101
|
Covid-19: Fat, Obesity, Inflammation, Ethnicity, and Sex Differences. Pathogens 2020; 9:pathogens9110887. [PMID: 33114495 PMCID: PMC7692736 DOI: 10.3390/pathogens9110887] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 01/08/2023] Open
Abstract
Although obesity is known to be a risk factor for COVID-19 severity, there is an urgent need to distinguish between different kinds of fat—visceral and subcutaneous fat—and their inflammation status in COVID-19. These different fat types have partially diverging biochemical roles in the human body, and they are differentially associated with SARS-CoV-2, which targets the angiotensin-converting enzyme 2 (ACE2) for cell entry. ACE2 is highly expressed in adipose tissue, especially in visceral fat, suggesting an important role for this tissue in determining COVID-19 disease severity. In this perspective article, we discuss group differences in the amount of visceral fat levels and the extent of inflammation in adipocytes of visceral fat tissue, which may, in part, drive population, cross-national, ethnic, and sex differences in COVID-19 disease. It is vital to steer the scientific community’s attention to the effects of visceral fat in creating individual and population differences in COVID-19 severity. This can help researchers unravel the reasons for the reported population, ethnic, and sex differences in COVID-19 severity and mortality.
Collapse
|
102
|
Vyas V, Hunter RJ, Longhi MP, Finlay MC. Inflammation and adiposity: new frontiers in atrial fibrillation. Europace 2020; 22:1609-1618. [PMID: 33006596 DOI: 10.1093/europace/euaa214] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/22/2020] [Indexed: 01/26/2023] Open
Abstract
Abstract
The aetiology of atrial fibrillation (AF) remains poorly understood, despite its growing prevalence and associated morbidity, mortality, and healthcare costs. Obesity is implicated in myriad different disease processes and is now recognized a major risk factor in the pathogenesis of AF. Moreover, the role of distinct adipose tissue depots is a matter of intense scientific interest with the depot directly surrounding the heart—epicardial adipose tissue (EAT) appearing to have the greatest correlation with AF presence and severity. Similarly, inflammation is implicated in the pathophysiology of AF with EAT thought to act as a local depot of inflammatory mediators. These can easily diffuse into atrial tissue with the potential to alter its structural and electrical properties. Various meta-analyses have indicated that EAT size is an independent risk factor for AF with adipose tissue expansion being inevitably associated with a local inflammatory process. Here, we first briefly review adipose tissue anatomy and physiology then move on to the epidemiological data correlating EAT, inflammation, and AF. We focus particularly on discussing the mechanistic basis of how EAT inflammation may precipitate and maintain AF. Finally, we review how EAT can be utilized to help in the clinical management of AF patients and discuss future avenues for research.
Collapse
Affiliation(s)
- Vishal Vyas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Department of Cardiac Electrophysiology, Barts Heart Centre, St. Bartholomew’s Hospital, West Smithfield, London, UK
| | - Ross J Hunter
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Department of Cardiac Electrophysiology, Barts Heart Centre, St. Bartholomew’s Hospital, West Smithfield, London, UK
| | - M Paula Longhi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Malcolm C Finlay
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Department of Cardiac Electrophysiology, Barts Heart Centre, St. Bartholomew’s Hospital, West Smithfield, London, UK
| |
Collapse
|
103
|
Ferreira LM, Li AM, Serafim TL, Sobral MC, Alpoim MC, Urbano AM. Intermediary metabolism: An intricate network at the crossroads of cell fate and function. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165887. [DOI: 10.1016/j.bbadis.2020.165887] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/01/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022]
|
104
|
Polyakova YV, Zavodovsky BV, Sivordova LE, Akhverdyan YR, Zborovskaya IA. Visfatin and Rheumatoid Arthritis: Pathogenetic Implications and Clinical Utility. Curr Rheumatol Rev 2020; 16:224-239. [DOI: 10.2174/1573397115666190409112621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Objective:
Analysis and generalization of data related to visfatin involvement in the
pathogenesis of inflammation at various stages of rheumatoid arthritis.
Data Synthesis:
Visfatin is an adipocytokine which has also been identified in non-adipose tissues.
It influences directly on the maturation of B cells, which are involved in autoantibody production
and T cell activation. Visfatin can promote inflammation via regulation of pro-inflammatory cytokines
including TNF, IL-1β and IL-6. The concentration of circulating visfatin in rheumatoid arthritis
patients is higher compared to healthy individuals. Several studies suggest that visfatin level is
associated with rheumatoid arthritis activity, and its elevation may precede clinical signs of the relapse.
In murine collagen-induced arthritis, visfatin levels were also found to be elevated both in
inflamed synovial cells and in joint vasculature. Visfatin blockers have been shown to confer fast
and long-term attenuation of pathological processes; however, most of their effects are transient.
Other factors responsible for hyperactivation of the immune system can participate in this process
at a later stage. Treatment of rheumatoid arthritis with a combination of these blockers and inhibitors
of other mediators of inflammation can potentially improve treatment outcomes compared to
current therapeutic strategies. Recent advances in the treatment of experimental arthritis in mice as
well as the application of emerging treatment strategies obtained from oncology for rheumatoid arthritis
management could be a source of novel adipokine-mediated anti-rheumatic drugs.
Conclusion:
The ongoing surge of interest in anticytokine therapy makes further study of visfatin
highly relevant as it may serve as a base for innovational RA treatment.
Collapse
Affiliation(s)
- Yulia V. Polyakova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Boris V. Zavodovsky
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Larisa E. Sivordova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Yuri R. Akhverdyan
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Irina A. Zborovskaya
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| |
Collapse
|
105
|
Abstract
Crohn's disease [CD] is an inflammatory bowel disease of unknown aetiology. During recent decades, significant technological advances led to development of -omic datasets allowing a detailed description of the disease. Unfortunately these have not, to date, resolved the question of the aetiology of CD. Thus, it may be necessary to [re]consider hypothesis-driven approaches to resolve the aetiology of CD. According to the cold chain hypothesis, the development of industrial and domestic refrigeration has led to frequent exposure of human populations to bacteria capable of growing in the cold. These bacteria, at low levels of exposure, particularly those of the genus Yersinia, are believed to be capable of inducing exacerbated inflammation of the intestine in genetically predisposed subjects. We discuss the consistency of this working hypothesis in light of recent data from epidemiological, clinical, pathological, microbiological, and molecular studies.
Collapse
Affiliation(s)
- Jean-Pierre Hugot
- Centre de recherche sur l’inflammation, UMR1149 INSERM and Université de Paris, Paris, France,Service des maladies digestives et respiratoires de l’enfant, Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris, Paris, France,Corresponding author: Jean-Pierre Hugot, MD, PhD, Service des maladies digestives et respiratoires de l’enfant, Hôpital Robert Debré, 48 Bd Sérurier, F75019 Paris, France. Tel.: [33] 1 40 03 57 12; fax: [33] 1 40 03 57 66;
| | - Anne Dumay
- Centre de recherche sur l’inflammation, UMR1149 INSERM and Université de Paris, Paris, France
| | - Frédérick Barreau
- IRSD, UMR1220, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Ulrich Meinzer
- Centre de recherche sur l’inflammation, UMR1149 INSERM and Université de Paris, Paris, France,Service des maladies digestives et respiratoires de l’enfant, Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris, Paris, France
| |
Collapse
|
106
|
Abstract
Integrated immunometabolic responses link dietary intake, energy utilization, and storage to immune regulation of tissue function and is therefore essential for the maintenance and restoration of homeostasis. Adipose-resident leukocytes have non-traditional immunological functions that regulate organismal metabolism by controlling insulin action, lipolysis, and mitochondrial respiration to control the usage of substrates for production of heat versus ATP. Energetically expensive vital functions such as immunological responses might have thus evolved to respond accordingly to dietary surplus and deficit of macronutrient intake. Here, we review the interaction of dietary intake of macronutrients and their metabolism with the immune system. We discuss immunometabolic checkpoints that promote healthspan and highlight how dietary fate and regulation of glucose, fat, and protein metabolism might affect immunity.
Collapse
Affiliation(s)
- Aileen H Lee
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Vishwa Deep Dixit
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
107
|
Lercher A, Baazim H, Bergthaler A. Systemic Immunometabolism: Challenges and Opportunities. Immunity 2020; 53:496-509. [PMID: 32937151 PMCID: PMC7491485 DOI: 10.1016/j.immuni.2020.08.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/13/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022]
Abstract
Over the past 10 years, the field of immunometabolism made great strides to unveil the crucial role of intracellular metabolism in regulating immune cell function. Emerging insights into how systemic inflammation and metabolism influence each other provide a critical additional dimension on the organismal level. Here, we discuss the concept of systemic immunometabolism and review the current understanding of the communication circuits that underlie the reciprocal impact of systemic inflammation and metabolism across organs in inflammatory and infectious diseases, as well as how these mechanisms apply to homeostasis. We present current challenges of systemic immunometabolic research, and in this context, highlight opportunities and put forward ideas to effectively explore organismal physiological complexity in both health and disease.
