1
|
van Tuijl J, van Heck JI, Bahrar H, Broeders W, Wijma J, ten Have YM, Giera M, Zweers-van Essen H, Rodwell L, Joosten LA, Netea MG, Afman LA, Bekkering S, Riksen NP. Single high-fat challenge and trained innate immunity: A randomized controlled cross-over trial. iScience 2024; 27:111103. [PMID: 39493874 PMCID: PMC11530819 DOI: 10.1016/j.isci.2024.111103] [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: 02/16/2024] [Revised: 05/11/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Brief exposure of monocytes to atherogenic molecules, such as oxidized lipoproteins, triggers a persistent pro-inflammatory phenotype, named trained immunity. In mice, transient high-fat diet leads to trained immunity, which aggravates atherogenesis. We hypothesized that a single high-fat challenge in humans induces trained immunity. In a randomized controlled cross-over study, 14 healthy individuals received a high-fat or reference shake, and blood was drawn before and after 1, 2, 4, 6, 24, and 72 h. Incubation of donor monocytes with the post-high-fat-shake serum induced trained immunity, regulated via Toll-like receptor 4. This was not mediated via triglyceride-rich lipoproteins, C12, 14, and 16, or metabolic endotoxemia. In vivo, however, the high-fat challenge did not affect monocyte phenotype and function. We conclude that a high-fat challenge leads to alterations in the serum composition that have the potential to induce trained immunity in vitro. However, this does not translate into a (persistent) hyperinflammatory monocyte phenotype in vivo.
Collapse
Affiliation(s)
- Julia van Tuijl
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Julia I.P. van Heck
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Wieteke Broeders
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Johan Wijma
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Yvonne M. ten Have
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Heidi Zweers-van Essen
- Department of Gastroenterology and Hepatology-Dietetics, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Laura Rodwell
- Section Biostatics, Department of Health Evidence, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Lydia A. Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University and Research, Wageningen 6700 HB, the Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Niels P. Riksen
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| |
Collapse
|
2
|
Swaby A, Atallah A, Varol O, Cristea A, Quail DF. Lifestyle and host determinants of antitumor immunity and cancer health disparities. Trends Cancer 2023; 9:1019-1040. [PMID: 37718223 DOI: 10.1016/j.trecan.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023]
Abstract
Lifestyle factors exert profound effects on host physiology and immunology. Disparities in cancer outcomes persist as a complex and multifaceted challenge, necessitating a comprehensive understanding of the interplay between host environment and antitumor immune responses. Determinants of health - such as obesity, diet, exercise, stress, or sleep disruption - have the potential for modification, yet some exert long-lasting effects and may challenge the notion of complete reversibility. Herein we review intersectional considerations of lifestyle immunity and the impact on tumor immunology and disparities in cancer outcomes, with a focus on obesity.
Collapse
Affiliation(s)
- Anikka Swaby
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Aline Atallah
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Ozgun Varol
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Alyssa Cristea
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Goodman Cancer Research Institute, Montreal, QC, Canada; Department of Experimental Medicine, McGill University, Montreal, QC, Canada; Department of Physiology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
3
|
DeBerge M, Chaudhary R, Schroth S, Thorp EB. Immunometabolism at the Heart of Cardiovascular Disease. JACC Basic Transl Sci 2023; 8:884-904. [PMID: 37547069 PMCID: PMC10401297 DOI: 10.1016/j.jacbts.2022.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 08/08/2023]
Abstract
Immune cell function among the myocardium, now more than ever, is appreciated to regulate cardiac function and pathophysiology. This is the case for both innate immunity, which includes neutrophils, monocytes, dendritic cells, and macrophages, as well as adaptive immunity, which includes T cells and B cells. This function is fueled by cell-intrinsic shifts in metabolism, such as glycolysis and oxidative phosphorylation, as well as metabolite availability, which originates from the surrounding extracellular milieu and varies during ischemia and metabolic syndrome. Immune cell crosstalk with cardiac parenchymal cells, such as cardiomyocytes and fibroblasts, is also regulated by complex cellular metabolic circuits. Although our understanding of immunometabolism has advanced rapidly over the past decade, in part through valuable insights made in cultured cells, there remains much to learn about contributions of in vivo immunometabolism and directly within the myocardium. Insight into such fundamental cell and molecular mechanisms holds potential to inform interventions that shift the balance of immunometabolism from maladaptive to cardioprotective and potentially even regenerative. Herein, we review our current working understanding of immunometabolism, specifically in the settings of sterile ischemic cardiac injury or cardiometabolic disease, both of which contribute to the onset of heart failure. We also discuss current gaps in knowledge in this context and therapeutic implications.
