressive symptomatology at 8 years after BS. (3) Results: The good responders group showed higher scores on SF-36 scales of physical functioning, role functioning-physical, role functioning-emotional, body pain and global general health compared with the non-responders. The expression of hsa-miR-101-3p, hsa-miR-15a-5p, hsa-miR-29c-3p, hsa-miR-144-3p and hsa-miR-19b-3p were lower in non-responders. Hsa-miR-19b-3p was the variable associated with the response to BS in a logistic regression model. (4) Conclusions: The mental health of patients after BS is limited by the success of the intervention. In addition, the expression of basal serum miRNAs related to depression/anxiety could predict the success of BS.
Collapse
Affiliation(s)
- Virginia Mela
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain; (V.M.); (M.D.A.-B.); (F.M.-R.); (A.S.-G.); (W.O.-O.); (L.G.-S.)
- Department of Medicine and Dermatology, Faculty of Medicine, University of Malaga, 29010 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
| | - Zaida Agüera
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
- Departament d’Infermeria de Salut Pública, Salut Mental i Maternoinfantil, Escola d’Infermeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain
| | - Maria D. Alvarez-Bermudez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain; (V.M.); (M.D.A.-B.); (F.M.-R.); (A.S.-G.); (W.O.-O.); (L.G.-S.)
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
| | - Flores Martín-Reyes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain; (V.M.); (M.D.A.-B.); (F.M.-R.); (A.S.-G.); (W.O.-O.); (L.G.-S.)
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Roser Granero
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain
- Department of Psychobiology and Methodology, Autonomous University of Barcelona, 08193 Barcelona, Spain
| | - Ana Sánchez-García
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain; (V.M.); (M.D.A.-B.); (F.M.-R.); (A.S.-G.); (W.O.-O.); (L.G.-S.)
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
| | - Wilfredo Oliva-Olivera
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain; (V.M.); (M.D.A.-B.); (F.M.-R.); (A.S.-G.); (W.O.-O.); (L.G.-S.)
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
| | - Monica Tomé
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Regional Universitario de Málaga, 29009 Malaga, Spain;
| | - Francisco J. Moreno-Ruiz
- Unidad de Gestión Clínica de Cirugía General y Digestiva, Hospital Regional Universitario de Málaga, 29010 Malaga, Spain;
| | - Rocío Soler-Humanes
- Unidad de Gestión Clínica de Cirugía General y Digestiva, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain; (R.S.-H.); (J.L.F.-S.)
| | - Jose L. Fernández-Serrano
- Unidad de Gestión Clínica de Cirugía General y Digestiva, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain; (R.S.-H.); (J.L.F.-S.)
| | - Pilar Sánchez-Gallegos
- Department of Surgical Specialities, Biochemistry and Immunology, Faculty of Medicine, University of Malaga, 29010 Malaga, Spain; (P.S.-G.); (J.M.M.-M.); (R.S.-M.)
| | - Jose M. Martínez-Moreno
- Department of Surgical Specialities, Biochemistry and Immunology, Faculty of Medicine, University of Malaga, 29010 Malaga, Spain; (P.S.-G.); (J.M.M.-M.); (R.S.-M.)
| | - Raquel Sancho-Marín
- Department of Surgical Specialities, Biochemistry and Immunology, Faculty of Medicine, University of Malaga, 29010 Malaga, Spain; (P.S.-G.); (J.M.M.-M.); (R.S.-M.)
| | - Fernando Fernández-Aranda
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain
- Department of Psychiatry, University Hospital of Bellvitge, 08907 Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08907 Barcelona, Spain
| | - Eduardo García-Fuentes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain; (V.M.); (M.D.A.-B.); (F.M.-R.); (A.S.-G.); (W.O.-O.); (L.G.-S.)
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- CIBER Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Francisco J. Tinahones
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain; (V.M.); (M.D.A.-B.); (F.M.-R.); (A.S.-G.); (W.O.-O.); (L.G.-S.)
- Department of Medicine and Dermatology, Faculty of Medicine, University of Malaga, 29010 Malaga, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
| | - Lourdes Garrido-Sánchez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, 29590 Malaga, Spain; (V.M.); (M.D.A.-B.); (F.M.-R.); (A.S.-G.); (W.O.-O.); (L.G.-S.)
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto Salud Carlos III, 28029 Madrid, Spain; (Z.A.); (R.G.); (F.F.-A.)
| |
Collapse
|
12
|
Mazitova AM, Márquez-Sánchez AC, Koltsova EK. Fat and inflammation: adipocyte-myeloid cell crosstalk in atherosclerosis. Front Immunol 2023; 14:1238664. [PMID: 37781401 PMCID: PMC10540690 DOI: 10.3389/fimmu.2023.1238664] [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: 06/12/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Adipose tissue inflammation has been implicated in various chronic inflammatory diseases and cancer. Perivascular adipose tissue (PVAT) surrounds the aorta as an extra layer and was suggested to contribute to atherosclerosis development. PVAT regulates the function of endothelial and vascular smooth muscle cells in the aorta and represent a reservoir for various immune cells which may participate in aortic inflammation. Recent studies demonstrate that adipocytes also express various cytokine receptors and, therefore, may directly respond to inflammatory stimuli. Here we will summarize current knowledge on immune mechanisms regulating adipocyte activation and the crosstalk between myeloid cells and adipocytes in pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Aleksandra M. Mazitova
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ana Cristina Márquez-Sánchez
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ekaterina K. Koltsova
- Cedars-Sinai Cancer, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
13
|
Tian Z, Yang S. Integrating the characteristic genes of macrophage pseudotime analysis in single-cell RNA-seq to construct a prediction model of atherosclerosis. Aging (Albany NY) 2023; 15:6361-6379. [PMID: 37421595 PMCID: PMC10373969 DOI: 10.18632/aging.204856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Macrophages play an important role in the occurrence and development of atherosclerosis. However, few existing studies have deliberately analyzed the changes in characteristic genes in the process of macrophage phenotype transformation. METHOD Carotid atherosclerotic plaque single-cell RNA (scRNA) sequencing data were analyzed to define the cells involved and determine their transcriptomic characteristics. KEGG enrichment analysis, CIBERSORT, ESTIMATE, support vector machine (SVM), random forest (RF), and weighted correlation network analysis (WGCNA) were applied to bulk sequencing data. All data were downloaded from Gene Expression Omnibus (GEO). RESULT Nine cell clusters were identified. M1 macrophages, M2 macrophages, and M2/M1 macrophages were identified as three clusters within the macrophages. According to pseudotime analysis, M2/M1 macrophages and M2 macrophages can be transformed into M1 macrophages. The ROC curve values of the six genes in the test group were statistically significant (AUC (IL1RN): 0.899, 95% CI: 0.764-0.990; AUC (NRP1): 0.817, 95% CI: 0.620-0.971; AUC (TAGLN): 0.846, 95% CI: 0.678-0.971; AUC (SPARCL1): 0.825, 95% CI: 0.620-0.988; AUC (EMP2): 0.808, 95% CI: 0.630-0.947; AUC (ACTA2): 0.784, 95% CI: 0.591-0.938). The atherosclerosis prediction model showed significant statistical significance in both the train group (AUC: 0.909, 95% CI: 0.842-0.967) and the test group (AUC: 0.812, 95% CI: 0.630-0.966). CONCLUSIONS IL1RNHigh M1, NRP1High M2, ACTA2High M2/M1, EMP2High M1/M1, SPACL1High M2/M1 and TAGLNHigh M2/M1 macrophages play key roles in the occurrence and development of arterial atherosclerosis. These marker genes of macrophage phenotypic transformation can also be used to establish a model to predict the occurrence of atherosclerosis.