Collapse
Affiliation(s)
- Alexander Lercher
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Hatoon Baazim
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria.
| |
Collapse
|
108
|
Masenga SK, Elijovich F, Koethe JR, Hamooya BM, Heimburger DC, Munsaka SM, Laffer CL, Kirabo A. Hypertension and Metabolic Syndrome in Persons with HIV. Curr Hypertens Rep 2020; 22:78. [PMID: 32880756 PMCID: PMC7467859 DOI: 10.1007/s11906-020-01089-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW With the advent of highly active antiretroviral therapy (ART), the life span of persons with HIV (PWH) has been nearly normalized. With aging, prevalence of the metabolic syndrome (MetS), including hypertension, has increased in the HIV population and exceeds that in the general population in some studies. This is due to a combination of traditional risk factors in addition to the effects attributable to the virus and ART. We review recent findings on the mechanisms contributing to MetS and hypertension in PWH, particularly those specific to the viral infection and to ART. RECENT FINDINGS Activation of the renin-angiotensin-aldosterone system (RAAS) and chronic immune activation contribute to the development of MetS and hypertension in PWH. HIV proteins and some ART agents alter adipocyte health contributing to dyslipidemias, weight gain, and insulin resistance. HIV infection also contributes to hypertension by direct effects on the RAAS that intertwine with inflammation by the RAAS also contributing to T cell activation. Recent data suggest that in addition to current ART, therapeutic targeting of the MetS and hypertension in PWH, by interfering with the RAAS, treating insulin resistance directly or by use of immunomodulators that dampen inflammation, may be critical for preventing or treating these risk factors and to improve overall cardiovascular complications in the HIV-infected aging population.
Collapse
Affiliation(s)
- Sepiso K Masenga
- HAND Research group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | - Fernando Elijovich
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John R Koethe
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benson M Hamooya
- HAND Research group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- School of Public Health, University of Zambia, Lusaka, Zambia
| | - Douglas C Heimburger
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Internal Medicine, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Sody M Munsaka
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | - Cheryl L Laffer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Room 536 Robinson Research Building, Nashville, TN, 37232-6602, USA.
| |
Collapse
|
109
|
Wang T, Shen Y, Luyten S, Yang Y, Jiang X. Tissue-resident memory CD8+ T cells in cancer immunology and immunotherapy. Pharmacol Res 2020; 159:104876. [DOI: 10.1016/j.phrs.2020.104876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
|
110
|
Vohra MS, Ahmad B, Serpell CJ, Parhar IS, Wong EH. Murine in vitro cellular models to better understand adipogenesis and its potential applications. Differentiation 2020; 115:62-84. [PMID: 32891960 DOI: 10.1016/j.diff.2020.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/08/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Adipogenesis has been extensively studied using in vitro models of cellular differentiation, enabling long-term regulation of fat cell metabolism in human adipose tissue (AT) material. Many studies promote the idea that manipulation of this process could potentially reduce the prevalence of obesity and its related diseases. It has now become essential to understand the molecular basis of fat cell development to tackle this pandemic disease, by identifying therapeutic targets and new biomarkers. This review explores murine cell models and their applications for study of the adipogenic differentiation process in vitro. We focus on the benefits and limitations of different cell line models to aid in interpreting data and selecting a good cell line model for successful understanding of adipose biology.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Bilal Ahmad
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom.
| | - Ishwar S Parhar
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University, Bandar Sunway, PJ 47500, Selangor, Malaysia.
| | - Eng Hwa Wong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
111
|
Majdoubi A, Lee JS, Kishta OA, Balood M, Moulefera MA, Ishido S, Talbot S, Cheong C, Alquier T, Thibodeau J. Lack of the E3 Ubiquitin Ligase March1 Affects CD8 T Cell Fate and Exacerbates Insulin Resistance in Obese Mice. Front Immunol 2020; 11:1953. [PMID: 32973799 PMCID: PMC7461985 DOI: 10.3389/fimmu.2020.01953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/20/2020] [Indexed: 01/08/2023] Open
Abstract
Obesity is a major risk factor for the development of insulin resistance and type 2 diabetes. However, the mechanisms that trigger the underlying adipose tissues inflammation are not completely understood. Here, we show that the E3 ubiquitin ligase March1 controls the phenotypic and functional properties of CD8+ T cells in mice white adipose tissue. In a diet-induced obesity model, mice lacking March1 [March1 knockout (KO)] show increased insulin resistance compared to their WT counterparts. Also, in obese March1 KO mice, the proportions of effector/memory (Tem) and resident/memory (Trm) CD8+ T cells were higher in the visceral adipose tissue, but not in the spleen. The effect of March1 on insulin resistance and on the phenotype of adipose tissue CD8+ T cells was independent of major histocompatibility complex class II ubiquitination. Interestingly, we adoptively transferred either WT or March1 KO splenic CD8+ T cells into obese WT chimeras that had been reconstituted with Rag1-deficient bone marrow. We observed an enrichment of Tem and Trm cells and exacerbated insulin resistance in mice that received March1 KO CD8 T cells. Mechanistically, we found that March1 deficiency alters the metabolic activity of CD8+ T cells. Our results provide additional evidence of the involvement of CD8+ T cells in adipose tissue inflammation and suggest that March1 controls the metabolic reprogramming of these cells.
Collapse
Affiliation(s)
- Abdelilah Majdoubi
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Jun Seong Lee
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Osama A Kishta
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Mohammad Balood
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | | | - Satoshi Ishido
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Sébastien Talbot
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Cheolho Cheong
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Thierry Alquier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Département de Médicine, Université de Montréal, Montreal, QC, Canada
| | - Jacques Thibodeau
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
112
|
Schetters STT, Kruijssen LJW, Crommentuijn MHW, Kalay H, den Haan JMM, van Kooyk Y. Immunological dynamics after subcutaneous immunization with a squalene-based oil-in-water adjuvant. FASEB J 2020; 34:12406-12418. [PMID: 33411367 PMCID: PMC7496326 DOI: 10.1096/fj.202000848r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/24/2020] [Accepted: 07/06/2020] [Indexed: 12/25/2022]
Abstract
The clinically successful adjuvant MF59 is used in seasonal influenza vaccines, which is proposed to enhance immunity by creating an immune-competent microenvironment in the muscle that allows recruitment of immune cells that drive adaptive immune responses. Here, we examined whether the clinically successful adjuvants MF59/AddaVax could be used for subcutaneous use and how antigen delivery can be synergized with cellular dynamics at the vaccination site. Subcutaneous injection of AddaVax leads to thickening of the skin, characterized by a neutrophil-monocyte recruitment sequence. Skin-infiltrating CCR2+Ly6Chigh monocytes showed differentiation to CD11b+Ly6C+MHCII+CD11c+CD64+ monocyte-derived DCs over time in the hypodermal layers of the skin, expressing high levels of CD209a/mDC-SIGN. Surprisingly, skin thickening was accompanied with increased white adipose tissue highly enriched with monocytes. Analysis of the skin-draining lymph nodes revealed early increases in neutrophils and moDCs at 12 hours after injection and later increases in migratory cDC2s. Subcutaneous vaccination with AddaVax enhanced antigen-specific CD8+ and CD4+ T cell responses, while moDC targeting using antigen-coupled CD209a antibody additionally boosted humoral responses. Hence, oil-in-water emulsions provide an attractive immune modulatory adjuvants aimed at increasing cellular responses, as well as antibody responses when combined with moDC targeting.
Collapse
Affiliation(s)
- Sjoerd T T Schetters
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Laura J W Kruijssen
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Matheus H W Crommentuijn
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Joke M M den Haan
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| |
Collapse
|
113
|
Stivers KB, Chilton PM, Beare JE, Dale JR, Alard P, LeBlanc AJ, Hoying JB. Adipose‐resident myeloid‐derived suppressor cells modulate immune cell homeostasis in healthy mice. Immunol Cell Biol 2020; 98:650-666. [DOI: 10.1111/imcb.12360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/31/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Katlin B Stivers
- Cardiovascular Innovation Institute University of Louisville School of Medicine Louisville KY40202USA
- Department of Microbiology & Immunology University of Louisville School of Medicine Louisville KY40202USA
| | - Paula M Chilton
- Cardiovascular Innovation Institute University of Louisville School of Medicine Louisville KY40202USA
- Department of Microbiology & Immunology University of Louisville School of Medicine Louisville KY40202USA
| | - Jason E Beare
- Cardiovascular Innovation Institute University of Louisville School of Medicine Louisville KY40202USA
| | - Jacob R Dale
- Cardiovascular Innovation Institute University of Louisville School of Medicine Louisville KY40202USA
| | - Pascale Alard
- Department of Microbiology & Immunology University of Louisville School of Medicine Louisville KY40202USA
| | - Amanda J LeBlanc
- Cardiovascular Innovation Institute University of Louisville School of Medicine Louisville KY40202USA
- Department of Physiology University of Louisville School of Medicine Louisville KY40292USA
| | - James B Hoying
- Cardiovascular Innovation Institute University of Louisville School of Medicine Louisville KY40202USA
- Department of Physiology University of Louisville School of Medicine Louisville KY40292USA
| |
Collapse
|
114
|
Misumi I, Starmer J, Uchimura T, Beck MA, Magnuson T, Whitmire JK. Obesity Expands a Distinct Population of T Cells in Adipose Tissue and Increases Vulnerability to Infection. Cell Rep 2020; 27:514-524.e5. [PMID: 30970254 PMCID: PMC6652206 DOI: 10.1016/j.celrep.2019.03.030] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 08/16/2018] [Accepted: 03/08/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity in humans is associated with poorer health outcomes after infections compared with non-obese individuals. Here, we examined the effects of white adipose tissue and obesity on T cell responses to viral infection in mice. We show that lymphocytic choriomeningitis virus (LCMV) grows to high titer in adipose tissue. Virus-specific T cells enter the adipose tissue to resolve infection but then remain as a memory population distinct from memory T cells in lymphoid tissues. Memory T cells in adipose tissue are abundant in lean mice, and diet-induced obesity further increases memory T cell number in adipose tissue and spleen. Upon re-challenge infection, memory T cells rapidly cause severe pathogenesis, leading to increases in lipase levels, calcification of adipose tissue, pancreatitis, and reduced survival in obese mice but not lean mice. Thus, obesity leads to a unique form of viral pathogenesis involving memory T cell-dependent adipocyte destruction and damage to other tissues. Obesity is associated with increased morbidity and mortality after viral infections. Using a mouse model of obesity, Misumi et al. identify a distinct population of memory T cells in white adipose tissue and a memory cell-dependent pathogenic response to infection that leads to acute fat necrosis, pancreatitis, and lethality.
