1
|
Carvalho D, Diaz-Amarilla P, Smith MR, Santi MD, Martinez-Busi M, Go YM, Jones DP, Duarte P, Savio E, Abin-Carriquiry JA, Arredondo F. Untargeted metabolomics of 3xTg-AD neurotoxic astrocytes. J Proteomics 2024; 310:105336. [PMID: 39448026 DOI: 10.1016/j.jprot.2024.105336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 10/26/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, affecting approximately 47 M people worldwide. Histological features and genetic risk factors, among other evidence, supported the amyloid hypothesis of the disease. This neuronocentric paradigm is currently undergoing a shift, considering evidence of the role of other cell types, such as microglia and astrocytes, in disease progression. Previously, we described a particular astrocyte subtype obtained from the 3xTg-AD murine model that displays neurotoxic properties in vitro. We continue here our exploratory analysis through the lens of metabolomics to identify potentially altered metabolites and biological pathways. Cell extracts from neurotoxic and control astrocytes were compared using high-resolution mass spectrometry-based metabolomics. Around 12 % of metabolic features demonstrated significant differences between neurotoxic and control astrocytes, including alterations in the key metabolite glutamate. Consistent with our previous transcriptomic study, the present results illustrate many homeostatic and regulatory functions of metabolites, suggesting that neurotoxic 3xTg-AD astrocytes exhibit alterations in the Krebs cycle as well as the prostaglandin pathway. This is the first metabolomic study performed in 3xTg-AD neurotoxic astrocytes. These results provide insight into metabolic alterations potentially associated with neurotoxicity and pathology progression in the 3xTg-AD mouse model and strengthen the therapeutic potential of astrocytes in AD. BIOLOGICAL SIGNIFICANCE: Our study is the first high-resolution metabolomic characterization of the novel neurotoxic 3xTg-AD astrocytes. We propose key metabolites and pathway alterations, as well as possible associations with gene expression alterations in the model. Our results are in line with recent hypotheses beyond the amyloid cascade, considering the involvement of several stress response cascades during the development of Alzheimer's disease. This work could inspire other researchers to initiate similar studies in related models. Furthermore, this work illustrates a powerful workflow for metabolite annotation and selection that can be implemented in other studies.
Collapse
Affiliation(s)
- Diego Carvalho
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay; Área de Matemática - DETEMA, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Pablo Diaz-Amarilla
- I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Mathew R Smith
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine; Department of Medicine, Emory University, GA, USA; Atlanta Veterans Affairs Healthcare System, Decatur, GA, USA
| | - María Daniela Santi
- I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay; Instituto Multidisciplinario de Biología Vegetal (IMBIV-CONICET), Ciudad Universitaria. X5000HUA, Córdoba, Argentina
| | - Marcela Martinez-Busi
- Plataforma de Servicios Analíticos, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine; Department of Medicine, Emory University, GA, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine; Department of Medicine, Emory University, GA, USA
| | - Pablo Duarte
- I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Eduardo Savio
- I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Juan A Abin-Carriquiry
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay; Laboratorio de Biofármacos, Instituto Pasteur de Montevideo, Montevideo, Uruguay.
| | - Florencia Arredondo
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay; I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay.