Collapse
Affiliation(s)
- Zemin Tian
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Shize Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| |
Collapse
|
14
|
Mizuno Y, Nakanishi Y, Kumanogoh A. Pathophysiological functions of semaphorins in the sympathetic nervous system. Inflamm Regen 2023; 43:30. [PMID: 37291626 DOI: 10.1186/s41232-023-00281-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Upon exposure to external stressors, the body senses them and activates the sympathetic nervous system (SNS) to maintain the homeostasis, which is known as the "fight-or-flight" response. Recent studies have revealed that the SNS also plays pivotal roles in regulating immune responses, such as hematopoiesis, leukocyte mobilization, and inflammation. Indeed, overactivation of the SNS causes many inflammatory diseases, including cardiovascular diseases, metabolic disorders, and autoimmune diseases. However, the molecular basis essential for SNS-mediated immune regulation is not completely understood. In this review, we focus on axon guidance cues, semaphorins, which play multifaceted roles in neural and immune systems. We summarize the functions of semaphorins in the crosstalk between the SNS and the immune system, exploring its pathophysiological roles.
Collapse
Affiliation(s)
- Yumiko Mizuno
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan
| | - Yoshimitsu Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan.
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan.
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
15
|
Hautz T, Salcher S, Fodor M, Sturm G, Ebner S, Mair A, Trebo M, Untergasser G, Sopper S, Cardini B, Martowicz A, Hofmann J, Daum S, Kalb M, Resch T, Krendl F, Weissenbacher A, Otarashvili G, Obrist P, Zelger B, Öfner D, Trajanoski Z, Troppmair J, Oberhuber R, Pircher A, Wolf D, Schneeberger S. Immune cell dynamics deconvoluted by single-cell RNA sequencing in normothermic machine perfusion of the liver. Nat Commun 2023; 14:2285. [PMID: 37085477 PMCID: PMC10121614 DOI: 10.1038/s41467-023-37674-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/27/2023] [Indexed: 04/23/2023] Open
Abstract
Normothermic machine perfusion (NMP) has emerged as an innovative organ preservation technique. Developing an understanding for the donor organ immune cell composition and its dynamic changes during NMP is essential. We aimed for a comprehensive characterization of immune cell (sub)populations, cell trafficking and cytokine release during liver NMP. Single-cell transcriptome profiling of human donor livers prior to, during NMP and after transplantation shows an abundance of CXC chemokine receptor 1+/2+ (CXCR1+/CXCR2+) neutrophils, which significantly decreased during NMP. This is paralleled by a large efflux of passenger leukocytes with neutrophil predominance in the perfusate. During NMP, neutrophils shift from a pro-inflammatory state towards an aged/chronically activated/exhausted phenotype, while anti-inflammatory/tolerogenic monocytes/macrophages are increased. We herein describe the dynamics of the immune cell repertoire, phenotypic immune cell shifts and a dominance of neutrophils during liver NMP, which potentially contribute to the inflammatory response. Our findings may serve as resource to initiate future immune-interventional studies.
Collapse
Affiliation(s)
- T Hautz
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - S Salcher
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - M Fodor
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - G Sturm
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - S Ebner
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - A Mair
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - M Trebo
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - G Untergasser
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
- Tyrolpath Obrist Brunhuber GmbH, Zams, Austria
| | - S Sopper
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - B Cardini
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - A Martowicz
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
- Tyrolpath Obrist Brunhuber GmbH, Zams, Austria
| | - J Hofmann
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - S Daum
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - M Kalb
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - T Resch
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - F Krendl
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - A Weissenbacher
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - G Otarashvili
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - P Obrist
- Tyrolpath Obrist Brunhuber GmbH, Zams, Austria
| | - B Zelger
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | - D Öfner
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - Z Trajanoski
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - J Troppmair
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - R Oberhuber
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - A Pircher
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - D Wolf
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria.
| | - S Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory and D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
16
|
Yu Y, Uchida-Fukuhara Y, Weng Y, He Y, Ikegame M, Wang Z, Yoshida K, Okamura H, Qiu L. Neuropilin 1 (NRP1) Positively Regulates Adipogenic Differentiation in C3H10T1/2 Cells. Int J Mol Sci 2023; 24:ijms24087394. [PMID: 37108554 PMCID: PMC10138427 DOI: 10.3390/ijms24087394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/25/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Neuropilin 1 (NRP1), a non-tyrosine kinase receptor for several ligands, is highly expressed in many kinds of mesenchymal stem cells (MSCs), but its function is poorly understood. In this study, we explored the roles of full-length NRP1 and glycosaminoglycan (GAG)-modifiable NRP1 in adipogenesis in C3H10T1/2 cells. The expression of full-length NRP1 and GAG-modifiable NRP1 increased during adipogenic differentiation in C3H10T1/2 cells. NRP1 knockdown repressed adipogenesis while decreasing the levels of Akt and ERK1/2 phosphorylation. Moreover, the scaffold protein JIP4 was involved in adipogenesis in C3H10T1/2 cells by interacting with NRP1. Furthermore, overexpression of non-GAG-modifiable NRP1 mutant (S612A) greatly promoted adipogenic differentiation, accompanied by upregulation of the phosphorylated Akt and ERK1/2. Taken together, these results indicate that NRP1 is a key regulator that promotes adipogenesis in C3H10T1/2 cells by interacting with JIP4 and activating the Akt and ERK1/2 pathway. Non-GAG-modifiable NRP1 mutant (S612A) accelerates the process of adipogenic differentiation, suggesting that GAG glycosylation is a negative post-translational modification of NRP1 in adipogenic differentiation.
Collapse
Affiliation(s)
- Yaqiong Yu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, China
| | - Yoko Uchida-Fukuhara
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Yao Weng
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Yuhan He
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, China
| | - Mika Ikegame
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Ziyi Wang
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Kaya Yoshida
- Department of Oral Healthcare Education, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan
| | - Hirohiko Okamura
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Lihong Qiu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, China
| |
Collapse
|
17
|
Mas-Rosario JA, Medor JD, Jeffway MI, Martínez-Montes JM, Farkas ME. Murine macrophage-based iNos reporter reveals polarization and reprogramming in the context of breast cancer. Front Oncol 2023; 13:1151384. [PMID: 37091169 PMCID: PMC10113556 DOI: 10.3389/fonc.2023.1151384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/23/2023] [Indexed: 04/25/2023] Open
Abstract
As part of the first line of defense against pathogens, macrophages possess the ability to differentiate into divergent phenotypes with varying functions. The process by which these cells change their characteristics, commonly referred to as macrophage polarization, allows them to change into broadly pro-inflammatory (M1) or anti-inflammatory (M2) subtypes, and depends on the polarizing stimuli. Deregulation of macrophage phenotypes can result in different pathologies or affect the nature of some diseases, such as cancer and atherosclerosis. Therefore, a better understanding of macrophage phenotype conversion in relevant models is needed to elucidate its potential roles in disease. However, there are few existing probes to track macrophage changes in multicellular environments. In this study, we generated an eGFP reporter cell line based on inducible nitric oxide synthase (iNos) promoter activity in RAW264.7 cells (RAW:iNos-eGFP). iNos is associated with macrophage activation to pro-inflammatory states and decreases in immune-suppressing ones. We validated the fidelity of the reporter for iNos following cytokine-mediated polarization and confirmed that reporter and parental cells behaved similarly. RAW:iNos-eGFP cells were then used to track macrophage responses in different in vitro breast cancer models, and their re-education from anti- to pro-inflammatory phenotypes via a previously reported pyrimido(5,4-b)indole small molecule, PBI1. Using two mouse mammary carcinoma cell lines, 4T1 and EMT6, effects on macrophages were assessed via conditioned media, two-dimensional/monolayer co-culture, and three-dimensional spheroid models. While conditioned media derived from 4T1 or EMT6 cells and monolayer co-cultures of each cancer cell line with RAW:iNos-eGFP cells all resulted in decreased fluorescence, the trends and extents of effects differed. We also observed decreases in iNos-eGFP signal in the macrophages in co-culture assays with 4T1- or EMT6-based spheroids. We then showed that iNos production is enhanced in these cancer models using PBI1, tracking increased fluorescence. Collectively, this work demonstrates that this reporter-based approach provides a facile means to study macrophage responses in complex, multicomponent environments. Beyond the initial studies presented here, this platform can be used with a variety of in vitro models and extended to in vivo applications with intravital imaging.