Collapse
Affiliation(s)
- Ichiro Misumi
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Joshua Starmer
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Toru Uchimura
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Melinda A Beck
- Department of Nutrition, UNC-Chapel Hill Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Terry Magnuson
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Jason K Whitmire
- Department of Genetics, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA; Department of Microbiology & Immunology, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
115
|
Collins N. Dietary Regulation of Memory T Cells. Int J Mol Sci 2020; 21:ijms21124363. [PMID: 32575427 PMCID: PMC7352243 DOI: 10.3390/ijms21124363] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Memory T cells are a fundamental component of immunological memory, providing rapid and potent host protection against secondary challenges. As such, memory T cells are key targets in the design of vaccination strategies and cancer immunotherapies, making it critical to understand the factors and mechanisms that regulate their biology. Diet is an environmental feature that impacts virtually all aspects of host physiology. However, the influence of specific dietary regiments and nutritional components on the immune system is only just starting to be uncovered. This article will review literature regarding the impact of diet and nutrition on memory T cell development, maintenance and function. It was recently shown that caloric restriction without undernutrition enhances memory T cell function, while diets high in fiber are also beneficial. However, memory T cell responses are dysfunctional in extreme nutritional states, such as undernutrition and diet-induced obesity. Therefore, diet and host nutritional status are major regulators of memory T cell biology and host fitness. To define the dietary balance required to promote optimal memory T cell responses could allow for the implementation of rational diet-based therapies that prevent or treat disease. Furthermore, that certain dietary regiments can enhance memory T cell function indicates the possibility of harnessing the underlying mechanisms in the design of novel vaccination strategies and cancer immunotherapies.
Collapse
Affiliation(s)
- Nicholas Collins
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
116
|
Hussan JR, Hunter PJ. Our natural "makeup" reveals more than it hides: Modeling the skin and its microbiome. WIREs Mech Dis 2020; 13:e1497. [PMID: 32539232 DOI: 10.1002/wsbm.1497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 01/23/2023]
Abstract
Skin is our primary interface with the environment. A structurally and functionally complex organ that hosts a dynamic ecosystem of microbes, and synthesizes many compounds that affect our well-being and psychosocial interactions. It is a natural platform of signal exchange between internal organs, skin resident microbes, and the environment. These interactions have gained a great deal of attention due to the increased prevalence of atopic diseases, and the co-occurrence of multiple allergic diseases related to allergic sensitization in early life. Despite significant advances in experimentally characterizing the skin, its microbial ecology, and disease phenotypes, high-levels of variability in these characteristics even for the same clinical phenotype are observed. Addressing this variability and resolving the relevant biological processes requires a systems approach. This review presents some of our current understanding of the skin, skin-immune, skin-neuroendocrine, skin-microbiome interactions, and computer-based modeling approaches to simulate this ecosystem in the context of health and disease. The review highlights the need for a systems-based understanding of this sophisticated ecosystem. This article is categorized under: Infectious Diseases > Computational Models.
Collapse
Affiliation(s)
- Jagir R Hussan
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Peter J Hunter
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
117
|
Bapat SP, Liang Y, Zheng Y. Characterization of Immune Cells from Adipose Tissue. ACTA ACUST UNITED AC 2020; 126:e86. [PMID: 31483101 DOI: 10.1002/cpim.86] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adipose tissue (AT) serves a crucial role in maintaining organismal metabolic homeostasis. Studies have demonstrated that AT is populated with a diverse array of immune cells that coordinate and regulate AT function. This adipo-immune system is highly dynamic, reflecting the physiologic state of the organism (e.g., obese, lean, aged, or young) as well as the constant physiologic remodeling of AT associated with the daily rhythms of fasting and feeding. Many of the adaptive and maladaptive functional changes of AT are regulated by changes in the quantity and quality of distinct sets of AT-resident immune cells. Here we present protocols to assess the dynamic state of the immune system within AT by constructing censuses of adipose-resident immune cells (macrophages, dendritic cells, neutrophils, eosinophils, NK cells, innate lymphocytes, T cells, and B cells, etc.) based on flow cytometry, which we term adipo-immune profiles (AIPs). Constructing AIPs can be an integral part of assessment for AT health and function. This article describes the protocols to generate such AIPs. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Sagar P Bapat
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Yuqiong Liang
- Nomis Foundation Laboratories of Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California
| | - Ye Zheng
- Nomis Foundation Laboratories of Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|
118
|
Ramalho R, Rao M, Zhang C, Agrati C, Ippolito G, Wang FS, Zumla A, Maeurer M. Immunometabolism: new insights and lessons from antigen-directed cellular immune responses. Semin Immunopathol 2020; 42:279-313. [PMID: 32519148 PMCID: PMC7282544 DOI: 10.1007/s00281-020-00798-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023]
Abstract
Modulation of immune responses by nutrients is an important area of study in cellular biology and clinical sciences in the context of cancer therapies and anti-pathogen-directed immune responses in health and disease. We review metabolic pathways that influence immune cell function and cellular persistence in chronic infections. We also highlight the role of nutrients in altering the tissue microenvironment with lessons from the tumor microenvironment that shapes the quality and quantity of cellular immune responses. Multiple layers of biological networks, including the nature of nutritional supplements, the genetic background, previous exposures, and gut microbiota status have impact on cellular performance and immune competence against molecularly defined targets. We discuss how immune metabolism determines the differentiation pathway of antigen-specific immune cells and how these insights can be explored to devise better strategies to strengthen anti-pathogen-directed immune responses, while curbing unwanted, non-productive inflammation.
Collapse
Affiliation(s)
- Renata Ramalho
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM, U4585 FCT), Applied Nutrition Studies Group G.E.N.A.-IUEM), Instituto Universitário Egas Moniz, Egas Moniz Higher Education School, Monte de Caparica, Portugal
| | - Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Chao Zhang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | | | | | - Fu-Sheng Wang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.
- I Medizinische Klinik, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
119
|
Han J, Khatwani N, Searles TG, Turk MJ, Angeles CV. Memory CD8 + T cell responses to cancer. Semin Immunol 2020; 49:101435. [PMID: 33272898 PMCID: PMC7738415 DOI: 10.1016/j.smim.2020.101435] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
Long-lived memory CD8+ T cells play important roles in tumor immunity. Studies over the past two decades have identified four subsets of memory CD8+ T cells - central, effector, stem-like, and tissue resident memory - that either circulate through blood, lymphoid and peripheral organs, or reside in tissues where cancers develop. In this article, we will review studies from both pre-clinical mouse models and human patients to summarize the phenotype, distribution and unique features of each memory subset, and highlight specific roles of each subset in anti-tumor immunity. Moreover, we will discuss how stem-cell like and resident memory CD8+ T cell subsets relate to exhausted tumor-infiltrating lymphocytes (TIL) populations. These studies reveal how memory CD8+ T cell subsets together orchestrate durable immunity to cancer.
Collapse
Affiliation(s)
- Jichang Han
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, United States
| | - Nikhil Khatwani
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, United States
| | - Tyler G Searles
- Norris Cotton Cancer Center, The Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, United States
| | - Mary Jo Turk
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, United States; Norris Cotton Cancer Center, The Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, United States
| | - Christina V Angeles
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, United States; The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
120
|
Pangrazzi L, Naismith E, Miggitsch C, Carmona Arana JA, Keller M, Grubeck-Loebenstein B, Weinberger B. The impact of body mass index on adaptive immune cells in the human bone marrow. IMMUNITY & AGEING 2020; 17:15. [PMID: 32514279 PMCID: PMC7251898 DOI: 10.1186/s12979-020-00186-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023]
Abstract
Background Obesity has been associated with chronic inflammation and oxidative stress. Both conditions play a determinant role in the pathogenesis of age-related diseases, such as immunosenescence. Adipose tissue can modulate the function of the immune system with the secretion of molecules influencing the phenotype of immune cells. The importance of the bone marrow (BM) in the maintenance of antigen-experienced adaptive immune cells has been documented in mice. Recently, some groups have investigated the survival of effector/memory T cells in the human BM. Despite this, whether high body mass index (BMI) may affect immune cells in the BM and the production of molecules supporting the maintenance of these cells it is unknown. Methods Using flow cytometry, the frequency and the phenotype of immune cell populations were measured in paired BM and PB samples obtained from persons with different BMI. Furthermore, the expression of BM cytokines was assessed. The influence of cytomegalovirus (CMV) on T cell subsets was additionally considered, dividing the donors into the CMV− and CMV+ groups. Results Our study suggests that increased BMI may affect both the maintenance and the phenotype of adaptive immune cells in the BM. While the BM levels of IL-15 and IL-6, supporting the survival of highly differentiated T cells, and oxygen radicals increased in overweight persons, the production of IFNγ and TNF by CD8+ T cells was reduced. In addition, the frequency of B cells and CD4+ T cells positively correlated with BMI in the BM of CMV− persons. Finally, the frequency of several T cell subsets, and the expression of senescence/exhaustion markers within these subpopulations, were affected by BMI. In particular, the levels of bona fide memory T cells may be reduced in overweight persons. Conclusion Our work suggests that, in addition to aging and CMV, obesity may represent an additional risk factor for immunosenescence in adaptive immune cells. Metabolic interventions may help in improving the fitness of the immune system in the elderly.