| |
Collapse
|
2
|
Traidl-Hoffmann C, Afghani J, Akdis CA, Akdis M, Aydin H, Bärenfaller K, Behrendt H, Bieber T, Bigliardi P, Bigliardi-Qi M, Bonefeld CM, Bösch S, Brüggen MC, Diemert S, Duchna HW, Fähndrich M, Fehr D, Fellmann M, Frei R, Garvey LH, Gharbo R, Gökkaya M, Grando K, Guillet C, Guler E, Gutermuth J, Herrmann N, Hijnen DJ, Hülpüsch C, Irvine AD, Jensen-Jarolim E, Kong HH, Koren H, Lang CCV, Lauener R, Maintz L, Mantel PY, Maverakis E, Möhrenschlager M, Müller S, Nadeau K, Neumann AU, O'Mahony L, Rabenja FR, Renz H, Rhyner C, Rietschel E, Ring J, Roduit C, Sasaki M, Schenk M, Schröder J, Simon D, Simon HU, Sokolowska M, Ständer S, Steinhoff M, Piccirillo DS, Taïeb A, Takaoka R, Tapparo M, Teixeira H, Thyssen JP, Traidl S, Uhlmann M, van de Veen W, van Hage M, Virchow C, Wollenberg A, Yasutaka M, Zink A, Schmid-Grendelmeier P. Navigating the evolving landscape of atopic dermatitis: Challenges and future opportunities: The 4th Davos declaration. Allergy 2024; 79:2605-2624. [PMID: 39099205 DOI: 10.1111/all.16247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024]
Abstract
The 4th Davos Declaration was developed during the Global Allergy Forum in Davos which aimed to elevate the care of patients with atopic dermatitis (AD) by uniting experts and stakeholders. The forum addressed the high prevalence of AD, with a strategic focus on advancing research, treatment, and management to meet the evolving challenges in the field. This multidisciplinary forum brought together top leaders from research, clinical practice, policy, and patient advocacy to discuss the critical aspects of AD, including neuroimmunology, environmental factors, comorbidities, and breakthroughs in prevention, diagnosis, and treatment. The discussions were geared towards fostering a collaborative approach to integrate these advancements into practical, patient-centric care. The forum underlined the mounting burden of AD, attributing it to significant environmental and lifestyle changes. It acknowledged the progress in understanding AD and in developing targeted therapies but recognized a gap in translating these innovations into clinical practice. Emphasis was placed on the need for enhanced awareness, education, and stakeholder engagement to address this gap effectively and to consider environmental and lifestyle factors in a comprehensive disease management strategy. The 4th Davos Declaration marks a significant milestone in the journey to improve care for people with AD. By promoting a holistic approach that combines research, education, and clinical application, the Forum sets a roadmap for stakeholders to collaborate to improve patient outcomes in AD, reflecting a commitment to adapt and respond to the dynamic challenges of AD in a changing world.
Collapse
Affiliation(s)
- Claudia Traidl-Hoffmann
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Jamie Afghani
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Cezmi A Akdis
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Mübecel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | | | - Katja Bärenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Heidrun Behrendt
- Center for Allergy and Environment (ZAUM), Technische Universität München, Germany
| | - Thomas Bieber
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Davos Biosciences, Davos, Switzerland
| | | | | | - Charlotte Menné Bonefeld
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Stefanie Bösch
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | - Hans-Werner Duchna
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Hochgebirgsklinik Davos, Davos, Switzerland
| | | | - Danielle Fehr
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | - Remo Frei
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Lena H Garvey
- Department of Dermatology and Allergy, Allergy Clinic, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Raschid Gharbo
- Psychosomatic Department, Hochgebirgsklinik, Davos, Switzerland
| | - Mehmet Gökkaya
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Karin Grando
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | - Carole Guillet
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | | | - Nadine Herrmann
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Dirk Jan Hijnen
- Diakonessenhuis Utrecht Zeist Doorn Locatie Utrecht, Erasmus MC, University Medical Center Utrecht, Utrecht, Netherlands
| | - Claudia Hülpüsch
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Alan D Irvine
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Erika Jensen-Jarolim
- Center of Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
- The interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Hillel Koren
- Environmental Health, LLC, Durham, North Carolina, USA
| | - Claudia C V Lang
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
| | - Roger Lauener
- Ostschweizer Kinderspital St. Gallen, St.Gallen, Switzerland
| | - Laura Maintz
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Pierre-Yves Mantel
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Emanuel Maverakis
- Department of Dermatology, University of California Davis, Sacramento, California, USA
| | | | - Svenja Müller