Collapse
Affiliation(s)
- Javier A. Mas-Rosario
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Ahmerst, MA, United States
| | - Josue D. Medor
- Department of Biochemistry & Molecular Biology, University of Massachusetts Amherst, Ahmerst, MA, United States
| | - Mary I. Jeffway
- Department of Biochemistry & Molecular Biology, University of Massachusetts Amherst, Ahmerst, MA, United States
| | - José M. Martínez-Montes
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Ahmerst, MA, United States
| | - Michelle E. Farkas
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Ahmerst, MA, United States
- Department of Chemistry, University of Massachusetts Amherst, Ahmerst, MA, United States
| |
Collapse
|
18
|
Yao J, Wu D, Qiu Y. Adipose tissue macrophage in obesity-associated metabolic diseases. Front Immunol 2022; 13:977485. [PMID: 36119080 PMCID: PMC9478335 DOI: 10.3389/fimmu.2022.977485] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Adipose tissue macrophage (ATM) has been appreciated for its critical contribution to obesity-associated metabolic diseases in recent years. Here, we discuss the regulation of ATM on both metabolic homeostatsis and dysfunction. In particular, the macrophage polarization and recruitment as well as the crosstalk between ATM and adipocyte in thermogenesis, obesity, insulin resistance and adipose tissue fibrosis have been reviewed. A better understanding of how ATM regulates adipose tissue remodeling may provide novel therapeutic strategies against obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Jingfei Yao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Dongmei Wu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yifu Qiu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- *Correspondence: Yifu Qiu,
| |
Collapse
|
19
|
Huang T, Song J, Gao J, Cheng J, Xie H, Zhang L, Wang YH, Gao Z, Wang Y, Wang X, He J, Liu S, Yu Q, Zhang S, Xiong F, Zhou Q, Wang CY. Adipocyte-derived kynurenine promotes obesity and insulin resistance by activating the AhR/STAT3/IL-6 signaling. Nat Commun 2022; 13:3489. [PMID: 35715443 PMCID: PMC9205899 DOI: 10.1038/s41467-022-31126-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
Aberrant amino acid metabolism is a common event in obesity. Particularly, subjects with obesity are characterized by the excessive plasma kynurenine (Kyn). However, the primary source of Kyn and its impact on metabolic syndrome are yet to be fully addressed. Herein, we show that the overexpressed indoleamine 2,3-dioxygenase 1 (IDO1) in adipocytes predominantly contributes to the excessive Kyn, indicating a central role of adipocytes in Kyn metabolism. Depletion of Ido1 in adipocytes abrogates Kyn accumulation, protecting mice against obesity. Mechanistically, Kyn impairs lipid homeostasis in adipocytes via activating the aryl hydrocarbon receptor (AhR)/Signal transducer and activator of transcription 3 /interleukin-6 signaling. Genetic ablation of AhR in adipocytes abolishes the effect of Kyn. Moreover, supplementation of vitamin B6 ameliorated Kyn accumulation, protecting mice from obesity. Collectively, our data support that adipocytes are the primary source of increased circulating Kyn, while elimination of accumulated Kyn could be a viable strategy against obesity.
Collapse
Affiliation(s)
- Teng Huang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Song
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Gao
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Cheng
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xie
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Zhang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Han Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Gao
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohui Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinhan He
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shiwei Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Qilin Yu
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Cong-Yi Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
20
|
Wu S, Huang Y, Huang X, Dai X. Lipopolysaccharide Accelerates Neuropilin-1 Protein Degradation by Activating the Large GTPase Dynamin-1 in Macrophages. Inflammation 2022; 45:1162-1173. [DOI: 10.1007/s10753-021-01610-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/23/2022]
|
21
|
Multiomics and digital monitoring during lifestyle changes reveal independent dimensions of human biology and health. Cell Syst 2021; 13:241-255.e7. [PMID: 34856119 DOI: 10.1016/j.cels.2021.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 09/15/2021] [Accepted: 11/09/2021] [Indexed: 01/04/2023]
Abstract
We explored opportunities for personalized and predictive health care by collecting serial clinical measurements, health surveys, genomics, proteomics, autoantibodies, metabolomics, and gut microbiome data from 96 individuals who participated in a data-driven health coaching program over a 16-month period with continuous digital monitoring of activity and sleep. We generated a resource of >20,000 biological samples from this study and a compendium of >53 million primary data points for 558,032 distinct features. Multiomics factor analysis revealed distinct and independent molecular factors linked to obesity, diabetes, liver function, cardiovascular disease, inflammation, immunity, exercise, diet, and hormonal effects. For example, ethinyl estradiol, a common oral contraceptive, produced characteristic molecular and physiological effects, including increased levels of inflammation and impact on thyroid, cortisol levels, and pulse, that were distinct from other sources of variability observed in our study. In total, this work illustrates the value of combining deep molecular and digital monitoring of human health. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
|
22
|
A widespread viral entry mechanism: The C-end Rule motif-neuropilin receptor interaction. Proc Natl Acad Sci U S A 2021; 118:2112457118. [PMID: 34772761 PMCID: PMC8670474 DOI: 10.1073/pnas.2112457118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 01/31/2023] Open
Abstract
Many phylogenetically distant animal viruses, including the new coronavirus severe acute respiratory syndrome coronavirus 2, have surface proteins with polybasic sites that are cleaved by host furin and furin-like proteases. Other than priming certain viral surface proteins for fusion, cleavage generates a carboxy-terminal RXXR sequence. This C-end Rule (CendR) motif is known to bind to neuropilin (NRP) receptors on the cell surface. NRPs are ubiquitously expressed, pleiotropic cell surface receptors with important roles in growth factor signaling, vascular biology, and neurobiology, as well as immune homeostasis and activation. The CendR–NRP receptor interaction promotes endocytic internalization and tissue spreading of different cargo, including viral particles. We propose that the interaction between viral surface proteins and NRPs plays an underappreciated and prevalent role in the transmission and pathogenesis of diverse viruses and represents a promising broad-spectrum antiviral target.
Collapse
|
23
|
Catrysse L, Maes B, Mehrotra P, Martens A, Hoste E, Martens L, Maueröder C, Remmerie A, Bujko A, Slowicka K, Sze M, Vikkula H, Ghesquière B, Scott CL, Saeys Y, van de Sluis B, Ravichandran K, Janssens S, van Loo G. A20 deficiency in myeloid cells protects mice from diet-induced obesity and insulin resistance due to increased fatty acid metabolism. Cell Rep 2021; 36:109748. [PMID: 34551300 DOI: 10.1016/j.celrep.2021.109748] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/04/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022] Open
Abstract
Obesity-induced inflammation is a major driving force in the development of insulin resistance, type 2 diabetes (T2D), and related metabolic disorders. During obesity, macrophages accumulate in the visceral adipose tissue, creating a low-grade inflammatory environment. Nuclear factor κB (NF-κB) signaling is a central coordinator of inflammatory responses and is tightly regulated by the anti-inflammatory protein A20. Here, we find that myeloid-specific A20-deficient mice are protected from diet-induced obesity and insulin resistance despite an inflammatory environment in their metabolic tissues. Macrophages lacking A20 show impaired mitochondrial respiratory function and metabolize more palmitate both in vitro and in vivo. We hypothesize that A20-deficient macrophages rely more on palmitate oxidation and metabolize the fat present in the diet, resulting in a lean phenotype and protection from metabolic disease. These findings reveal a role for A20 in regulating macrophage immunometabolism.