Collapse
Affiliation(s)
| | | | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
4
|
Wood EK, Sullivan EL. The Influence of Diet on Metabolism and Health Across the Lifespan in Nonhuman Primates. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 24. [PMID: 35425871 DOI: 10.1016/j.coemr.2022.100336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The macro and micronutrient composition and the overall quantity of the diet are important predictors of physical and psychological health and, as a consequence, behavior. Translational preclinical models are critical to identifying the mechanisms underlying these relationships. Nonhuman primate models are particularly instrumental to this line of research as they exhibit considerable genetic, social, and physiological similarities, as well as similarities in their developmental trajectories to humans. This review aims to discuss recent contributions to the field of diet and metabolism and health using nonhuman primate models. The influence of diet composition on health and physiology across the lifespan will be the primary focus, including recent work examining the impact of maternal diet programming of offspring physiologic and behavioral developmental outcomes.
Collapse
Affiliation(s)
- Elizabeth K Wood
- Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239
| | - Elinor L Sullivan
- Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239
- Oregon National Primate Research Center, 505 NW 185 Avenue, Beaverton, OR 97006
| |
Collapse
|
5
|
Schiattarella GG, Alcaide P, Condorelli G, Gillette TG, Heymans S, Jones EAV, Kallikourdis M, Lichtman A, Marelli-Berg F, Shah S, Thorp EB, Hill JA. Immunometabolic Mechanisms of Heart Failure with Preserved Ejection Fraction. NATURE CARDIOVASCULAR RESEARCH 2022; 1:211-222. [PMID: 35755006 PMCID: PMC9229992 DOI: 10.1038/s44161-022-00032-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing in prevalence worldwide, already accounting for at least half of all heart failure (HF). As most patients with HFpEF are obese with metabolic syndrome, metabolic stress has been implicated in syndrome pathogenesis. Recently, compelling evidence for bidirectional crosstalk between metabolic stress and chronic inflammation has emerged, and alterations in systemic and cardiac immune responses are held to participate in HFpEF pathophysiology. Indeed, based on both preclinical and clinical evidence, comorbidity-driven systemic inflammation, coupled with metabolic stress, have been implicated together in HFpEF pathogenesis. As metabolic alterations impact immune function(s) in HFpEF, major changes in immune cell metabolism are also recognized in HFpEF and in HFpEF-predisposing conditions. Both arms of immunity - innate and adaptive - are implicated in the cardiomyocyte response in HFpEF. Indeed, we submit that crosstalk among adipose tissue, the immune system, and the heart represents a critical component of HFpEF pathobiology. Here, we review recent evidence in support of immunometabolic mechanisms as drivers of HFpEF pathogenesis, discuss pivotal biological mechanisms underlying the syndrome, and highlight questions requiring additional inquiry.
Collapse
Affiliation(s)
- Gabriele G. Schiattarella
- Center for Cardiovascular Research (CCR), Department of Cardiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Gianluigi Condorelli
- Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Italy,Cardio Center, Humanitas Research Hospital IRCCS, Rozzano, Italy
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands,Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Elizabeth A. V. Jones
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Maastricht, Netherlands,Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marinos Kallikourdis
- Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Italy,Adaptive Immunity Lab, Humanitas Research Hospital IRCCS, Rozzano, Italy
| | - Andrew Lichtman
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sanjiv Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Edward B. Thorp
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
6
|
Ahn YJ, Wang L, Tavakoli S, Nguyen HN, Short JD, Asmis R. Glutaredoxin 1 controls monocyte reprogramming during nutrient stress and protects mice against obesity and atherosclerosis in a sex-specific manner. Nat Commun 2022; 13:790. [PMID: 35145079 PMCID: PMC8831602 DOI: 10.1038/s41467-022-28433-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
High-calorie diet-induced nutrient stress promotes thiol oxidative stress and the reprogramming of blood monocytes, giving rise to dysregulated, obesogenic, proatherogenic monocyte-derived macrophages. We report that in chow-fed, reproductively senescent female mice but not in age-matched male mice, deficiency in the thiol transferase glutaredoxin 1 (Grx1) promotes dysregulated macrophage phenotypes as well as rapid weight gain and atherogenesis. Grx1 deficiency derepresses distinct expression patterns of reactive oxygen species and reactive nitrogen species generators in male versus female macrophages, poising female but not male macrophages for increased peroxynitrate production. Hematopoietic Grx1 deficiency recapitulates this sexual dimorphism in high-calorie diet-fed LDLR-/- mice, whereas macrophage-restricted overexpression of Grx1 eliminates the sex differences unmasked by high-calorie diet-feeding and protects both males and females against atherogenesis. We conclude that loss of monocytic Grx1 activity disrupts the immunometabolic balance in mice and derepresses sexually dimorphic oxidative stress responses in macrophages. This mechanism may contribute to the sex differences reported in cardiovascular disease and obesity in humans. High-calorie diet promotes thiol oxidative stress and the reprogramming of blood monocytes, giving rise to obesogenic and proatherogenic macrophages. Here the authors report that loss of monocytic thiol transferase glutaredoxin 1 results in the derepression of sex-specific oxidative stress responses in macrophages, promoting atherogenesis and obesity in female mice.