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Kari Nadeau
- Stanford University School of Medicine, Stanford, California, USA
| | - Avidan U Neumann
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
| | | | - Harald Renz
- Institute of Laboratory Medicine, Philipps University, Marburg, Germany
| | - Claudio Rhyner
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Ernst Rietschel
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Johannes Ring
- Klinik und Poliklinik für Dermatologie und Allergologie am Biederstein, Technische Universität München, Munich, Germany
| | - Caroline Roduit
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
- Ostschweizer Kinderspital St. Gallen, St.Gallen, Switzerland
| | - Mari Sasaki
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
| | - Mirjam Schenk
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jens Schröder
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Schleswig-Holstein (UK-SH), Kiel, Germany
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Milena Sokolowska
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Sonja Ständer
- Center for Chronic Pruritus and Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- School of Medicine, Weill Cornell Medicine-Qatar, Ar-Rayyan, Qatar
- College of Medicine, Qatar University, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | - Doris Straub Piccirillo
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Alain Taïeb
- INSERM 1312, University of Bordeaux, Bordeaux, France
| | - Roberto Takaoka
- Department of Dermatology, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | | | | | - Jacob Pontoppidan Thyssen
- Department of Dermatology and Venerology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Miriam Uhlmann
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Marianne van Hage
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institute and Karolinska University Hospital Stockholm, Solna, Sweden
| | - Christian Virchow
- Department of Pneumology, Intensive Care Medicine, Center for Internal Medicine, Universitätsmedizin Rostock, Rostock, Germany
| | - Andreas Wollenberg
- Department of Dermatology and Allergy, Ludwig-Maximilian-University, Munich, Germany
- Department of Dermatology and Allergy, University Hospital Augsburg, Augsburg, Germany
- Comprehensive Center of Inflammation Medicine, University Hospital Schleswig Holstein Campus Luebeck, Lubeck, Germany
| | - Mitamura Yasutaka
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Alexander Zink
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Medicine Solna, Division of Dermatology and Venereology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Schmid-Grendelmeier
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
| |
Collapse
|
3
|
Komaravolu RK, Mehta-D'souza P, Conner T, Allen M, Lumry J, Batushansky A, Pezant NP, Montgomery CG, Griffin TM. Sex-specific effects of injury and beta-adrenergic activation on metabolic and inflammatory mediators in a murine model of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2024; 32:1097-1112. [PMID: 38527663 PMCID: PMC11330734 DOI: 10.1016/j.joca.2024.03.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/09/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
OBJECTIVE Metabolic processes are intricately linked to the resolution of innate inflammation and tissue repair, two critical steps for treating post-traumatic osteoarthritis (PTOA). Based on lipolytic and immunoregulatory actions of norepinephrine, we hypothesized that intra-articular β-adrenergic receptor (βAR) stimulation would suppress PTOA-associated inflammation in the infrapatellar fat pad (IFP) and synovium. DESIGN We used the βAR agonist isoproterenol to perturb intra-articular metabolism 3.5 weeks after applying a non-invasive single-load compression injury to knees of 12-week-old male and female mice. We examined the acute effects of intra-articular isoproterenol treatment relative to saline on IFP histology, multiplex gene expression of synovium-IFP tissue, synovial fluid metabolomics, and mechanical allodynia. RESULTS Injured knees developed PTOA pathology characterized by heterotopic ossification, articular cartilage loss, and IFP atrophy and fibrosis. Isoproterenol suppressed the upregulation of pro-fibrotic genes and downregulated the expression of adipose genes and pro-inflammatory genes (Adam17, Cd14, Icam1, Csf1r, and Casp1) in injured joints of female (but not male) mice. Analysis of published single-cell RNA-seq data identified elevated catecholamine-associated gene expression in resident-like synovial-IFP macrophages after injury. Injury substantially altered synovial fluid metabolites by increasing amino acids, peptides, sphingolipids, phospholipids, bile acids, and dicarboxylic acids, but these changes were not appreciably altered by isoproterenol. Intra-articular injection of either isoproterenol or saline increased mechanical allodynia in female mice, whereas neither substance affected male mice. CONCLUSIONS Acute βAR activation altered synovial-IFP transcription in a sex and injury-dependent manner, suggesting that women with PTOA may be more sensitive than men to treatments targeting sympathetic neural signaling pathways.