Collapse
Affiliation(s)
- Leen Catrysse
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Bastiaan Maes
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, B-9052 Ghent, Belgium
| | - Parul Mehrotra
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Arne Martens
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Esther Hoste
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Liesbet Martens
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Christian Maueröder
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Anneleen Remmerie
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Anna Bujko
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Karolina Slowicka
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Mozes Sze
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Hanna Vikkula
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Bart Ghesquière
- Metabolomics Core Facility, VIB Center for Cancer Biology, VIB, B-3000 Leuven, Belgium
| | - Charlotte L Scott
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Yvan Saeys
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Applied Mathematics, Computer Science and Statistics, Ghent University, B-9052 Ghent, Belgium
| | - Bart van de Sluis
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, NL- 9713 Groningen, the Netherlands
| | - Kodi Ravichandran
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium; Center for Cell Clearance and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Sophie Janssens
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, B-9052 Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium.
| |
Collapse
|
24
|
Chuckran CA, Liu C, Bruno TC, Workman CJ, Vignali DA. Neuropilin-1: a checkpoint target with unique implications for cancer immunology and immunotherapy. J Immunother Cancer 2021; 8:jitc-2020-000967. [PMID: 32675311 PMCID: PMC7368550 DOI: 10.1136/jitc-2020-000967] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Checkpoint blockade immunotherapy established a new paradigm in cancer treatment: for certain patients curative treatment requires immune reinvigoration. Despite this monumental advance, only 20%–30% of patients achieve an objective response to standard of care immunotherapy, necessitating the consideration of alternative targets. Optimal strategies will not only stimulate CD8+ T cells, but concomitantly modulate immunosuppressive cells in the tumor microenvironment (TME), most notably regulatory T cells (Treg cells). In this context, the immunoregulatory receptor Neuropilin-1 (NRP1) is garnering renewed attention as it reinforces intratumoral Treg cell function amidst inflammation in the TME. Loss of NRP1 on Treg cells in mouse models restores antitumor immunity without sacrificing peripheral tolerance. Enrichment of NRP1+ Treg cells is observed in patients across multiple malignancies with cancer, both intratumorally and in peripheral sites. Thus, targeting NRP1 may safely undermine intratumoral Treg cell fitness, permitting enhanced inflammatory responses with existing immunotherapies. Furthermore, NRP1 has been recently found to modulate tumor-specific CD8+ T cell responses. Emerging data suggest that NRP1 restricts CD8+ T cell reinvigoration in response to checkpoint inhibitors, and more importantly, acts as a barrier to the long-term durability of CD8+ T cell-mediated tumor immunosurveillance. These novel and distinct regulatory mechanisms present an exciting therapeutic opportunity. This review will discuss the growing literature on NRP1-mediated immune modulation which provides a strong rationale for categorizing NRP1 as both a key checkpoint in the TME as well as an immunotherapeutic target with promise either alone or in combination with current standard of care therapeutic regimens.
Collapse
Affiliation(s)
- Christopher A Chuckran
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Tumor Microenvironment Center and the Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chang Liu
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Tumor Microenvironment Center and the Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Tumor Microenvironment Center and the Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Creg J Workman
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Tumor Microenvironment Center and the Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Dario Aa Vignali
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA .,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
25
|
Zhen Y, Shu W, Hou X, Wang Y. Innate Immune System Orchestrates Metabolic Homeostasis and Dysfunction in Visceral Adipose Tissue During Obesity. Front Immunol 2021; 12:702835. [PMID: 34421909 PMCID: PMC8377368 DOI: 10.3389/fimmu.2021.702835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/22/2021] [Indexed: 01/22/2023] Open
Abstract
Arising incidence of metabolic disorders and related diseases caused by obesity is a global health concern. Elucidating the role of the immune system in this process will help to understand the related mechanisms and develop treatment strategies. Here, we have focused on innate immune cells in visceral adipose tissue (VAT) and summarized the roles of these cells in maintaining the homeostasis of VAT. Furthermore, this review reveals the importance of quantitative and functional changes of innate immune cells when the metabolic microenvironment changes due to obesity or excess lipids, and confirms that these changes eventually lead to the occurrence of chronic inflammation and metabolic diseases of VAT. Two perspectives are reviewed, which include sequential changes in various innate immune cells in the steady state of VAT and its imbalance during obesity. Cross-sectional interactions between various innate immune cells at the same time point are also reviewed. Through delineation of a comprehensive perspective of VAT homeostasis in obesity-induced chronic inflammation, and ultimately metabolic dysfunction and disease, we expect to clarify the complex interactive networks among distinct cell populations and propose that these interactions should be taken into account in the development of biotherapeutic strategies.
Collapse
Affiliation(s)
- Yu Zhen
- Department of Dermatology, The First Hospital of Jilin University, Changchun, China
| | - Wentao Shu
- Department of Biobank, Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Xintong Hou
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China.,Institute of Immunology, Jilin University, Changchun, China
| | - Yinan Wang
- Department of Biobank, Division of Clinical Research, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Diaz-Marin R, Crespo-Garcia S, Wilson AM, Buscarlet M, Dejda A, Fournier F, Juneau R, Alquier T, Sapieha P. Myeloid-resident neuropilin-1 influences brown adipose tissue in obesity. Sci Rep 2021; 11:15767. [PMID: 34344941 PMCID: PMC8333363 DOI: 10.1038/s41598-021-95064-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 07/20/2021] [Indexed: 11/09/2022] Open
Abstract
The beneficial effects of brown adipose tissue (BAT) on obesity and associated metabolic diseases are mediated through its capacity to dissipate energy as heat. While immune cells, such as tissue-resident macrophages, are known to influence adipose tissue homeostasis, relatively little is known about their contribution to BAT function. Here we report that neuropilin-1 (NRP1), a multiligand single-pass transmembrane receptor, is highly expressed in BAT-resident macrophages. During diet-induced obesity (DIO), myeloid-resident NRP1 influences interscapular BAT mass, and consequently vascular morphology, innervation density and ultimately core body temperature during cold exposure. Thus, NRP1-expressing myeloid cells contribute to the BAT homeostasis and potentially its thermogenic function in DIO.
Collapse
Affiliation(s)
- Roberto Diaz-Marin
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, 5415 Assumption Boulevard, Montréal, QC, H1T 2M4, Canada
| | - Sergio Crespo-Garcia
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, 5415 Assumption Boulevard, Montréal, QC, H1T 2M4, Canada
| | - Ariel M Wilson
- Department of Ophthalmology, Maisonneuve-Rosemont Research Centre, Université de Montréal, Montréal, QC, H1T2M4, Canada
| | - Manuel Buscarlet
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, 5415 Assumption Boulevard, Montréal, QC, H1T 2M4, Canada
| | - Agnieszka Dejda
- Department of Ophthalmology, Maisonneuve-Rosemont Research Centre, Université de Montréal, Montréal, QC, H1T2M4, Canada
| | - Frédérik Fournier
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, 5415 Assumption Boulevard, Montréal, QC, H1T 2M4, Canada
| | - Rachel Juneau
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, 5415 Assumption Boulevard, Montréal, QC, H1T 2M4, Canada
| | - Thierry Alquier
- Montreal Diabetes Research Centre and Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue Saint-Denis, Montréal, QC, H2X0A9, Canada
| | - Przemyslaw Sapieha
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, 5415 Assumption Boulevard, Montréal, QC, H1T 2M4, Canada. .,Department of Ophthalmology, Maisonneuve-Rosemont Research Centre, Université de Montréal, Montréal, QC, H1T2M4, Canada.
| |
Collapse
|
27
|
Andriessen EMMA, Binet F, Fournier F, Hata M, Dejda A, Mawambo G, Crespo‐Garcia S, Pilon F, Buscarlet M, Beauchemin K, Bougie V, Cumberlidge G, Wilson AM, Bourgault S, Rezende FA, Beaulieu N, Delisle J, Sapieha P. Myeloid-resident neuropilin-1 promotes choroidal neovascularization while mitigating inflammation. EMBO Mol Med 2021; 13:e11754. [PMID: 33876574 PMCID: PMC8103107 DOI: 10.15252/emmm.201911754] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/11/2021] [Accepted: 02/24/2021] [Indexed: 01/03/2023] Open
Abstract
Age-related macular degeneration (AMD) in its various forms is a leading cause of blindness in industrialized countries. Here, we provide evidence that ligands for neuropilin-1 (NRP1), such as Semaphorin 3A and VEGF-A, are elevated in the vitreous of patients with AMD at times of active choroidal neovascularization (CNV). We further demonstrate that NRP1-expressing myeloid cells promote and maintain CNV. Expression of NRP1 on cells of myeloid lineage is critical for mitigating production of inflammatory factors such as IL6 and IL1β. Therapeutically trapping ligands of NRP1 with an NRP1-derived trap reduces CNV. Collectively, our findings identify a role for NRP1-expressing myeloid cells in promoting pathological angiogenesis during CNV and introduce a therapeutic approach to counter neovascular AMD.