Collapse
Affiliation(s)
- Yong Joo Ahn
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Luxi Wang
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sina Tavakoli
- Departments of Radiology and Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huynh Nga Nguyen
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - John D Short
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Reto Asmis
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
7
|
Sugino KY, Mandala A, Janssen RC, Gurung S, Trammell M, Day MW, Brush RS, Papin JF, Dyer DW, Agbaga MP, Friedman JE, Castillo-Castrejon M, Jonscher KR, Myers DA. Western diet-induced shifts in the maternal microbiome are associated with altered microRNA expression in baboon placenta and fetal liver. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:945768. [PMID: 36935840 PMCID: PMC10012127 DOI: 10.3389/fcdhc.2022.945768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Maternal consumption of a high-fat, Western-style diet (WD) disrupts the maternal/infant microbiome and contributes to developmental programming of the immune system and nonalcoholic fatty liver disease (NAFLD) in the offspring. Epigenetic changes, including non-coding miRNAs in the fetus and/or placenta may also underlie this risk. We previously showed that obese nonhuman primates fed a WD during pregnancy results in the loss of beneficial maternal gut microbes and dysregulation of cellular metabolism and mitochondrial dysfunction in the fetal liver, leading to a perturbed postnatal immune response with accelerated NAFLD in juvenile offspring. Here, we investigated associations between WD-induced maternal metabolic and microbiome changes, in the absence of obesity, and miRNA and gene expression changes in the placenta and fetal liver. After ~8-11 months of WD feeding, dams were similar in body weight but exhibited mild, systemic inflammation (elevated CRP and neutrophil count) and dyslipidemia (increased triglycerides and cholesterol) compared with dams fed a control diet. The maternal gut microbiome was mainly comprised of Lactobacillales and Clostridiales, with significantly decreased alpha diversity (P = 0.0163) in WD-fed dams but no community-wide differences (P = 0.26). At 0.9 gestation, mRNA expression of IL6 and TNF in maternal WD (mWD) exposed placentas trended higher, while increased triglycerides, expression of pro-inflammatory CCR2, and histological evidence for fibrosis were found in mWD-exposed fetal livers. In the mWD-exposed fetus, hepatic expression levels of miR-204-5p and miR-145-3p were significantly downregulated, whereas in mWD-exposed placentas, miR-182-5p and miR-183-5p were significantly decreased. Notably, miR-1285-3p expression in the liver and miR-183-5p in the placenta were significantly associated with inflammation and lipid synthesis pathway genes, respectively. Blautia and Ruminococcus were significantly associated with miR-122-5p in liver, while Coriobacteriaceae and Prevotellaceae were strongly associated with miR-1285-3p in the placenta; both miRNAs are implicated in pathways mediating postnatal growth and obesity. Our findings demonstrate that mWD shifts the maternal microbiome, lipid metabolism, and inflammation prior to obesity and are associated with epigenetic changes in the placenta and fetal liver. These changes may underlie inflammation, oxidative stress, and fibrosis patterns that drive NAFLD and metabolic disease risk in the next generation.
Collapse
Affiliation(s)
- Kameron Y. Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Ashok Mandala
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Sunam Gurung
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - MaJoi Trammell
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Michael W. Day
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Richard S. Brush
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - James F. Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - David W. Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Martin-Paul Agbaga
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Marisol Castillo-Castrejon
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Karen R. Jonscher
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- CORRESPONDENCE: Karen R. Jonscher,
| | - Dean A. Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
8
|
Hulme KD, Noye EC, Short KR, Labzin LI. Dysregulated Inflammation During Obesity: Driving Disease Severity in Influenza Virus and SARS-CoV-2 Infections. Front Immunol 2021; 12:770066. [PMID: 34777390 PMCID: PMC8581451 DOI: 10.3389/fimmu.2021.770066] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Acute inflammation is a critical host defense response during viral infection. When dysregulated, inflammation drives immunopathology and tissue damage. Excessive, damaging inflammation is a hallmark of both pandemic influenza A virus (IAV) infections and Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infections. Chronic, low-grade inflammation is also a feature of obesity. In recent years, obesity has been recognized as a growing pandemic with significant mortality and associated costs. Obesity is also an independent risk factor for increased disease severity and death during both IAV and SARS-CoV-2 infection. This review focuses on the effect of obesity on the inflammatory response in the context of viral respiratory infections and how this leads to increased viral pathology. Here, we will review the fundamentals of inflammation, how it is initiated in IAV and SARS-CoV-2 infection and its link to disease severity. We will examine how obesity drives chronic inflammation and trained immunity and how these impact the immune response to IAV and SARS-CoV-2. Finally, we review both medical and non-medical interventions for obesity, how they impact on the inflammatory response and how they could be used to prevent disease severity in obese patients. As projections of global obesity numbers show no sign of slowing down, future pandemic preparedness will require us to consider the metabolic health of the population. Furthermore, if weight-loss alone is insufficient to reduce the risk of increased respiratory virus-related mortality, closer attention must be paid to a patient’s history of health, and new therapeutic options identified.