Collapse
Affiliation(s)
- Luca Pangrazzi
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Rennweg, 10 Innsbruck, Austria.,Present Address: Center for Mind/Brain Sciences (CIMeC), University of Trento, Corso Bettini, 31 Rovereto, Italy
| | - Erin Naismith
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Rennweg, 10 Innsbruck, Austria
| | - Carina Miggitsch
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Rennweg, 10 Innsbruck, Austria.,Present Address: Private Kinderwunsch-Clinic Dr. J. Zech GmbH, Grabenweg 64, 6020 Innsbruck, Austria
| | - Jose' Antonio Carmona Arana
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Rennweg, 10 Innsbruck, Austria
| | - Michael Keller
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Rennweg, 10 Innsbruck, Austria
| | - Beatrix Grubeck-Loebenstein
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Rennweg, 10 Innsbruck, Austria
| | - Birgit Weinberger
- Department of Immunology, Institute for Biomedical Aging Research, University of Innsbruck, Rennweg, 10 Innsbruck, Austria
| |
Collapse
|
121
|
Spallanzani RG, Zemmour D, Xiao T, Jayewickreme T, Li C, Bryce PJ, Benoist C, Mathis D. Distinct immunocyte-promoting and adipocyte-generating stromal components coordinate adipose tissue immune and metabolic tenors. Sci Immunol 2020; 4:4/35/eaaw3658. [PMID: 31053654 DOI: 10.1126/sciimmunol.aaw3658] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/06/2019] [Indexed: 12/13/2022]
Abstract
Regulatory T cells (Tregs) are key brakes on the visceral adipose tissue (VAT) inflammation that regulates local and systemic metabolic tenor. Breakdown of this regulation promotes type 2 diabetes. The cytokine IL-33 expands and sustains the unique Treg population residing within VAT. Here, relying on single-cell RNA sequencing, we identified the major IL-33 producers in VAT to be particular mesenchymal stromal cell subtypes, related to but distinct from adipocyte progenitor cells. We explored modulation of the VAT stromal cell landscape with physiologic variables such as age and sex, as well as its remodeling in pathogenic states like obesity. Last, we uncovered a VAT Treg:stromal cell negative regulatory loop that keeps the potent effect of IL-33 under rein.
Collapse
Affiliation(s)
- Raul German Spallanzani
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David Zemmour
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Tianli Xiao
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Teshika Jayewickreme
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Chaoran Li
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Paul J Bryce
- Immunology & Inflammation Therapeutic Area, Sanofi US, Cambridge, MA 02139, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
122
|
Porsche CE, Delproposto JB, Patrick E, Zamarron BF, Lumeng CN. Adipose tissue dendritic cell signals are required to maintain T cell homeostasis and obesity-induced expansion. Mol Cell Endocrinol 2020; 505:110740. [PMID: 31987897 PMCID: PMC7197735 DOI: 10.1016/j.mce.2020.110740] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023]
Abstract
Adipose tissue derived chronic inflammation is a critical component of obesity induced type II diabetes. Major histocompatibility complex II (MHCII) mediated T cell activation within adipose tissue is one mechanism that contributes to this phenotype. However, the contribution of dendritic cells as professional antigen presenting cells in adipose issue has not previously been explored. Using ItgaxCre x MHCIIfl/fl (M11cKO) mice we observed adipose tissue specific changes in adipose tissue leukocytes. While there was a complete knockout of MHCII in dendritic cells, MHCII was also absent on the majority of macrophages. This resulted in reduction of TCR expression in CD4+ T cells in obese adipose tissue, and an increase in CD8+ and CD4+ CD8+ double positive T cells with decreased CD4+ T cells independent of diet type. Increased CD8+ cells were not observed in the spleen, suggesting adipose tissue T cell regulation is tissue specific. In vitro studies demonstrated more potent antigen presentation function in adipose tissue dendritic cells compared to macrophages. Obese M11cKO mice had decreased CD11c+ adipose tissue macrophages. Despite the changes of immune cellularity in adipose tissue, M11cKO largely did not change inflammatory gene expression in adipose tissue and did not demonstrate differences in glucose and insulin intolerance. Overall MHCII expression on CD11c+ cells is important for maintaining CD4+ and CD8+ adipose tissue T cells, but these cellular changes fail to alter inflammatory output and systemic metabolism.
Collapse
Affiliation(s)
- Cara E Porsche
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jennifer B Delproposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Elise Patrick
- College of Literature Sciences and Arts, University of Michigan, Ann Arbor, USA
| | - Brian F Zamarron
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carey N Lumeng
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
123
|
Frizzell H, Fonseca R, Christo SN, Evrard M, Cruz-Gomez S, Zanluqui NG, von Scheidt B, Freestone D, Park SL, McWilliam HEG, Villadangos JA, Carbone FR, Mackay LK. Organ-specific isoform selection of fatty acid–binding proteins in tissue-resident lymphocytes. Sci Immunol 2020; 5:5/46/eaay9283. [DOI: 10.1126/sciimmunol.aay9283] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 02/06/2020] [Indexed: 12/16/2022]
Abstract
Tissue-resident memory T (TRM) cells exist throughout the body, where they are poised to mediate local immune responses. Although studies have defined a common mechanism of residency independent of location, there is likely to be a level of specialization that adapts TRM cells to their given tissue of lodgment. It has been shown that TRM cells in the skin rely on the uptake of exogenous fatty acids for their survival and up-regulate fatty acid–binding protein 4 (FABP4) and FABP5 as part of their transcriptional program. However, FABPs exist as a larger family of isoforms, with different members selected in a tissue-specific fashion that is optimized for local fatty acid availability. Here, we show that although TRM cells in a range of tissue widely express FABPs, they are not restricted to FABP4 and FABP5. Instead, TRM cells show varying patterns of isoform usage that are determined by tissue-derived factors. These patterns are malleable because TRM cells relocated to different organs modify their FABP expression in line with their new location. As a consequence, these results argue for tissue-specific overlays to the TRM cell residency program, including FABP expression that is tailored to the particular tissue of TRM cell lodgment.
Collapse
Affiliation(s)
- H. Frizzell
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - R. Fonseca
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - S. N. Christo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - M. Evrard
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - S. Cruz-Gomez
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - N. G. Zanluqui
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - B. von Scheidt
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - D. Freestone
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - S. L. Park
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - H. E. G. McWilliam
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, Melbourne, VIC, Australia
| | - J. A. Villadangos
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, Melbourne, VIC, Australia
| | - F. R. Carbone
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - L. K. Mackay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
124
|
Kumar RK, Jin Y, Watts SW, Rockwell CE. Naïve, Regulatory, Activated, and Memory Immune Cells Co-exist in PVATs That Are Comparable in Density to Non-PVAT Fats in Health. Front Physiol 2020; 11:58. [PMID: 32116768 PMCID: PMC7026504 DOI: 10.3389/fphys.2020.00058] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Perivascular adipose tissue (PVAT), the fat surrounding peripheral blood vessels, is protective and reduces the contraction of blood vessels in health. PVAT is composed of adipocytes, stromal cells, and immune cells. Recent work supports eosinophils as one of the cell types key to the anti-contractile nature of PVAT in health. Hence, we hypothesized that there exists a basally activated immune cell community in healthy PVAT that is distinctly different from non-PVAT fats. PVATs were from around mesenteric resistance vessels (MRPVAT – white fat) and thoracic aorta (APVAT – brown fat). Non-PVATs included retroperitoneal (RP fat – white fat) and subscapular (SS fat – brown fat) while the spleen was a positive control. Tissues were harvested from adult male and female Sprague Dawley rats. Six primary immune cell types were identified in PVATs. T cells (CD4 and CD8), B cells, natural killer (NK) cells, macrophages, mast cells, and neutrophils in the stromal vascular fraction of each fat were identified using nine-color flow cytometry. PVATs contained a higher number of total immune cells vs. their respective non-PVAT fats in females. Females had a higher number of T cells in MRPVAT vs. males. Females also had a greater number of T cells and total immune cells in APVAT vs. males. Further, activation, differentiation, and/or polarization of various immune cell types were similarly determined by flow cytometry. PVATs were similar to their respective non-PVAT fats in density of recently activated B cells (B220+ CD25+). However, MRPVAT in females had a higher number of naïve CD4 T cells vs. MRPVAT in males and APVAT in females. MRPVAT also had denser naïve CD8 T cells vs. APVAT in females. Overall, this research for the first time has identified a community of discrete populations of immune cells (naive/recently activated/regulatory/memory) in healthy PVATs. Contrary to our hypothesis, PVATs are more similar than different in density to their respective non-PVAT fats.