Collapse
Affiliation(s)
| | - François Binet
- SemaThera Inc.MontrealQCCanada
- Department of OphthalmologyUniversity of MontrealMontrealQCCanada
| | - Frédérik Fournier
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontrealQCCanada
| | - Masayuki Hata
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontrealQCCanada
| | - Agnieszka Dejda
- Department of OphthalmologyUniversity of MontrealMontrealQCCanada
| | - Gaëlle Mawambo
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontrealQCCanada
| | - Sergio Crespo‐Garcia
- Department of OphthalmologyUniversity of MontrealMontrealQCCanada
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontrealQCCanada
| | - Frédérique Pilon
- Department of OphthalmologyUniversity of MontrealMontrealQCCanada
| | - Manuel Buscarlet
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontrealQCCanada
| | | | | | | | - Ariel M Wilson
- Department of OphthalmologyUniversity of MontrealMontrealQCCanada
| | - Steve Bourgault
- Department of ChemistryUniversité du Québec à MontréalMontrealQCCanada
| | - Flavio A Rezende
- Department of OphthalmologyUniversity of MontrealMontrealQCCanada
| | | | - Jean‐Sébastien Delisle
- Department of MedicineMaisonneuve‐Rosemont Hospital Research CentreUniversity of MontrealMontrealQCCanada
| | - Przemyslaw Sapieha
- Department of Biomedical SciencesUniversity of MontrealMontrealQCCanada
- SemaThera Inc.MontrealQCCanada
- Department of OphthalmologyUniversity of MontrealMontrealQCCanada
- Department of Biochemistry and Molecular MedicineUniversity of MontrealMontrealQCCanada
| |
Collapse
|
28
|
Nobs SP, Kopf M. Tissue-resident macrophages: guardians of organ homeostasis. Trends Immunol 2021; 42:495-507. [PMID: 33972166 DOI: 10.1016/j.it.2021.04.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Tissue-resident macrophages (MTR) have recently emerged as a key rheostat capable of regulating the balance between organ health and disease. In most organs, ontogenetically and functionally distinct macrophage subsets fulfill a plethora of functions specific to their tissue environment. In this review, we summarize recent findings regarding the ontogeny and functions of macrophage populations in different mammalian tissues, describing how these cells regulate tissue homeostasis and how they can contribute to inflammation. Furthermore, we highlight new developments concerning certain general principles of tissue macrophage biology, including the importance of metabolism for understanding macrophage activation states and the influence of intrinsic and extrinsic factors on macrophage metabolic control. We also shed light on certain open questions in the field and how answering these might pave the way for tissue-specific therapeutic approaches.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Department of Immunology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
29
|
Balestrieri B, Granata F, Loffredo S, Petraroli A, Scalia G, Morabito P, Cardamone C, Varricchi G, Triggiani M. Phenotypic and Functional Heterogeneity of Low-Density and High-Density Human Lung Macrophages. Biomedicines 2021; 9:biomedicines9050505. [PMID: 34064389 PMCID: PMC8147777 DOI: 10.3390/biomedicines9050505] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pulmonary macrophages are a highly heterogeneous cell population distributed in different lung compartments. METHODS We separated two subpopulations of macrophages from human lung parenchyma according to flotation over density gradients. RESULTS Two-thirds 65.4% of the lung macrophages have a density between 1.065 and 1.078 (high-density macrophages: HDMs), and the remaining one-third (34.6) had a density between 1.039 and 1.052 (low-density macrophages: LDMs). LDMs had a larger area (691 vs. 462 μm2) and cell perimeter (94 vs. 77 μm) compared to HDMs. A significantly higher percentage of HDMs expressed CD40, CD45, and CD86 compared to LDMs. In contrast, a higher percentage of LDMs expressed the activation markers CD63 and CD64. The release of TNF-α, IL-6, IL-10 and IL-12 induced by lipopolysaccharide (LPS) was significantly higher in HDMs than in LDMs. CONCLUSION The human lung contains two subpopulations of macrophages that differ in buoyancy, morphometric parameters, surface marker expression and response to LPS. These subpopulations of macrophages probably play distinct roles in lung inflammation and immune responses.
Collapse
Affiliation(s)
- Barbara Balestrieri
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Correspondence: (B.B.); (F.G.)
| | - Francescopaolo Granata
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Center of Excellence, World Allergy Organization (WAO), 80131 Naples, Italy
- Correspondence: (B.B.); (F.G.)
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Center of Excellence, World Allergy Organization (WAO), 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Angelica Petraroli
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Center of Excellence, World Allergy Organization (WAO), 80131 Naples, Italy
| | - Giulia Scalia
- Clinical and Experimental Cytometry Unit, CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy;
| | - Paolo Morabito
- Laboratory of Clinical Pathology, A. Cardarelli Hospital, 80131 Naples, Italy;
| | - Chiara Cardamone
- Division of Allergy and Clinical Immunology, University of Salerno, 84084 Fisciano, Italy; (C.C.); (M.T.)
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (S.L.); (A.P.); (G.V.)
- Center of Excellence, World Allergy Organization (WAO), 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, 84084 Fisciano, Italy; (C.C.); (M.T.)
| |
Collapse
|
30
|
Keck S, Galati-Fournier V, Kym U, Moesch M, Usemann J, Müller I, Subotic U, Tharakan SJ, Krebs T, Stathopoulos E, Schmittenbecher P, Cholewa D, Romero P, Reingruber B, Bruder E, Group NS, Holland-Cunz S. Lack of Mucosal Cholinergic Innervation Is Associated With Increased Risk of Enterocolitis in Hirschsprung's Disease. Cell Mol Gastroenterol Hepatol 2021; 12:507-545. [PMID: 33741501 PMCID: PMC8258990 DOI: 10.1016/j.jcmgh.2021.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Hirschsprung's disease (HSCR) is a congenital intestinal motility disorder defined by the absence of enteric neuronal cells (ganglia) in the distal gut. The development of HSCR-associated enterocolitis remains a life-threatening complication. Absence of enteric ganglia implicates innervation of acetylcholine-secreting (cholinergic) nerve fibers. Cholinergic signals have been reported to control excessive inflammation, but the impact on HSCR-associated enterocolitis is unknown. METHODS We enrolled 44 HSCR patients in a prospective multicenter study and grouped them according to their degree of colonic mucosal acetylcholinesterase-positive innervation into low-fiber and high-fiber patient groups. The fiber phenotype was correlated with the tissue cytokine profile as well as immune cell frequencies using Luminex analysis and fluorescence-activated cell sorting analysis of colonic tissue and immune cells. Using confocal immunofluorescence microscopy, macrophages were identified in close proximity to nerve fibers and characterized by RNA-seq analysis. Microbial dysbiosis was analyzed in colonic tissue using 16S-rDNA gene sequencing. Finally, the fiber phenotype was correlated with postoperative enterocolitis manifestation. RESULTS The presence of mucosal nerve fiber innervation correlated with reduced T-helper 17 cytokines and cell frequencies. In high-fiber tissue, macrophages co-localized with nerve fibers and expressed significantly less interleukin 23 than macrophages from low-fiber tissue. HSCR patients lacking mucosal nerve fibers showed microbial dysbiosis and had a higher incidence of postoperative enterocolitis. CONCLUSIONS The mucosal fiber phenotype might serve as a prognostic marker for enterocolitis development in HSCR patients and may offer an approach to personalized patient care and new therapeutic options.