Collapse
Affiliation(s)
- Katina D Hulme
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ellesandra C Noye
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Larisa I Labzin
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
9
|
Williams H, Mack CD, Li SCH, Fletcher JP, Medbury HJ. Nature versus Number: Monocytes in Cardiovascular Disease. Int J Mol Sci 2021; 22:ijms22179119. [PMID: 34502027 PMCID: PMC8430468 DOI: 10.3390/ijms22179119] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 01/01/2023] Open
Abstract
Monocytes play a key role in cardiovascular disease (CVD) as their influx into the vessel wall is necessary for the development of an atherosclerotic plaque. Monocytes are, however, heterogeneous differentiating from classical monocytes through the intermediate subset to the nonclassical subset. While it is recognized that the percentage of intermediate and nonclassical monocytes are higher in individuals with CVD, accompanying changes in inflammatory markers suggest a functional impact on disease development that goes beyond the increased proportion of these ‘inflammatory’ monocyte subsets. Furthermore, emerging evidence indicates that changes in monocyte proportion and function arise in dyslipidemia, with lipid lowering medication having some effect on reversing these changes. This review explores the nature and number of monocyte subsets in CVD addressing what they are, when they arise, the effect of lipid lowering treatment, and the possible implications for plaque development. Understanding these associations will deepen our understanding of the clinical significance of monocytes in CVD.
Collapse
Affiliation(s)
- Helen Williams
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Corinne D. Mack
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Stephen C. H. Li
- Chemical Pathology, NSW Health Pathology, Westmead Hospital and Institute of Clinical Pathology and Medical Research, Westmead, Sydney, NSW 2145, Australia;
- Blacktown/Mt Druitt Clinical School, Blacktown Hospital, Western Sydney University, Blacktown, NSW 2148, Australia
| | - John P. Fletcher
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Heather J. Medbury
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
- Correspondence:
| |
Collapse
|
10
|
Gene expression profile of CD14 + blood monocytes following lifestyle-induced weight loss in individuals with metabolic syndrome. Sci Rep 2020; 10:17855. [PMID: 33082492 PMCID: PMC7576128 DOI: 10.1038/s41598-020-74973-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 10/06/2020] [Indexed: 11/08/2022] Open
Abstract
Lifestyle-induced weight loss is regarded as an efficient therapy to reverse metabolic syndrome (MetS) and to prevent disease progression. The objective of this study was to investigate whether lifestyle-induced weight loss modulates gene expression in circulating monocytes. We analyzed and compared gene expression in monocytes (CD14+ cells) and subcutaneous adipose tissue biopsies by unbiased mRNA profiling. Samples were obtained before and after diet-induced weight loss in well-defined male individuals in a prospective controlled clinical trial (ICTRP Trial Number: U1111-1158-3672). The BMI declined significantly (− 12.6%) in the treatment arm (N = 39) during the 6-month weight loss intervention. This was associated with a significant reduction in hsCRP (− 45.84%) and circulating CD14+ cells (− 21.0%). Four genes were differentially expressed (DEG’s) in CD14+ cells following weight loss (ZRANB1, RNF25, RB1CC1 and KMT2C). Comparative analyses of paired CD14+ monocytes and subcutaneous adipose tissue samples before and after weight loss did not identify common genes differentially regulated in both sample types. Lifestyle-induced weight loss is associated with specific changes in gene expression in circulating CD14+ monocytes, which may affect ubiquitination, histone methylation and autophagy.
Collapse
|
11
|
Dietary 23-hydroxy ursolic acid protects against diet-induced weight gain and hyperglycemia by protecting monocytes and macrophages against nutrient stress-triggered reprogramming and dysfunction and preventing adipose tissue inflammation. J Nutr Biochem 2020; 86:108483. [PMID: 32860922 DOI: 10.1016/j.jnutbio.2020.108483] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022]
Abstract
The aim of this study was to determine whether the atheroprotective phytochemical 23-hydroxy ursolic acid protects against diet-induced obesity and hyperglycemia by preventing nutrient stress-induced monocyte reprogramming. After a two week run-in period on a defined, phytochemical-free low-fat maintenance diet, 12-week old female C57BL/6J mice were either kept on the maintenance diet for additional 13 weeks or switched to either a high-calorie diet, a high-calorie diet supplemented with either 0.05% 23-hydroxy ursolic acid or a high-calorie diet supplemented with 0.2% 23-hydroxy ursolic acid. Dietary supplementation with 23-hydroxy ursolic acid reduced weight gain and adipose tissue mass, prevented hyperglycemia, hyperleptinemia and adipose tissue inflammation, and preserved glucose tolerance. 23-Hydroxy ursolic acid also preserved blood monocyte mitogen-activated protein kinase phosphatase-1 activity, a biomarker of monocyte health, and reduced macrophage content in the adipose tissue. Targeted gene profiling by qRT-PCR using custom-designed TaqMan® Array Cards revealed that dietary 23-hydroxy ursolic acid converts macrophages into a transcriptionally hyperactive phenotype with enhanced antioxidant defenses and anti-inflammatory potential. In conclusion, our findings show that dietary 23-hydroxy ursolic acid exerts both anti-obesogenic effects through multiple mechanisms. These include improving glucose tolerance, preventing hyperleptinemia, maintaining blood monocyte function, reducing recruitment of monocyte-derived macrophages into adipose tissues during nutrient stress, and converting these macrophages into an anti-inflammatory, potentially inflammation-resolving phenotype, all contributing to reduced adipose tissue inflammation. Our data suggest that 23-hydroxy ursolic acid may serve as an oral therapeutic and dietary supplement suited for patients at risk for obesity, impaired glucose tolerance and cardiovascular disease.