Collapse
Affiliation(s)
- Ramya K Kumar
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Yining Jin
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Cheryl E Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
125
|
Oliveira BM, Pinto A, Correia A, Ferreira PG, Vilanova M, Teixeira L. Characterization of Myeloid Cellular Populations in Mesenteric and Subcutaneous Adipose Tissue of Holstein-Friesian Cows. Sci Rep 2020; 10:1771. [PMID: 32019985 PMCID: PMC7000716 DOI: 10.1038/s41598-020-58678-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
Immune cells resident in adipose tissue have important functions in local and systemic metabolic homeostasis. Nevertheless, these immune cell populations remain poorly characterized in bovines. Recently, we described diverse lymphocyte subpopulations in adipose tissue of Holstein-Friesian cows. Here, we aimed at characterising myeloid cell populations present in bovine adipose tissue using multicolour flow cytometry, cell sorting and histochemistry/immunohistochemistry. Macrophages, CD14+CD11b+MHC-II+CD45+ cells, were identified in mesenteric and subcutaneous adipose tissue, though at higher proportions in the latter. Mast cells, identified as SSC-AhighCD11b−/+CD14−MHC-II−CH138A−CD45+ cells, were also observed in adipose tissue and found at higher proportions than macrophages in mesenteric adipose tissue. Neutrophils, presenting a CH138A+CD11b+ phenotype, were also detected in mesenteric and subcutaneous adipose tissue, however, at much lower frequencies than in the blood. Our gating strategy allowed identification of eosinophils in blood but not in adipose tissue although being detected by morphological analysis at low frequencies in some animals. A population not expressing CD45 and with the CH138A+ CD11b−MHC-II− phenotype, was found abundant and present at higher proportions in mesenteric than subcutaneous adipose tissue. The work reported here may be useful for further studies addressing the function of the described cells.
Collapse
Affiliation(s)
- Bárbara M Oliveira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana Pinto
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Alexandra Correia
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Paula G Ferreira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Manuel Vilanova
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Luzia Teixeira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal. .,UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
126
|
Whole transcriptional analysis identifies markers of B, T and plasma cell signaling pathways in the mesenteric adipose tissue associated with Crohn's disease. J Transl Med 2020; 18:44. [PMID: 32000799 PMCID: PMC6993458 DOI: 10.1186/s12967-020-02220-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/10/2020] [Indexed: 12/21/2022] Open
Abstract
Background Crohn’s disease (CD) is a multifactorial disease characterized by chronic intestinal inflammation. The increased visceral adiposity near the affected intestinal area, of which mesenteric adipose tissue (MAT) is the main component, is a feature of CD. Both protective and pathological roles have been attributed to this disease-associated tissue in CD. To understand the contribution of MAT to CD pathophysiology, a molecular and cellular signature of disease-associated MAT in CD patients was provided. Methods We performed an observational study with whole transcriptional analysis by RNA sequencing (RNA-seq) of MAT and ileal mucosa from CD patients with active disease and controls. qPCR and immunohistology were performed for validation analysis. Results RNA-seq identified 17 significantly regulated genes (|FC| > 1.5; FDR < 0.05) in CD-MAT compared to non-IBD controls, with a marked upregulation of plasma cell genes (i.e., IGLL5, MZB1, CD79A, POU2AF1, FCRL5, JCHAIN, DERL3, SDC1, PIM2). A less strict statistical cutoff value (|FC| > 1.5, nominal p ≤ 0.05) yielded a larger list of 651 genes in CD-MAT compared to controls. CD ileum showed the significant regulation compared to control ileum of 849 genes (|FC| > 1.5; FDR < 0.05) or 2654 genes (|FC| > 1.5, nominal p ≤ 0.05). Ingenuity Pathway Analysis revealed the significant regulation of pathways related to T- and B cell functionality in the MAT of CD patients. Despite the differences between the MAT and ileal signatures of CD patients, we identified a subset of 204 genes significantly modulated in both tissues compared to controls. This common signature included genes related to the plasma cell signature. Genes such as S100A8, S100A9 (calprotectin) and IL1B, which are associated with acute inflammatory response, were exclusively regulated in the ileal mucosa of CD disease. In contrast, some genes encoding for lymphocyte receptors such as MS4A1, CD3D and CD79A were exclusively regulated in CD-MAT, exhibiting a different pattern of immune cell activation compared to the ileal mucosa in CD patients. qPCR and immunohistology confirmed the presence of large infiltrates of CD3+ CD20+ lymphocytes and CD138+ plasma cells in CD-MAT. Conclusion Our data strongly supports the role of CD-associated MAT as a site for T-, B- and plasma cell activation, and suggests that it could also act as a reservoir of memory immune responses.
Collapse
|
127
|
Troha K, Ayres JS. Metabolic Adaptations to Infections at the Organismal Level. Trends Immunol 2020; 41:113-125. [PMID: 31959515 DOI: 10.1016/j.it.2019.12.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 12/08/2019] [Accepted: 12/08/2019] [Indexed: 02/07/2023]
Abstract
Metabolic processes occurring during host-microbiota-pathogen interactions can favorably or negatively influence host survival during infection. Defining the metabolic needs of the three players, the mechanisms through which they acquire nutrients, and whether each participant cooperates or competes with each other to meet their own metabolic demands during infection has the potential to reveal new approaches to treat disease. Here, we review topical findings in organismal metabolism and infection and highlight four emerging lines of investigation: how host-microbiota metabolic partnerships protect against infection; competition for glucose between host and pathogen; significance of infection-induced anorexia; and redefinition of the role of iron during infection. We also discuss how these discoveries shape our understanding of infection biology and their likely therapeutic value.
Collapse
Affiliation(s)
- Katia Troha
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Janelle S Ayres
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
128
|
Abstract
Tertiary lymphoid organs (TLOs), also known as inducible lymphoid organs, tertiary lymphoid structures, tertiary lymphoid tissues, or ectopic lymphoid organs are accumulations of cells in chronic inflammation that have been observed in most tissues in autoimmunity, infection, and cancer in mouse and man. They share many properties with secondary lymphoid organs (SLOs), particularly lymph nodes, with regard to cellular composition, function, and regulation. TLOs include T and B cells, dendritic cells, follicular dendritic cells, and many other stromal cells, and high endothelial venules (HEVs) and lymphatic vessels. They serve as sites of antigen presentation and tolerance induction; they are harmful in autoimmunity and can be both harmful and beneficial in cancer. SLO induction in ontogeny is mediated by interactions of several cell types, including CD4+ CD3- lymphoid tissue inducer (LTi) RORγt+ cells that express LTαβ and interact with mesenchymal lymphoid tissue organizer (LTo) FAP+ cells in the presence of lymphatic and blood vessels. A variety of inducer cells initiate TLOs, including bona fide LTi cells, T cells, B cells, and NK cells. The mesenchymal organizer cells are less well characterized but can include FAP+ cells. Current challenges include identification of methods to inhibit TLOs in autoimmunity without affecting SLOs, and enhancement of TLOs for defense against tumors.
Collapse
Affiliation(s)
- Nancy H Ruddle
- Epidemiology of Microbial Diseases, Yale School of Public Health, 60 College St., New Haven, CT, 06510, USA.
| |
Collapse
|
129
|
Bourgeois C, Gorwood J, Barrail-Tran A, Lagathu C, Capeau J, Desjardins D, Le Grand R, Damouche A, Béréziat V, Lambotte O. Specific Biological Features of Adipose Tissue, and Their Impact on HIV Persistence. Front Microbiol 2019; 10:2837. [PMID: 31921023 PMCID: PMC6927940 DOI: 10.3389/fmicb.2019.02837] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Although white AT can contribute to anti-infectious immune responses, it can also be targeted and perturbed by pathogens. The AT's immune involvement is primarily due to strong pro-inflammatory responses (with both local and paracrine effects), and the large number of fat-resident macrophages. Adipocytes also exert direct antimicrobial responses. In recent years, it has been found that memory T cells accumulate in AT, where they provide efficient secondary responses against viral pathogens. These observations have prompted researchers to re-evaluate the links between obesity and susceptibility to infections. In contrast, AT serves as a reservoir for several persistence pathogens, such as human adenovirus Ad-36, Trypanosoma gondii, Mycobacterium tuberculosis, influenza A virus, and cytomegalovirus (CMV). The presence and persistence of bacterial DNA in AT has led to the concept of a tissue-specific microbiota. The unexpected coexistence of immune cells and pathogens within the specific AT environment is intriguing, and its impact on anti-infectious immune responses requires further evaluation. AT has been recently identified as a site of HIV persistence. In the context of HIV infection, AT is targeted by both the virus and the antiretroviral drugs. AT's intrinsic metabolic features, large overall mass, and wide distribution make it a major tissue reservoir, and one that may contribute to the pathophysiology of chronic HIV infections. Here, we review the immune, metabolic, viral, and pharmacological aspects that contribute to HIV persistence in AT. We also evaluate the respective impacts of both intrinsic and HIV-induced factors on AT's involvement as a viral reservoir. Lastly, we examine the potential consequences of HIV persistence on the metabolic and immune activities of AT.
Collapse
Affiliation(s)
- Christine Bourgeois
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Aurélie Barrail-Tran
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| | - Claire Lagathu
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Jacqueline Capeau
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Delphine Desjardins
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Roger Le Grand
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Abderaouf Damouche
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Véronique Béréziat
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Olivier Lambotte
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| |
Collapse
|
130
|
Gilleron J, Bouget G, Ivanov S, Meziat C, Ceppo F, Vergoni B, Djedaini M, Soprani A, Dumas K, Jacquel A, Yvan-Charvet L, Venteclef N, Tanti JF, Cormont M. Rab4b Deficiency in T Cells Promotes Adipose Treg/Th17 Imbalance, Adipose Tissue Dysfunction, and Insulin Resistance. Cell Rep 2019; 25:3329-3341.e5. [PMID: 30566860 DOI: 10.1016/j.celrep.2018.11.083] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/04/2018] [Accepted: 11/21/2018] [Indexed: 12/18/2022] Open
Abstract
Obesity modifies T cell populations in adipose tissue, thereby contributing to adipose tissue inflammation and insulin resistance. Here, we show that Rab4b, a small GTPase governing endocytic trafficking, is pivotal in T cells for the development of these pathological events. Rab4b expression is decreased in adipose T cells from mice and patients with obesity. The specific depletion of Rab4b in T cells causes adipocyte hypertrophy and insulin resistance in chow-fed mice and worsens insulin resistance in obese mice. This phenotype is driven by an increase in adipose Th17 and a decrease in adipose Treg due to a cell-autonomous skew of differentiation toward Th17. The Th17/Treg imbalance initiates adipose tissue inflammation and reduces adipogenesis, leading to lipid deposition in liver and muscles. Therefore, we propose that the obesity-induced loss of Rab4b in adipose T cells may contribute to maladaptive white adipose tissue remodeling and insulin resistance by altering adipose T cell fate.