Collapse
Affiliation(s)
- Simone Keck
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland.
| | - Virginie Galati-Fournier
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| | - Urs Kym
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| | - Michèle Moesch
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| | - Jakob Usemann
- Department of Pediatric Pulmonology, University Children's Hospital Basel (UKBB), Basel, and Division of Respiratory Medicine, University Children's Hospital Zurich, Zurich, Switzerland
| | - Isabelle Müller
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| | - Ulrike Subotic
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland; Department of Pediatric Surgery, University Children's Hospital Zurich, Zurich, Switzerland
| | - Sasha J Tharakan
- Department of Pediatric Surgery, University Children's Hospital Zurich, Zurich, Switzerland
| | - Thomas Krebs
- Department of Pediatric Surgery, Children's Hospital of Eastern Switzerland, St Gallen, Switzerland
| | - Eleuthere Stathopoulos
- Department of Pediatric Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | | | - Dietmar Cholewa
- Department of Pediatric Surgery, University Hospital of Bern, Bern, Switzerland
| | - Philipp Romero
- Department of Pediatric Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Bertram Reingruber
- Department of Pediatric Surgery, Florence Nightingale Hospital, Düsseldorf, Germany
| | - Elisabeth Bruder
- Institute for Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Nig Study Group
- NIG Study Group, Lausanne, Switzerland; Department of Pathology, University Hospital of Lausanne (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Stefan Holland-Cunz
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| |
Collapse
|
31
|
Cristinziano L, Poto R, Criscuolo G, Ferrara AL, Galdiero MR, Modestino L, Loffredo S, de Paulis A, Marone G, Spadaro G, Varricchi G. IL-33 and Superantigenic Activation of Human Lung Mast Cells Induce the Release of Angiogenic and Lymphangiogenic Factors. Cells 2021; 10:cells10010145. [PMID: 33445787 PMCID: PMC7828291 DOI: 10.3390/cells10010145] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 02/06/2023] Open
Abstract
Human lung mast cells (HLMCs) express the high-affinity receptor FcεRI for IgE and are strategically located in different compartments of human lung, where they play a role in several inflammatory disorders and cancer. Immunoglobulin superantigens (e.g., protein A of Staphylococcus aureus and protein L of Peptostreptococcus magnus) bind to the variable regions of either the heavy (VH3) or light chain (κ) of IgE. IL-33 is a cytokine expressed by epithelial cells that exerts pleiotropic functions in the lung. The present study investigated whether immunoglobulin superantigens protein A and protein L and IL-33 caused the release of inflammatory (histamine), angiogenic (VEGF-A) and lymphangiogenic (VEGF-C) factors from HLMCs. The results show that protein A and protein L induced the rapid (30 min) release of preformed histamine from HLMCs. By contrast, IL-33 did not induce the release of histamine from lung mast cells. Prolonged incubation (12 h) of HLMCs with superantigens and IL-33 induced the release of VEGF-A and VEGF-C. Preincubation with IL-33 potentiated the superantigenic release of histamine, angiogenic and lymphangiogenic factors from HLMCs. Our results suggest that IL-33 might enhance the inflammatory, angiogenic and lymphangiogenic activities of lung mast cells in pulmonary disorders.
Collapse
Affiliation(s)
- Leonardo Cristinziano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Luca Modestino
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
32
|
Abstract
Neuropilin-1 (NRP-1), a member of a family of signaling proteins, was shown to serve as an entry factor and potentiate SARS Coronavirus 2 (SARS-CoV-2) infectivity in vitro. This cell surface receptor with its disseminated expression is important in angiogenesis, tumor progression, viral entry, axonal guidance, and immune function. NRP-1 is implicated in several aspects of a SARS-CoV-2 infection including possible spread through the olfactory bulb and into the central nervous system and increased NRP-1 RNA expression in lungs of severe Coronavirus Disease 2019 (COVID-19). Up-regulation of NRP-1 protein in diabetic kidney cells hint at its importance in a population at risk of severe COVID-19. Involvement of NRP-1 in immune function is compelling, given the role of an exaggerated immune response in disease severity and deaths due to COVID-19. NRP-1 has been suggested to be an immune checkpoint of T cell memory. It is unknown whether involvement and up-regulation of NRP-1 in COVID-19 may translate into disease outcome and long-term consequences, including possible immune dysfunction. It is prudent to further research NRP-1 and its possibility of serving as a therapeutic target in SARS-CoV-2 infections. We anticipate that widespread expression, abundance in the respiratory and olfactory epithelium, and the functionalities of NRP-1 factor into the multiple systemic effects of COVID-19 and challenges we face in management of disease and potential long-term sequelae.
Collapse
Affiliation(s)
- Bindu S. Mayi
- Department of Basic Sciences, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, Florida, United States of America
- * E-mail:
| | - Jillian A. Leibowitz
- Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, Florida, United States of America
| | - Arden T. Woods
- Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, Florida, United States of America
| | - Katherine A. Ammon
- USF Morsani College of Medicine, Tampa, Florida, United States of America
| | - Alphonse E. Liu
- Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, Florida, United States of America
| | - Aarti Raja
- Department of Biological Sciences, Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| |
Collapse
|
33
|
Yoshitake R, Hirose Y, Murosaki S, Matsuzaki G. Heat-killed Lactobacillus plantarum L-137 attenuates obesity and associated metabolic abnormalities in C57BL/6 J mice on a high-fat diet. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2020; 40:84-91. [PMID: 33996364 PMCID: PMC8099634 DOI: 10.12938/bmfh.2020-040] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/24/2020] [Indexed: 12/31/2022]
Abstract
Heat-killed Lactobacillus plantarum L-137 (HK L-137) has anti-allergic, antitumor, and antiviral effects in mice, as well as an anti-inflammatory effect in rats with metabolic syndrome through regulation of immunity. To evaluate the influence of HK L-137 on chronic inflammation in mice with diet-induced obesity, C57BL/6 J mice were fed a normal diet (16% of energy as fat) or a high-fat diet (62% of energy as fat) with or without 0.002% HK L-137 for 4 to 20 weeks. It was found that HK L-137 supplementation alleviated weight gain and elevation of plasma glucose, cholesterol, alanine aminotransferase, and aspartate transaminase levels in mice with diet-induced obesity. Expression of several inflammation-related genes, including F4/80, CD11c, and IL-1β, in the epididymal adipose tissue of these mice was significantly downregulated by HK L-137. In addition, plasma levels of lipopolysaccharide-binding protein, a marker of endotoxemia, tended to be decreased by administration of HK L-137. These findings suggest that HK L-137 supplementation ameliorates obesity-induced metabolic abnormalities and adipose tissue inflammation, possibly through improvement of intestinal permeability.