Collapse
|
12
|
Bekkering S, Saner C, Riksen NP, Netea MG, Sabin MA, Saffery R, Stienstra R, Burgner DP. Trained Immunity: Linking Obesity and Cardiovascular Disease across the Life-Course? Trends Endocrinol Metab 2020; 31:378-389. [PMID: 32305098 DOI: 10.1016/j.tem.2020.01.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/17/2019] [Accepted: 01/09/2020] [Indexed: 02/06/2023]
Abstract
Obesity, a chronic inflammatory disease, is the most prevalent modifiable risk factor for cardiovascular disease. The mechanisms underlying inflammation in obesity are incompletely understood. Recent developments have challenged the dogma of immunological memory occurring exclusively in the adaptive immune system and show that the innate immune system has potential to be reprogrammed. This innate immune memory (trained immunity) is characterized by epigenetic and metabolic reprogramming of myeloid cells following endogenous or exogenous stimulation, resulting in enhanced inflammation to subsequent stimuli. Trained immunity phenotypes have now been reported for other immune and non-immune cells. Here, we provide a novel perspective on the putative role of trained immunity in mediating the adverse cardiovascular effects of obesity and highlight potential translational pathways.
Collapse
Affiliation(s)
- Siroon Bekkering
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Christoph Saner
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Endocrinology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Niels P Riksen
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Matthew A Sabin
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Endocrinology, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Rinke Stienstra
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - David P Burgner
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
13
|
Westerman K, Fernández‐Sanlés A, Patil P, Sebastiani P, Jacques P, Starr JM, J. Deary I, Liu Q, Liu S, Elosua R, DeMeo DL, Ordovás JM. Epigenomic Assessment of Cardiovascular Disease Risk and Interactions With Traditional Risk Metrics. J Am Heart Assoc 2020; 9:e015299. [PMID: 32308120 PMCID: PMC7428544 DOI: 10.1161/jaha.119.015299] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/10/2020] [Indexed: 12/16/2022]
Abstract
Background Epigenome-wide association studies for cardiometabolic risk factors have discovered multiple loci associated with incident cardiovascular disease (CVD). However, few studies have sought to directly optimize a predictor of CVD risk. Furthermore, it is challenging to train multivariate models across multiple studies in the presence of study- or batch effects. Methods and Results Here, we analyzed existing DNA methylation data collected using the Illumina HumanMethylation450 microarray to create a predictor of CVD risk across 3 cohorts: Women's Health Initiative, Framingham Heart Study Offspring Cohort, and Lothian Birth Cohorts. We trained Cox proportional hazards-based elastic net regressions for incident CVD separately in each cohort and used a recently introduced cross-study learning approach to integrate these individual scores into an ensemble predictor. The methylation-based risk score was associated with CVD time-to-event in a held-out fraction of the Framingham data set (hazard ratio per SD=1.28, 95% CI, 1.10-1.50) and predicted myocardial infarction status in the independent REGICOR (Girona Heart Registry) data set (odds ratio per SD=2.14, 95% CI, 1.58-2.89). These associations remained after adjustment for traditional cardiovascular risk factors and were similar to those from elastic net models trained on a directly merged data set. Additionally, we investigated interactions between the methylation-based risk score and both genetic and biochemical CVD risk, showing preliminary evidence of an enhanced performance in those with less traditional risk factor elevation. Conclusions This investigation provides proof-of-concept for a genome-wide, CVD-specific epigenomic risk score and suggests that DNA methylation data may enable the discovery of high-risk individuals who would be missed by alternative risk metrics.