Collapse
Affiliation(s)
- Jérôme Gilleron
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France
| | - Gwennaëlle Bouget
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France
| | - Stoyan Ivanov
- Université Côte d'Azur, Nice, France; INSERM U1065, Centre Méditerranéen de Médecine Moléculaire C3M, Team "Metabolism and Cancer," Nice, France
| | - Cindy Meziat
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France
| | - Franck Ceppo
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France
| | - Bastien Vergoni
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France
| | - Mansour Djedaini
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France
| | - Antoine Soprani
- Sorbonne Université, Université Pierre et Marie Curie, INSERM, UMR S_1138 Cordeliers Research Center, Paris, France; Clinique Geoffroy Saint-Hilaire, Ramsey Générale de Santé, Paris, France
| | - Karine Dumas
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France
| | - Arnaud Jacquel
- Université Côte d'Azur, Nice, France; INSERM U1065, Centre Méditerranéen de Médecine Moléculaire C3M, Team "Cell Death, Differentiation, and Cancer," Nice, France
| | - Laurent Yvan-Charvet
- Université Côte d'Azur, Nice, France; INSERM U1065, Centre Méditerranéen de Médecine Moléculaire C3M, Team "Metabolism and Cancer," Nice, France
| | - Nicolas Venteclef
- Sorbonne Université, Université Pierre et Marie Curie, INSERM, UMR S_1138 Cordeliers Research Center, Paris, France
| | - Jean-François Tanti
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France
| | - Mireille Cormont
- INSERM UMR1065, Mediterranean Center of Molecular Medicine C3M, Team "Cellular and Molecular Physiopathology of Obesity and Diabetes," Nice, France; Université Côte d'Azur, Nice, France.
| |
Collapse
|
131
|
Couturier J, Nuotio-Antar AM, Agarwal N, Wilkerson GK, Saha P, Kulkarni V, Lakhashe SK, Esquivel J, Nehete PN, Ruprecht RM, Sastry KJ, Meyer JM, Hill LR, Lake JE, Balasubramanyam A, Lewis DE. Lymphocytes upregulate CD36 in adipose tissue and liver. Adipocyte 2019; 8:154-163. [PMID: 31035848 PMCID: PMC6768236 DOI: 10.1080/21623945.2019.1609202] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
CD36 is a multifunctional scavenger receptor and lipid transporter implicated in metabolic and inflammatory pathologies, as well as cancer progression. CD36 is known to be expressed by adipocytes and monocytes/macrophages, but its expression by T cells is not clearly established. We found that CD4 and CD8 T cells in adipose tissue and liver of humans, monkeys, and mice upregulated CD36 expression (ranging from ~5–40% CD36+), whereas little to no CD36 was expressed by T cells in blood, spleen, and lymph nodes. CD36 was expressed predominantly by resting CD38-, HLA.DR-, and PD-1- adipose tissue T cells in monkeys, and increased during high-fat feeding in mice. Adipose tissue and liver promote a distinct phenotype in resident T cells characterized by CD36 upregulation.
Collapse
Affiliation(s)
- Jacob Couturier
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alli M. Nuotio-Antar
- US Department of Agriculture/Agricultural Research Center, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Neeti Agarwal
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Gregory K. Wilkerson
- Department of Comparative Medicine, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - Pradip Saha
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Viraj Kulkarni
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Samir K. Lakhashe
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Juan Esquivel
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Pramod N. Nehete
- Department of Comparative Medicine, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - Ruth M. Ruprecht
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
- Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - K. Jagannadha Sastry
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer M. Meyer
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lori R. Hill
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jordan E. Lake
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ashok Balasubramanyam
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Dorothy E. Lewis
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
132
|
Bentley EG, Pugh G, Gledhill LR, Flynn RJ. An analysis of the immune compartment within bovine adipose tissue. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 100:103411. [PMID: 31202894 DOI: 10.1016/j.dci.2019.103411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 06/09/2023]
Abstract
Adipose tissue (AT) has wide functions as an active endocrine organ acting as a site of nutrient storage and thermogenesis. Recently it has been identified as having a key role in murine and human immunity and inflammation. Type 1 or type 2 immune responses and their respective cytokines have been linked to white or brown AT, respectively. Most dramatic is the involvement of type-2 innate lymphoid cells (ILC2s) in stimulating eosinophil recruitment via interleukin (IL)-13 which in turn stimulates alternative macrophage activation via IL-4/IL-13. Recruited leukocytes are capable of influencing the cellular composition and function of adipose tissue and present a route to combat human obesity, however these processes are poorly understood in ruminants. Here we have characterised the resident leukocytes populations within bovine mesenteric AT (MAT) and subcutaneous AT (SAT), compared with the corresponding mesenteric lymph node (MLN). Concurring with related studies, we find bovine AT has its own resident leukocyte populations where eosinophils and neutrophils dominate. Importantly the proportion of eosinophils or neutrophils corresponded to the adipocyte size found in both depots. Further exploration of this area may have important implications on the food production industry or could be applied to improve the course of pathogenesis during disease.
Collapse
Affiliation(s)
- Eleanor G Bentley
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, L3 5RF, UK
| | - Glesni Pugh
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, L3 5RF, UK
| | - Laura R Gledhill
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, L3 5RF, UK
| | - Robin J Flynn
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, L3 5RF, UK.
| |
Collapse
|
133
|
Martini DJ, Kline MR, Liu Y, Shabto JM, Williams MA, Khan AI, Lewis C, Collins H, Akce M, Kissick HT, Carthon BC, Shaib WL, Alese OB, Pillai RN, Steuer CE, Wu CS, Lawson DH, Kudchadkar RR, El‐Rayes BF, Ramalingam SS, Owonikoko TK, Harvey RD, Master VA, Bilen MA. Adiposity may predict survival in patients with advanced stage cancer treated with immunotherapy in phase 1 clinical trials. Cancer 2019; 126:575-582. [DOI: 10.1002/cncr.32576] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/27/2019] [Accepted: 08/14/2019] [Indexed: 12/21/2022]
|
134
|
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: 24] [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
|
135
|
The Bone Marrow Protects and Optimizes Immunological Memory during Dietary Restriction. Cell 2019; 178:1088-1101.e15. [PMID: 31442402 PMCID: PMC6818271 DOI: 10.1016/j.cell.2019.07.049] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/28/2019] [Accepted: 07/29/2019] [Indexed: 12/31/2022]
Abstract
Mammals evolved in the face of fluctuating food availability. How the immune system adapts to transient nutritional stress remains poorly understood. Here, we show that memory T cells collapsed in secondary lymphoid organs in the context of dietary restriction (DR) but dramatically accumulated within the bone marrow (BM), where they adopted a state associated with energy conservation. This response was coordinated by glucocorticoids and associated with a profound remodeling of the BM compartment, which included an increase in T cell homing factors, erythropoiesis, and adipogenesis. Adipocytes, as well as CXCR4-CXCL12 and S1P-S1P1R interactions, contributed to enhanced T cell accumulation in BM during DR. Memory T cell homing to BM during DR was associated with enhanced protection against infections and tumors. Together, this work uncovers a fundamental host strategy to sustain and optimize immunological memory during nutritional challenges that involved a temporal and spatial reorganization of the memory pool within "safe haven" compartments.
Collapse
|
136
|
Architecture of antimicrobial skin defense. Cytokine Growth Factor Rev 2019; 49:70-84. [PMID: 31473081 DOI: 10.1016/j.cytogfr.2019.08.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
The skin is the largest and the most exposed organ in the body and its defense is regulated at several anatomical levels. Here, we explore how skin layers, including the epidermis, dermis, adipose tissue, and skin appendages, as well as cutaneous microbiota, contribute to the function of skin antimicrobial defense. We highlight recent studies that reveal the differential and complementary responses of skin layers to bacterial, viral, and fungal infection. In particular, we focus on key soluble mediators in the layered skin defense, such as antimicrobial peptides, as well as on lipid antimicrobials, cytokines, chemokines, and barrier-maintaining molecules. We include our own evaluative analyses of transcriptomic datasets of human skin to map the involvement of antimicrobial peptides in skin protection under both steady state and infectious conditions. Furthermore, we explore the versatility of the mechanisms underlying skin defense by highlighting the role of the immune and nervous systems in their interaction with cutaneous microbes, and by illustrating the multifunctionality of selected antimicrobial peptides in skin protection.