Collapse
Affiliation(s)
- Rieko Yoshitake
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, University of The Ryukyus, Nishihara, Okinawa 903-0213, Japan
| | - Yoshitaka Hirose
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, University of The Ryukyus, Nishihara, Okinawa 903-0213, Japan
| | - Shinji Murosaki
- Nihon Pharmaceutical University, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Goro Matsuzaki
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, University of The Ryukyus, Nishihara, Okinawa 903-0213, Japan
| |
Collapse
|
34
|
Li Y, Yun K, Mu R. A review on the biology and properties of adipose tissue macrophages involved in adipose tissue physiological and pathophysiological processes. Lipids Health Dis 2020; 19:164. [PMID: 32646451 PMCID: PMC7350193 DOI: 10.1186/s12944-020-01342-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity exhibits a correlation with metabolic inflammation and endoplasmic reticulum stress, promoting the progression of metabolic disease such as diabetes, hyperlipidemia, hyperuricemia and so on. Adipose tissue macrophages (ATMs) are central players in obesity-associated inflammation and metabolic diseases. Macrophages are involved in lipid and energy metabolism and mitochondrial function in adipocytes. Macrophage polarization is accompanied by metabolic shifting between glycolysis and mitochondrial oxidative phosphorylation. Here, this review focuses on macrophage metabolism linked to functional phenotypes with an emphasis on macrophage polarization in adipose tissue physiological and pathophysiological processes. In particular, the interplay between ATMs and adipocytes in energy metabolism, glycolysis, OXPHOS, iron handing and even interactions with the nervous system have been reviewed. Overall, the understanding of protective and pathogenic roles of ATMs in adipose tissue can potentially provide strategies to prevent and treat obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Yunjia Li
- The First Clinical Medicine Faculty, China Medical University, Shenyang, 110001, China
| | - Ke Yun
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Runqing Mu
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
35
|
HIV gp120 Induces the Release of Proinflammatory, Angiogenic, and Lymphangiogenic Factors from Human Lung Mast Cells. Vaccines (Basel) 2020; 8:vaccines8020208. [PMID: 32375243 PMCID: PMC7349869 DOI: 10.3390/vaccines8020208] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Human lung mast cells (HLMCs) express the high-affinity receptor FcεRI for IgE and are involved in chronic pulmonary diseases occurring at high frequency among HIV-infected individuals. Immunoglobulin superantigens bind to the variable regions of either the heavy or light chain of immunoglobulins (Igs). Glycoprotein 120 (gp120) of HIV-1 is a typical immunoglobulin superantigen interacting with the heavy chain, variable 3 (VH3) region of human Igs. The present study investigated whether immunoglobulin superantigen gp120 caused the release of different classes of proinflammatory and immunoregulatory mediators from HLMCs. The results show that gp120 from different clades induced the rapid (30 min) release of preformed mediators (histamine and tryptase) from HLMCs. gp120 also caused the de novo synthesis of cysteinyl leukotriene C4 (LTC4) and prostaglandin D2 (PGD2) from HLMCs. Incubation (6 h) of HLMC with gp120 induced the release of angiogenic (VEGF-A) and lymphangiogenic (VEGF-C) factors from HLMCs. The activating property of gp120 was mediated through the interaction with IgE VH3+ bound to FcεRI. Our data indicate that HIV gp120 is a viral superantigen, which induces the release of different proinflammatory, angiogenic, and lymphangiogenic factors from HLMCs. These observations could contribute to understanding, at least in part, the pathophysiology of chronic pulmonary diseases in HIV-infected individuals.
Collapse
|
36
|
Bolus WR, Hasty AH. Contributions of innate type 2 inflammation to adipose function. J Lipid Res 2019; 60:1698-1709. [PMID: 29891508 PMCID: PMC6795076 DOI: 10.1194/jlr.r085993] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/25/2018] [Indexed: 12/17/2022] Open
Abstract
A critical contributor to the health consequences of the obesity epidemic is dysregulated adipose tissue (AT) homeostasis. While white, brown, and beige AT function is altered in obesity-related disease, white AT is marked by progressive inflammation and adipocyte dysfunction and has been the focus of extensive "immunometabolism" research in the past decade. The exact triggering events initiating and sustaining AT inflammation are still under study, but it has been shown that reducing inflammation improves insulin action in AT. Scientific efforts seeking interventions to mitigate obesity-associated AT inflammation continue, and many groups are now determining how lean healthy AT homeostasis is maintained in order to leverage these mechanisms as therapeutic targets. Such studies have revealed that an elaborate network of immune cells, cytokines, and other cellular mediators coordinate AT function. Recent studies elucidated the involvement of the innate immune system in AT homeostasis (e.g., beiging and insulin sensitivity), including M2-like macrophages, eosinophils, innate lymphoid type 2 cells, and several others. In this review, we summarize the existing literature on innate type 2 inflammation in AT; additionally, we draw attention to areas of debate where seemingly conflicting data promises to yield more surprising and elegant biology as studies continue to dissect AT physiology.
Collapse
Affiliation(s)
- W Reid Bolus
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville TN 37232
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville TN 37232
- Veterans Affairs Tennessee Valley Healthcare System, Nashville TN 37212
| |
Collapse
|
37
|
Savedoroudi P, Bennike TB, Kastaniegaard K, Talebpour M, Ghassempour A, Stensballe A. Serum proteome changes and accelerated reduction of fat mass after laparoscopic gastric plication in morbidly obese patients. J Proteomics 2019; 203:103373. [PMID: 31054967 DOI: 10.1016/j.jprot.2019.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
Laparoscopic Gastric Plication (LGP) is a relatively new bariatric surgical procedure which no part of the stomach is removed. It is not clearly understood how LGP leads to fatty tissue reduction. We aimed to investigate the impact of LGP on serum proteome and understand molecular mechanisms of LGP-induced weight loss post-surgery. A Prospective observational study of 16 obese individuals who underwent LGP was performed. A Label-free quantitative shotgun proteomics approach was used to compare serum proteome of subjects before surgery with serum of the same individuals 1 to 2 months post-surgery (T1) and 4 to 5 months post-surgery (T2). The proteome analysis revealed that 48 proteins were differentially regulated between pre-surgery and T1, and seven proteins between pre-surgery and T2 of which six proteins were shared between the two timepoints. Among differentially regulated proteins, four proteins (SRGN, FETUB, LCP1 and CFP) have not previously been described in the context of BMI/weight loss. Despite few differences following LGP, most regulated serum proteins are in accordance with alternative weight loss procedures. Pathway analysis revealed changes to lipid- and inflammatory pathways, including PPARα/RXRα, LXR/RXR and FXR/RXR activation, especially at T1. At T2, the pathways related to inflammation and immune system are most affected. SIGNIFICANCE: Among the available clinical therapies for morbid obesity, bariatric surgery is considered as the most effective approach to achieve long-term weight loss, alongside a significant improvement in metabolic syndrome. However, very little is known about the underlying mechanism associated with significant weight loss post-surgery. Understanding such mechanisms could lead to development of safer non-surgical weight loss approaches. We here present the first analysis of the impact of LGP on the serum proteome, to bring new insights into the underlying molecular mechanism. Our findings indicate that LGP has a comprehensive systemic effect based on the blood serum proteome profile which might account for accelerated reduction of fat mass after surgery, thus, food restriction is not the only reason for weight loss following this unique surgical approach. As secretory regions of the stomach are preserved in LGP and it is associated with minimal physiological and anatomical changes, the findings are of high importance in the field of bariatric surgery and weight loss.
Collapse
Affiliation(s)
- Parisa Savedoroudi
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran; Department of Health Science and Technology, Aalborg University, Denmark.
| | - Tue Bjerg Bennike
- Department of Health Science and Technology, Aalborg University, Denmark.
| | | | - Mohammad Talebpour
- Laparoscopic Surgery Ward, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Ghassempour
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran.