Collapse
Affiliation(s)
- Kenneth Westerman
- JM‐USDA Human Nutrition Research Center on Aging at Tufts UniversityBostonMA
| | - Alba Fernández‐Sanlés
- Cardiovascular Epidemiology and Genetics Research GroupREGICOR Study GroupIMIM (Hospital del Mar Medical Research Institute)BarcelonaCataloniaSpain
- Pompeu Fabra University (UPF)BarcelonaCataloniaSpain
| | - Prasad Patil
- Department of BiostatisticsBoston University School of Public HealthBostonMA
| | - Paola Sebastiani
- Department of BiostatisticsBoston University School of Public HealthBostonMA
| | - Paul Jacques
- JM‐USDA Human Nutrition Research Center on Aging at Tufts UniversityBostonMA
| | - John M. Starr
- Department of PsychologyUniversity of EdinburghUnited Kingdom
- Centre for Cognitive Ageing and Cognitive EpidemiologyUniversity of EdinburghUnited Kingdom
| | - Ian J. Deary
- Department of PsychologyUniversity of EdinburghUnited Kingdom
- Centre for Cognitive Ageing and Cognitive EpidemiologyUniversity of EdinburghUnited Kingdom
| | - Qing Liu
- Department of EpidemiologyBrown University School of Public HealthProvidenceRI
| | - Simin Liu
- Department of EpidemiologyBrown University School of Public HealthProvidenceRI
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics Research GroupREGICOR Study GroupIMIM (Hospital del Mar Medical Research Institute)BarcelonaCataloniaSpain
- CIBER Cardiovascular Diseases (CIBERCV)MadridSpain
- Medicine DepartmentMedical SchoolUniversity of Vic‐Central University of Catalonia (UVic‐UCC)VicCataloniaSpain
| | - Dawn L. DeMeo
- Channing Division of Network MedicineDepartment of MedicineBrigham and Women’s HospitalBostonMA
| | - José M. Ordovás
- JM‐USDA Human Nutrition Research Center on Aging at Tufts UniversityBostonMA
- IMDEA AlimentaciónCEIUAMMadridSpain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| |
Collapse
|
14
|
Westerman K, Sebastiani P, Jacques P, Liu S, DeMeo D, Ordovás JM. DNA methylation modules associate with incident cardiovascular disease and cumulative risk factor exposure. Clin Epigenetics 2019; 11:142. [PMID: 31615550 PMCID: PMC6792327 DOI: 10.1186/s13148-019-0705-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/12/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Epigenome-wide association studies using DNA methylation have the potential to uncover novel biomarkers and mechanisms of cardiovascular disease (CVD) risk. However, the direction of causation for these associations is not always clear, and investigations to-date have often failed to replicate at the level of individual loci. METHODS Here, we undertook module- and region-based DNA methylation analyses of incident CVD in the Women's Health Initiative (WHI) and Framingham Heart Study Offspring Cohort (FHS) in order to find more robust epigenetic biomarkers for cardiovascular risk. We applied weighted gene correlation network analysis (WGCNA) and the Comb-p algorithm to find methylation modules and regions associated with incident CVD in the WHI dataset. RESULTS We discovered two modules whose activation correlated with CVD risk and replicated across cohorts. One of these modules was enriched for development-related processes and overlaps strongly with epigenetic aging sites. For the other, we showed preliminary evidence for monocyte-specific effects and statistical links to cumulative exposure to traditional cardiovascular risk factors. Additionally, we found three regions (associated with the genes SLC9A1, SLC1A5, and TNRC6C) whose methylation associates with CVD risk. CONCLUSIONS In sum, we present several epigenetic associations with incident CVD which reveal disease mechanisms related to development and monocyte biology. Furthermore, we show that epigenetic modules may act as a molecular readout of cumulative cardiovascular risk factor exposure, with implications for the improvement of clinical risk prediction.
Collapse
Affiliation(s)
- Kenneth Westerman
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Paul Jacques
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Simin Liu
- Department of Epidemiology, Brown University, Providence, RI, USA
| | - Dawn DeMeo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - José M Ordovás
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
- IMDEA Alimentación, CEI, UAM, Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
15
|
Pence BD, Yarbro JR. Classical monocytes maintain ex vivo glycolytic metabolism and early but not later inflammatory responses in older adults. Immun Ageing 2019; 16:3. [PMID: 30700992 PMCID: PMC6348080 DOI: 10.1186/s12979-019-0143-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/21/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Inflammaging is a condition of chronic low-grade inflammation due to the aging process and is associated with a variety of chronic diseases. Monocytes are innate immune cells which contribute to inflammation and are dysregulated during aging, demonstrated reduced phagocytosis, increased inflammation, and alterations in subset proportions. Metabolism is known to determine immune cell function, with quiescent and anti-inflammatory cells primarily relying on fatty acid oxidation, while activated and inflammatory cells primarily rely on glycolysis. We have previously shown an age-related decrease in mitochondrial respiratory capacity in monocytes, so we hypothesized here that a compensatory shift toward glycolysis would occur which would also exacerbate inflammation. RESULTS Using Seahorse assays, we profiled glycolysis in classical monocytes isolated from older (60-80 yr) and younger (18-35 yr) adults. Aging did not affect parameters of basal glycolysis in the glycolysis stress test, nor did it alter glycolytic activation early (2 h) or later (24 h) post-LPS stimulation. Cytokine gene expression was unchanged between aged and young subjects at 2 h post-LPS but was reduced in older subjects at 24 h post-LPS either significantly (IL1B) or near-significantly (IL6, IL10). CONCLUSIONS Aging appears not to affect glycolytic metabolism ex vivo in classical monocytes, but may reduce cytokine expression at later timepoints. Studies examining monocytes stimulated with age-altered circulating factors or with other pattern recognition receptor agonists may shed further light on monocyte metabolism as a determinant of immunosenescence and inflammaging.