Collapse
|
137
|
Labarthe L, Henriquez S, Lambotte O, Di Santo JP, Le Grand R, Pflumio F, Arcangeli ML, Legrand N, Bourgeois C. Frontline Science: Exhaustion and senescence marker profiles on human T cells in BRGSF-A2 humanized mice resemble those in human samples. J Leukoc Biol 2019; 107:27-42. [PMID: 31378988 DOI: 10.1002/jlb.5hi1018-410rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 07/17/2019] [Accepted: 07/25/2019] [Indexed: 12/22/2022] Open
Abstract
This work sought to confirm the human-like expression of exhaustion and senescence markers in a mouse model with a humanized immune system (HIS): the Balb/c Rag2KO IL2rgcKO SirpαNOD Flk2KO HLA-A2HHD (BRGSF-A2) mouse reconstituted with human CD34+ cord blood cells. With regard to senescence markers, the percentage of CD57+ T cells was higher in the bone marrow (BM) than in the spleen or blood. The same was true for KLRG1+ hCD8+ T cells. With regard to exhaustion markers, the percentage of programmed death 1 (PD-1+ ) T cells was higher in the BM than in the spleen or blood; the same was true for TIGIT+ hCD4+ cells. These tissue-specific differences were related to both higher proportions of memory T cells in BM and intrinsic differences in expression within the memory fraction. In blood samples from HIS mice and healthy human donors (HDs), we found that the percentage of KLRG1+ cells among hCD8+ T cells was lower in HIS compared to HDs. The opposite was true for CD4+ T cells. Unexpectedly, a high frequency of KLRG1+ cells was observed among naive T cells in HIS mice. CD57 expression on T cells was similar in blood samples from HIS mice and HDs. Likewise, PD-1 expression was similar in the two systems, although a relatively low proportion of HIS hCD4+ T cells expressed TIGIT. The BRGSF-A2 HIS mouse's exhaustion and senescence profile was tissue specific and relatively human like; hence, this mouse might be a valuable tool for determining the preclinical efficacy of immunotherapies.
Collapse
Affiliation(s)
- Laura Labarthe
- IDMIT Department, CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IBFJ, F-92265, Paris, France.,genOway Paris, F-92265, Fontenay-aux-Roses, France
| | - Soledad Henriquez
- IDMIT Department, CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IBFJ, F-92265, Paris, France
| | - Olivier Lambotte
- IDMIT Department, CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IBFJ, F-92265, Paris, France.,Assistance Publique-Hôpitaux de Paris, Service de Médecine Interne et Immunologie Clinique, Groupe Hospitalier Universitaire Paris Sud, Hôpital Bicêtre, F-94276, Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, F-75015, Paris, France.,INSERM U1223, F-75015, Paris, France
| | - Roger Le Grand
- IDMIT Department, CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IBFJ, F-92265, Paris, France
| | - Françoise Pflumio
- IRCM, CEA-Université Paris 7-Université Paris Sud 11, INSERM U1274, Paris, France
| | | | | | - Christine Bourgeois
- IDMIT Department, CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IBFJ, F-92265, Paris, France
| |
Collapse
|
138
|
Rosshart SP, Herz J, Vassallo BG, Hunter A, Wall MK, Badger JH, McCulloch JA, Anastasakis DG, Sarshad AA, Leonardi I, Collins N, Blatter JA, Han SJ, Tamoutounour S, Potapova S, Foster St Claire MB, Yuan W, Sen SK, Dreier MS, Hild B, Hafner M, Wang D, Iliev ID, Belkaid Y, Trinchieri G, Rehermann B. Laboratory mice born to wild mice have natural microbiota and model human immune responses. Science 2019; 365:365/6452/eaaw4361. [PMID: 31371577 DOI: 10.1126/science.aaw4361] [Citation(s) in RCA: 336] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/06/2019] [Accepted: 06/27/2019] [Indexed: 12/11/2022]
Abstract
Laboratory mouse studies are paramount for understanding basic biological phenomena but also have limitations. These include conflicting results caused by divergent microbiota and limited translational research value. To address both shortcomings, we transferred C57BL/6 embryos into wild mice, creating "wildlings." These mice have a natural microbiota and pathogens at all body sites and the tractable genetics of C57BL/6 mice. The bacterial microbiome, mycobiome, and virome of wildlings affect the immune landscape of multiple organs. Their gut microbiota outcompete laboratory microbiota and demonstrate resilience to environmental challenges. Wildlings, but not conventional laboratory mice, phenocopied human immune responses in two preclinical studies. A combined natural microbiota- and pathogen-based model may enhance the reproducibility of biomedical studies and increase the bench-to-bedside safety and success of immunological studies.
Collapse
Affiliation(s)
- Stephan P Rosshart
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA.
| | - Jasmin Herz
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Brian G Vassallo
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Ashli Hunter
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Morgan K Wall
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan H Badger
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - John A McCulloch
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Dimitrios G Anastasakis
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute for Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD 20892, USA
| | - Aishe A Sarshad
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute for Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD 20892, USA
| | - Irina Leonardi
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nicholas Collins
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joshua A Blatter
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Seong-Ji Han
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samira Tamoutounour
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Svetlana Potapova
- Laboratory of Animal Sciences Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Mark B Foster St Claire
- Laboratory of Animal Sciences Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Wuxing Yuan
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD 20892, USA.,Leidos Biomedical Research, Inc., Microbiome and Genetics Core, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shurjo K Sen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD 20892, USA.,Leidos Biomedical Research, Inc., Microbiome and Genetics Core, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew S Dreier
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Benedikt Hild
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Markus Hafner
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute for Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD 20892, USA
| | - David Wang
- Departments of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Iliyan D Iliev
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD 20892, USA
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD 20892, USA.
| |
Collapse
|
139
|
Abstract
Tissue-resident memory T (TRM) cells have emerged as a major component of T cell biology. Recent investigations have greatly advanced our understanding of TRMs. Common features have been discovered to distinguish memory T cells residing in various mucosal and non-mucosal tissues from their circulating counterparts. Given that most organs and tissues contain a unique microenvironment, local signal-induced tissue-specific features are tightly associated with the differentiation, homeostasis, and protective functions of TRMs. Here, we discuss recent advances in the TRM field with a special emphasis on the interaction between local signals and TRMs in the context of individual tissue environment.
Collapse
Affiliation(s)
- Yong Liu
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South Univeristy, Changsha, Hunan 410008, China
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
140
|
Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B, Descamps H, Lundgren P, Bleriot C, Liu Z, Deczkowska A, Keren-Shaul H, David E, Zmora N, Eldar SM, Lubezky N, Shibolet O, Hill DA, Lazar MA, Colonna M, Ginhoux F, Shapiro H, Elinav E, Amit I. Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner. Cell 2019; 178:686-698.e14. [PMID: 31257031 PMCID: PMC7068689 DOI: 10.1016/j.cell.2019.05.054] [Citation(s) in RCA: 703] [Impact Index Per Article: 140.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/18/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022]
Abstract
Immune cells residing in white adipose tissue have been highlighted as important factors contributing to the pathogenesis of metabolic diseases, but the molecular regulators that drive adipose tissue immune cell remodeling during obesity remain largely unknown. Using index and transcriptional single-cell sorting, we comprehensively map all adipose tissue immune populations in both mice and humans during obesity. We describe a novel and conserved Trem2+ lipid-associated macrophage (LAM) subset and identify markers, spatial localization, origin, and functional pathways associated with these cells. Genetic ablation of Trem2 in mice globally inhibits the downstream molecular LAM program, leading to adipocyte hypertrophy as well as systemic hypercholesterolemia, body fat accumulation, and glucose intolerance. These findings identify Trem2 signaling as a major pathway by which macrophages respond to loss of tissue-level lipid homeostasis, highlighting Trem2 as a key sensor of metabolic pathologies across multiple tissues and a potential therapeutic target in metabolic diseases.
Collapse
Affiliation(s)
- Diego Adhemar Jaitin
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lorenz Adlung
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Christoph A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Assaf Weiner
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Baoguo Li
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Hélène Descamps
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Lundgren
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Camille Bleriot
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Biopolis, Singapore 138648, Singapore
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | | | - Hadas Keren-Shaul
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Niv Zmora
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shai Meron Eldar
- Division of Surgery, Tel-Aviv Medical Center and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Nir Lubezky
- Division of Surgery, Tel-Aviv Medical Center and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Oren Shibolet
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - David A Hill
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Biopolis, Singapore 138648, Singapore; Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
141
|
Macdougall CE, Wood EG, Solomou A, Scagliotti V, Taketo MM, Gaston-Massuet C, Marelli-Berg FM, Charalambous M, Longhi MP. Constitutive Activation of β-Catenin in Conventional Dendritic Cells Increases the Insulin Reserve to Ameliorate the Development of Type 2 Diabetes in Mice. Diabetes 2019; 68:1473-1484. [PMID: 31048369 DOI: 10.2337/db18-1243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 04/11/2019] [Indexed: 11/13/2022]
Abstract
β-Cell failure is central to the development of type 2 diabetes mellitus (T2DM). Dysregulation of metabolic and inflammatory processes during obesity contributes to the loss of islet function and impaired β-cell insulin secretion. Modulating the immune system, therefore, has the potential to ameliorate diseases. We report that inducing sustained expression of β-catenin in conventional dendritic cells (cDCs) provides a novel mechanism to enhance β-cell insulin secretion. Intriguingly, cDCs with constitutively activated β-catenin induced islet expansion by increasing β-cell proliferation in a model of diet-induced obesity. We further found that inflammation in these islets was reduced. Combined, these effects improved β-cell insulin secretion, suggesting a unique compensatory mechanism driven by cDCs to generate a greater insulin reserve in response to obesity-induced insulin resistance. Our findings highlight the potential of immune modulation to improve β-cell mass and function in T2DM.