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Denmark.
| |
Collapse
|
38
|
Varricchi G, Loffredo S, Borriello F, Pecoraro A, Rivellese F, Genovese A, Spadaro G, Marone G. Superantigenic Activation of Human Cardiac Mast Cells. Int J Mol Sci 2019; 20:ijms20081828. [PMID: 31013832 PMCID: PMC6514993 DOI: 10.3390/ijms20081828] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 02/06/2023] Open
Abstract
B cell superantigens, also called immunoglobulin superantigens, bind to the variable regions of either the heavy or light chain of immunoglobulins mirroring the lymphocyte-activating properties of classical T cell superantigens. Protein A of Staphylococcus aureus, protein L of Peptostreptococcus magnus, and gp120 of HIV are typical immunoglobulin superantigens. Mast cells are immune cells expressing the high-affinity receptor for IgE (FcεRI) and are strategically located in the human heart, where they play a role in several cardiometabolic diseases. Here, we investigated whether immunoglobulin superantigens induced the activation of human heart mast cells (HHMCs). Protein A induced the de novo synthesis of cysteinyl leukotriene C4 (LTC4) from HHMCs through the interaction with IgE VH3+ bound to FcεRI. Protein L stimulated the production of prostaglandin D2 (PGD2) from HHMCs through the interaction with κ light chains of IgE. HIV glycoprotein gp120 induced the release of preformed (histamine) and de novo synthesized mediators, such as cysteinyl leukotriene C4 (LTC4), angiogenic (VEGF-A), and lymphangiogenic (VEGF-C) factors by interacting with the VH3 region of IgE. Collectively, our data indicate that bacterial and viral immunoglobulin superantigens can interact with different regions of IgE bound to FcεRI to induce the release of proinflammatory, angiogenic, and lymphangiogenic factors from human cardiac mast cells.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
| | - Francesco Borriello
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, 02115 MA, USA.
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 4NS London, UK.
| | - Arturo Genovese
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council (CNR), 80100 Naples, Italy.
| |
Collapse
|
39
|
Role of innate immune cells in metabolism: from physiology to type 2 diabetes. Semin Immunopathol 2019; 41:531-545. [DOI: 10.1007/s00281-019-00736-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/15/2019] [Indexed: 12/19/2022]
|
40
|
Daryabor G, Kabelitz D, Kalantar K. An update on immune dysregulation in obesity-related insulin resistance. Scand J Immunol 2019; 89:e12747. [PMID: 30593678 DOI: 10.1111/sji.12747] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/22/2018] [Accepted: 12/25/2018] [Indexed: 12/29/2022]
Abstract
Obesity is associated with chronic low-grade inflammation of the adipose tissue (AT) that might develop into systemic inflammation, insulin resistance (IR) and an increased risk of type 2 diabetes mellitus (T2DM) in severe obese rodents and humans. In the lean state, small normal adipocytes and AT macrophages interact with each other to maintain metabolic homeostasis but during obesity, enlarged adipocytes secrete inflammatory mediators and express immune receptors to recruit immune cells and aggravate the inflammation. The better understanding of the obesity-related inflammatory milieu and the sequential events leading to IR could be helpful in designing new preventive and therapeutic strategies. The present review will discuss the cellular and molecular abnormalities participating in the pathogenesis of obesity in obese individuals as well as high-fat diet (HFD)-fed mice, a mouse model of obesity.
Collapse
Affiliation(s)
- Gholamreza Daryabor
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Kurosh Kalantar
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
41
|
Zhang F, Zarkada G, Han J, Li J, Dubrac A, Ola R, Genet G, Boyé K, Michon P, Künzel SE, Camporez JP, Singh AK, Fong GH, Simons M, Tso P, Fernández-Hernando C, Shulman GI, Sessa WC, Eichmann A. Lacteal junction zippering protects against diet-induced obesity. Science 2018; 361:599-603. [PMID: 30093598 PMCID: PMC6317738 DOI: 10.1126/science.aap9331] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 04/04/2018] [Accepted: 06/27/2018] [Indexed: 12/18/2022]
Abstract
Excess dietary lipid uptake causes obesity, a major global health problem. Enterocyte-absorbed lipids are packaged into chylomicrons, which enter the bloodstream through intestinal lymphatic vessels called lacteals. Here, we show that preventing lacteal chylomicron uptake by inducible endothelial genetic deletion of Neuropilin1 (Nrp1) and Vascular endothelial growth factor receptor 1 (Vegfr1; also known as Flt1) renders mice resistant to diet-induced obesity. Absence of NRP1 and FLT1 receptors increased VEGF-A bioavailability and signaling through VEGFR2, inducing lacteal junction zippering and chylomicron malabsorption. Restoring permeable lacteal junctions by VEGFR2 and vascular endothelial (VE)-cadherin signaling inhibition rescued chylomicron transport in the mutant mice. Zippering of lacteal junctions by disassembly of cytoskeletal VE-cadherin anchors prevented chylomicron uptake in wild-type mice. These data suggest that lacteal junctions may be targets for preventing dietary fat uptake.
Collapse
Affiliation(s)
- Feng Zhang
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Georgia Zarkada
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Jinah Han
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Jinyu Li
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Alexandre Dubrac
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Roxana Ola
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
- Department of Basic, Preventive and Clinical Science, University of Transylvania, 500019 Brasov, Romania
| | - Gael Genet
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Kevin Boyé
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Pauline Michon
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
- INSERM U970, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Steffen E Künzel
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Joao Paulo Camporez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Abhishek K Singh
- Departments of Comparative Medicine and Pathology, Vascular Biology and Therapeutics Program and Integrative Cell Signaling and Neurobiology of Metabolism Program, Yale University School of Medicine, New Haven, CT, USA
| | - Guo-Hua Fong
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06030-3501, USA
| | - Michael Simons
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH 45237-0507, USA
| | - Carlos Fernández-Hernando
- Departments of Comparative Medicine and Pathology, Vascular Biology and Therapeutics Program and Integrative Cell Signaling and Neurobiology of Metabolism Program, Yale University School of Medicine, New Haven, CT, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - William C Sessa
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510-3221, USA.
- INSERM U970, Paris Cardiovascular Research Center, 75015 Paris, France
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
42
|
Minchin JEN, Scahill CM, Staudt N, Busch-Nentwich EM, Rawls JF. Deep phenotyping in zebrafish reveals genetic and diet-induced adiposity changes that may inform disease risk. J Lipid Res 2018; 59:1536-1545. [PMID: 29794036 DOI: 10.1194/jlr.d084525] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/22/2018] [Indexed: 02/06/2023] Open
Abstract
The regional distribution of adipose tissues is implicated in a wide range of diseases. For example, proportional increases in visceral adipose tissue increase the risk for insulin resistance, diabetes, and CVD. Zebrafish offer a tractable model system by which to obtain unbiased and quantitative phenotypic information on regional adiposity, and deep phenotyping can explore complex disease-related adiposity traits. To facilitate deep phenotyping of zebrafish adiposity traits, we used pairwise correlations between 67 adiposity traits to generate stage-specific adiposity profiles that describe changing adiposity patterns and relationships during growth. Linear discriminant analysis classified individual fish according to an adiposity profile with 87.5% accuracy. Deep phenotyping of eight previously uncharacterized zebrafish mutants identified neuropilin 2b as a novel gene that alters adipose distribution. When we applied deep phenotyping to identify changes in adiposity during diet manipulations, zebrafish that underwent food restriction and refeeding had widespread adiposity changes when compared with continuously fed, equivalently sized control animals. In particular, internal adipose tissues (e.g., visceral adipose) exhibited a reduced capacity to replenish lipid following food restriction. Together, these results in zebrafish establish a new deep phenotyping technique as an unbiased and quantitative method to help uncover new relationships between genotype, diet, and adiposity.
Collapse
Affiliation(s)
- James E N Minchin
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom, EH16 4TJ .,Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710
| | - Catherine M Scahill
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom, CB10 1SA
| | - Nicole Staudt
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom, CB10 1SA
| | - Elisabeth M Busch-Nentwich
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom, CB10 1SA.,Department of Medicine, University of Cambridge, Cambridge, United Kingdom, CB2 0QQ
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710.,Department of Medicine, University of Cambridge, Cambridge, United Kingdom, CB2 0QQ
| |
Collapse
|
43
|
Bordon Y. Macrophages: Running on empty. Nat Rev Immunol 2018; 18:292-293. [PMID: 29616683 DOI: 10.1038/nri.2018.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|