Collapse
Affiliation(s)
- Brandt D. Pence
- School of Health Studies, Memphis, TN 38152 USA
- Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN 38152 USA
- 304 Elma Roane Fieldhouse, University of Memphis, Memphis, TN 38152 USA
| | | |
Collapse
|
16
|
Breznik JA, Naidoo A, Foley KP, Schulz C, Lau TC, Loukov D, Sloboda DM, Bowdish DME, Schertzer JD. TNF, but not hyperinsulinemia or hyperglycemia, is a key driver of obesity-induced monocytosis revealing that inflammatory monocytes correlate with insulin in obese male mice. Physiol Rep 2018; 6:e13937. [PMID: 30548217 PMCID: PMC6286899 DOI: 10.14814/phy2.13937] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 11/08/2018] [Accepted: 11/11/2018] [Indexed: 12/24/2022] Open
Abstract
Inflammation contributes to obesity-related hyperinsulinemia and insulin resistance, which often precede type 2 diabetes. Inflammation is one way that obesity can promote insulin resistance. It is not clear if the extent of obesity, hyperinsulinemia, or hyperglycemia, underpins changes in cellular immunity during diet-induced obesity. In particular, the requirement for obesity or directionality in the relationship between insulin resistance and monocyte characteristics is poorly defined. Inflammatory cytokines such as tumor necrosis factor (TNF) can contribute to insulin resistance. It is unclear if TNF alters monocytosis or specific markers of cellular immunity in the context of obesity. We measured bone marrow and blood monocyte characteristics in WT and TNF-/- mice that were fed obesogenic, high fat (HF) diets. We also used hyperglycemic Akita mice and mice implanted with insulin pellets in order to determine if glucose or insulin were sufficient to alter monocyte characteristics. We found that diet-induced obesity in male mice increased the total number of monocytes in blood, but not in bone marrow. Immature, inflammatory (Ly6Chigh ) monocytes decreased within the bone marrow and increased within peripheral blood of HF-fed mice. We found that neither hyperinsulinemia nor hyperglycemia was sufficient to induce the observed changes in circulating monocytes in the absence of diet-induced obesity. In obese HF-fed mice, antibiotic treatment lowered insulin and insulin resistance, but did not alter circulating monocyte characteristics. Fewer Ly6Chigh monocytes were present within the blood of HF-fed TNF-/- mice in comparison to HF-fed wild-type (WT) mice. The prevalence of immature Ly6Chigh monocytes in the blood correlated with serum insulin and insulin resistance irrespective of the magnitude of adipocyte or adipose tissue hypertrophy in obese mice. These data suggest that diet-induced obesity instigates a TNF-dependent increase in circulating inflammatory monocytes, which predicts increased blood insulin and insulin resistance independently from markers of adiposity or adipose tissue expansion.
Collapse
Affiliation(s)
- Jessica A. Breznik
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonCanada
- McMaster Immunology Research CentreMcMaster UniversityHamiltonCanada
- Michael G. DeGroote Institute for Infectious Disease ResearchMcMaster UniversityHamiltonCanada
| | - Avee Naidoo
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonCanada
- McMaster Immunology Research CentreMcMaster UniversityHamiltonCanada
- Michael G. DeGroote Institute for Infectious Disease ResearchMcMaster UniversityHamiltonCanada
| | - Kevin P. Foley
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonCanada
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamiltonCanada
| | - Christian Schulz
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonCanada
- McMaster Immunology Research CentreMcMaster UniversityHamiltonCanada
- Michael G. DeGroote Institute for Infectious Disease ResearchMcMaster UniversityHamiltonCanada
| | - Trevor C. Lau
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonCanada
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamiltonCanada
| | - Dessi Loukov
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonCanada
- McMaster Immunology Research CentreMcMaster UniversityHamiltonCanada
- Michael G. DeGroote Institute for Infectious Disease ResearchMcMaster UniversityHamiltonCanada
| | - Deborah M. Sloboda
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonCanada
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamiltonCanada
- Department of Obstetrics and Gynecology and PediatricsMcMaster UniversityHamiltonCanada
| | - Dawn M. E. Bowdish
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonCanada
- McMaster Immunology Research CentreMcMaster UniversityHamiltonCanada
- Michael G. DeGroote Institute for Infectious Disease ResearchMcMaster UniversityHamiltonCanada
| | - Jonathan D. Schertzer
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonCanada
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamiltonCanada
| |
Collapse
|
17
|