Collapse
Affiliation(s)
- Claire E Macdougall
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Elizabeth G Wood
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Antonia Solomou
- Department of Medical and Molecular Genetics, King's College London, Guys Hospital, London, U.K
| | - Valeria Scagliotti
- Department of Medical and Molecular Genetics, King's College London, Guys Hospital, London, U.K
| | - Makoto Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Carles Gaston-Massuet
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, U.K
| | - Marika Charalambous
- Department of Medical and Molecular Genetics, King's College London, Guys Hospital, London, U.K
| | - M Paula Longhi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, U.K.
| |
Collapse
|
142
|
Proteomic analysis reveals greater abundance of complement and inflammatory proteins in subcutaneous adipose tissue from postpartum cows treated with sodium salicylate. J Proteomics 2019; 204:103399. [DOI: 10.1016/j.jprot.2019.103399] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/22/2019] [Accepted: 05/27/2019] [Indexed: 02/08/2023]
|
143
|
Wong Y, Nakamizo S, Tan KJ, Kabashima K. An Update on the Role of Adipose Tissues in Psoriasis. Front Immunol 2019; 10:1507. [PMID: 31316526 PMCID: PMC6609873 DOI: 10.3389/fimmu.2019.01507] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022] Open
Abstract
Psoriasis is a common chronic inflammatory skin disease that is increasingly being recognized as a disease that not only affects the skin but also has multi-systemic implications. The pathophysiological link between psoriasis and obesity is becoming increasingly elucidated by recent studies. The cross-talk between adipocytes and the immune system via various mediators such as adipokines could explain how obesity contributes to psoriasis. The effects of obesity on adipocytes include upregulation of pro-inflammatory adipokines such as leptin and resistin, downregulation of anti-inflammatory adipokine, and also the stimulation of pro-inflammatory cytokine production by macrophages. This article provides an update on the role of adipose tissues in psoriasis.
Collapse
Affiliation(s)
| | - Satoshi Nakamizo
- Singapore Immunology Network, Skin Research Institute of Singapore, ASTAR, Singapore, Singapore
| | - Kahbing J Tan
- Singapore Immunology Network, Skin Research Institute of Singapore, ASTAR, Singapore, Singapore
| | - Kenji Kabashima
- Singapore Immunology Network, Skin Research Institute of Singapore, ASTAR, Singapore, Singapore.,Department Dermatology, Kyoto University School of Medicine, Kyoto, Japan
| |
Collapse
|
144
|
Life-long control of cytomegalovirus (CMV) by T resident memory cells in the adipose tissue results in inflammation and hyperglycemia. PLoS Pathog 2019; 15:e1007890. [PMID: 31220189 PMCID: PMC6605679 DOI: 10.1371/journal.ppat.1007890] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 07/02/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Cytomegalovirus (CMV) is a ubiquitous herpesvirus infecting most of the world’s population. CMV has been rigorously investigated for its impact on lifelong immunity and potential complications arising from lifelong infection. A rigorous adaptive immune response mounts during progression of CMV infection from acute to latent states. CD8 T cells, in large part, drive this response and have very clearly been demonstrated to take up residence in the salivary gland and lungs of infected mice during latency. However, the role of tissue resident CD8 T cells as an ongoing defense mechanism against CMV has not been studied in other anatomical locations. Therefore, we sought to identify additional locations of anti-CMV T cell residency and the physiological consequences of such a response. Through RT-qPCR we found that mouse CMV (mCMV) infected the visceral adipose tissue and that this resulted in an expansion of leukocytes in situ. We further found, through flow cytometry, that adipose tissue became enriched in cytotoxic CD8 T cells that are specific for mCMV antigens from day 7 post infection through the lifespan of an infected animal (> 450 days post infection) and that carry markers of tissue residence. Furthermore, we found that inflammatory cytokines are elevated alongside the expansion of CD8 T cells. Finally, we show a correlation between the inflammatory state of adipose tissue in response to mCMV infection and the development of hyperglycemia in mice. Overall, this study identifies adipose tissue as a location of viral infection leading to a sustained and lifelong adaptive immune response mediated by CD8 T cells that correlates with hyperglycemia. These data potentially provide a mechanistic link between metabolic syndrome and chronic infection. Mouse cytomegalovirus (mCMV) infection results in initial systemic viremia that is thereafter controlled by the adaptive immune system. Control is mediated in part by T cells that render the virus undetectable systemically, and latent in specific organs, including the lungs and salivary glands. It remains unclear how latent virus is controlled across tissues given the large pool of systemic mCMV-specific T cells. We explored mCMV control in the adipose tissue, whose cellular constituents are potentially susceptible to infection. We found that mCMV infects the adipose tissue during the acute phase, causing local inflammation and a lifelong mCMV-specific CD8 T cell immune response. The response consisted largely from non-recirculating, tissue-resident T cells. The infected adipose tissue showed signs of metabolic changes, that may potentially predispose the infected host to metabolic dysregulation as evidenced by hyperglycemia. Accumulation and persistence of mCMV specific non-circulating resident CD8 T cells (Trm) in adipose tissue reveal a likely generalized mechanism of mCMV tissue reservoir control by Trm cells and identify the adipose tissue as a persistent mCMV reservoir, with potential implications for metabolic health.
Collapse
|
145
|
Caputa G, Castoldi A, Pearce EJ. Metabolic adaptations of tissue-resident immune cells. Nat Immunol 2019; 20:793-801. [DOI: 10.1038/s41590-019-0407-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/26/2019] [Indexed: 12/25/2022]
|
146
|
O'Sullivan D. The metabolic spectrum of memory T cells. Immunol Cell Biol 2019; 97:636-646. [DOI: 10.1111/imcb.12274] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/27/2022]
Affiliation(s)
- David O'Sullivan
- Department of Immunometabolism Max Planck Institute of Immunobiology and Epigenetics Freiburg Germany
| |
Collapse
|
147
|
Abstract
White adipose tissue (WAT) is exposed to pathogens that breach epithelial barriers, but the role of adipose T cells in immunity isn't clear. In this issue of Immunity, Han et al. (2017) find that WAT harbors a reservoir of memory T cells that provide antimicrobial immunity at the expense of lipid synthesis and metabolism.
Collapse
Affiliation(s)
- Stanley Cheuk
- Dermatology and Venereology Unit, Department of Medicine Solna, Karolinska Institutet, 17176 Stockholm, Sweden; Department of Dermatology, Karolinska University Hospital Solna, 17176 Stockholm, Sweden
| | - Liv Eidsmo
- Dermatology and Venereology Unit, Department of Medicine Solna, Karolinska Institutet, 17176 Stockholm, Sweden; Department of Dermatology, Karolinska University Hospital Solna, 17176 Stockholm, Sweden.
| |
Collapse
|
148
|
Lu J, Zhao J, Meng H, Zhang X. Adipose Tissue-Resident Immune Cells in Obesity and Type 2 Diabetes. Front Immunol 2019; 10:1173. [PMID: 31191541 PMCID: PMC6540829 DOI: 10.3389/fimmu.2019.01173] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 05/08/2019] [Indexed: 12/21/2022] Open
Abstract
Inflammation is an important contributor to the pathogenesis of obesity-related type 2 diabetes (T2D). Adipose tissue-resident immune cells have been observed, and the potential contribution of these cells to metabolic dysfunction has been appreciated in recent years. This review focused on adipose tissue-resident immune cells that are dysregulated in the context of obesity and T2D. We comprehensively overviewed emerging knowledge regarding the phenotypic and functional properties of these cells and local factors that control their development. We discussed their function in controlling the immune response cascade and disease progression. We also characterized the metabolic profiles of these cells to explain the functional consequences in obese adipose tissues. Finally, we discussed the potential therapeutic targeting of adipose tissue-resident immune cells with the aim of addressing novel therapeutic approaches for the treatment of this disease.
Collapse
Affiliation(s)
- Jingli Lu
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Junjie Zhao
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Haiyang Meng
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
149
|
Rao M, Dodoo E, Zumla A, Maeurer M. Immunometabolism and Pulmonary Infections: Implications for Protective Immune Responses and Host-Directed Therapies. Front Microbiol 2019; 10:962. [PMID: 31134013 PMCID: PMC6514247 DOI: 10.3389/fmicb.2019.00962] [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: 03/02/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
The biology and clinical efficacy of immune cells from patients with infectious diseases or cancer are associated with metabolic programming. Host immune- and stromal-cell genetic and epigenetic signatures in response to the invading pathogen shape disease pathophysiology and disease outcomes. Directly linked to the immunometabolic axis is the role of the host microbiome, which is also discussed here in the context of productive immune responses to lung infections. We also present host-directed therapies (HDT) as a clinically viable strategy to refocus dysregulated immunometabolism in patients with infectious diseases, which requires validation in early phase clinical trials as adjuncts to conventional antimicrobial therapy. These efforts are expected to be continuously supported by newly generated basic and translational research data to gain a better understanding of disease pathology while devising new molecularly defined platforms and therapeutic options to improve the treatment of patients with pulmonary infections, particularly in relation to multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Ernest Dodoo
- Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London, NIHR Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| |
Collapse
|
150
|
Francis MS, Auerbuch V. Editorial: The Pathogenic Yersiniae-Advances in the Understanding of Physiology and Virulence, Second Edition. Front Cell Infect Microbiol 2019; 9:119. [PMID: 31058103 PMCID: PMC6482262 DOI: 10.3389/fcimb.2019.00119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/03/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Matthew S Francis
- Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Victoria Auerbuch
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|