Nguyen HN, Ahn YJ, Medina EA, Asmis R. Dietary 23-hydroxy ursolic acid protects against atherosclerosis and obesity by preventing dyslipidemia-induced monocyte priming and dysfunction. Atherosclerosis 2018; 275:333-341. [PMID: 30015296 DOI: 10.1016/j.atherosclerosis.2018.06.882] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/16/2018] [Accepted: 06/26/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIMS We demonstrated that dietary ursolic acid (UA) reduces atherosclerotic lesion size and improves kidney function in diabetic mice. Based on structure-function analyses of naturally occurring UA analogs, we synthesized 23-hydroxy ursolic acid (23-OHUA), a compound with structural features predicted to enhance its bioavailability and anti-atherogenic properties compared to UA. The goal of this study was to determine the anti-obesogenic and atheroprotective properties of 23-OHUA and its mechanism of action. METHODS We performed chemotaxis assays to determine IC50 of phytochemicals on primed THP-1 monocytes. We fed 12-week old female LDLR-/- mice a high-fat diet (HFD) or a HFD supplemented with either 0.05% UA or 0.05% 23-OHUA, and measured monocyte priming, weight gain and atherosclerotic lesion size after 6 and 20 weeks. RESULTS Both dietary UA and 23-OHUA prevented dyslipidemia-induced loss of MKP-1 activity, and hyper-chemotactic activity, hallmarks of blood monocytes priming and dysfunction, but they did not affect plasma lipids or blood glucose levels nor WBC and monocyte counts. After 20 weeks, mice fed 23-OHUA showed 11% less weight gain compared to HFD-fed control mice and a 40% reduction in atherosclerotic plaque size, whereas UA reduced lesion size by only 19% and did not reduce weight gain. CONCLUSIONS Dietary 23-OHUA reduces weight gain and attenuates atherogenesis in mice by protecting monocytes against metabolic stress-induced priming and dysfunction. Based on its mechanism of action, 23-OHUA may represent a novel therapeutic approach for the prevention and treatment of obesity and atherosclerosis.
Collapse
Affiliation(s)
- Huynh Nga Nguyen
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Yong Joo Ahn
- Department of Internal Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Edward Antonio Medina
- Department of Pathology, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Reto Asmis
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA; Department of Internal Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
18
|
Jiang W, Agrawal DK, Boosani CS. Cell‑specific histone modifications in atherosclerosis (Review). Mol Med Rep 2018; 18:1215-1224. [PMID: 29901135 PMCID: PMC6072136 DOI: 10.3892/mmr.2018.9142] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022] Open
Abstract
Histone modifications are the key epigenetic mechanisms that have been identified to regulate gene expression in many human diseases. However, in the early developmental stages, such as in utero and the postnatal stages, histone modifications are essential for gene regulation and cell growth. Atherosclerosis represents a classical example of the involvement of different cell types, and their cumulative effects in the development of atheroma and the progression of the disease. Post translational modifications on proteins either induces their functional activity or renders them inactive. Post translational modifications such as methylation or acetylation on histones have been well characterized, and their role in enhancing or inhibiting specific gene expression was clearly elucidated. In the present review article, the critical roles of different histone modifications that occur in atherosclerosis have been summarized. Different histone proteins have been identified to serve a critical role in the development of atherosclerosis. Specifically, histone methylation and histone acetylation in monocytes, macrophages, vascular smooth muscle cells and in endothelial cells during the progression of atherosclerosis, have been well reported. In recent years, different target molecules and genes that regulate histone modifications have been examined for their effects in the treatment of atherosclerosis in animal models and in clinical trials. An increasing body of evidence suggests that these epigenetic changes resulting from DNA methylation and non-coding RNA may also be associated with histone modifications, thereby indicating that novel therapeutic strategies can be developed by targeting these post translational modifications, which may in turn aid in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Wanlin Jiang
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Chandra S Boosani
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| |
Collapse
|
19
|
Aging impairs mitochondrial respiratory capacity in classical monocytes. Exp Gerontol 2018; 108:112-117. [PMID: 29655929 DOI: 10.1016/j.exger.2018.04.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 12/27/2022]
Abstract
Aging is a critical healthcare concern, with age-related inflammation disposing individuals to a variety of diseases. Monocytes are affected by the aging process, with increased inflammation and impaired cellular functions such as phagocytosis. Mechanisms by which aging alters monocyte function are unknown, but recent research suggests that the balance of metabolic processes determine immune cell phenotype and function. Given the known association between aging and mitochondrial dysfunction in other tissues, we hypothesized that aging would impair mitochondrial function in monocytes. To test this, we isolated classical monocytes from young and older adults and tested mitochondrial function by a Seahorse assay. Aging reduced mitochondrial respiratory capacity and spare capacity in monocytes. Mitochondrial dysfunction is a potential mechanism by which aging alters monocyte phenotype and may impair inflammatory functions, especially in low-glucose environments where oxidative metabolism is necessary to meet energy demands.
Collapse
|