Collapse
MESH Headings
- Animals
- Female
- Male
- Mice
- Isoproterenol/pharmacology
- Adrenergic beta-Agonists/pharmacology
- Disease Models, Animal
- Sex Factors
- Synovial Membrane/metabolism
- Adipose Tissue/metabolism
- Inflammation Mediators/metabolism
- Receptors, Adrenergic, beta/metabolism
- Injections, Intra-Articular
- Knee Injuries/complications
- Knee Injuries/metabolism
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/etiology
- Cartilage, Articular/metabolism
- Cartilage, Articular/drug effects
- Cartilage, Articular/pathology
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Ravi K Komaravolu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Padmaja Mehta-D'souza
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Taylor Conner
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Madeline Allen
- Department of Health and Exercise Science, University of Oklahoma, Norman, OK 73019, USA.
| | - Jessica Lumry
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Nathan P Pezant
- Center for Biomedical Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Courtney G Montgomery
- Center for Biomedical Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.
| | - Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Oklahoma City VA Health Care System, Oklahoma City, OK 73104, USA; Oklahoma Center for Geroscience and the Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
4
|
Das MK, Savidge B, Pearl JE, Yates T, Miles G, Pareek M, Haldar P, Cooper AM. Altered hepatic metabolic landscape and insulin sensitivity in response to pulmonary tuberculosis. PLoS Pathog 2024; 20:e1012565. [PMID: 39331683 PMCID: PMC11463835 DOI: 10.1371/journal.ppat.1012565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/09/2024] [Accepted: 08/26/2024] [Indexed: 09/29/2024] Open
Abstract
Chronic inflammation triggers development of metabolic disease, and pulmonary tuberculosis (TB) generates chronic systemic inflammation. Whether TB induced-inflammation impacts metabolic organs and leads to metabolic disorder is ill defined. The liver is the master regulator of metabolism and to determine the impact of pulmonary TB on this organ we undertook an unbiased mRNA and protein analyses of the liver in mice with TB and reanalysed published data on human disease. Pulmonary TB led to upregulation of genes in the liver related to immune signalling and downregulation of genes encoding metabolic processes. In liver, IFN signalling pathway genes were upregulated and this was reflected in increased biochemical evidence of IFN signalling, including nuclear location of phosphorylated Stat-1 in hepatocytes. The liver also exhibited reduced expression of genes encoding the gluconeogenesis rate-limiting enzymes Pck1 and G6pc. Phosphorylation of CREB, a transcription factor controlling gluconeogenesis was drastically reduced in the livers of mice with pulmonary TB as was phosphorylation of other glucose metabolism-related kinases, including GSK3a, AMPK, and p42. In support of the upregulated IFN signalling being linked to the downregulated metabolic functions in the liver, we found suppression of gluconeogenic gene expression and reduced CREB phosphorylation in hepatocyte cell lines treated with interferons. The impact of reduced gluconeogenic gene expression in the liver was seen when infected mice were less able to convert pyruvate, a gluconeogenesis substrate, to the same extent as uninfected mice. Infected mice also showed evidence of reduced systemic and hepatic insulin sensitivity. Similarly, in humans with TB, we found that changes in a metabolite-based signature of insulin resistance correlates temporally with successful treatment of active TB and with progression to active TB following exposure. These data support the hypothesis that TB drives interferon-mediated alteration of hepatic metabolism resulting in reduced gluconeogenesis and drives systemic reduction of insulin sensitivity.
Collapse
Affiliation(s)
- Mrinal K. Das
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
| | - Ben Savidge
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
| | - John E. Pearl
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
| | - Thomas Yates
- Diabetes Research Centre, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Gareth Miles
- Leicester Cancer Research Centre, University of Leicester, Clinical Sciences Building, Leicester, United Kingdom
| | - Manish Pareek
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Pranabashis Haldar
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- NIHR Respiratory Biomedical Research Centre, Leicester, Glenfield Hospital, Groby Road, Leicester, United Kingdom
| | - Andrea M. Cooper
- Department of Respiratory Sciences, Leicester TB Research Group, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| |
Collapse
|
5
|
Demangel C, Surace L. Host-pathogen interactions from a metabolic perspective: methods of investigation. Microbes Infect 2024; 26:105267. [PMID: 38007087 DOI: 10.1016/j.micinf.2023.105267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/21/2023] [Accepted: 11/21/2023] [Indexed: 11/27/2023]
Abstract
Metabolism shapes immune homeostasis in health and disease. This review presents the range of methods that are currently available to investigate the dialog between metabolism and immunity at the systemic, tissue and cellular levels, particularly during infection.
Collapse
Affiliation(s)
- Caroline Demangel
- Institut Pasteur, Université Paris Cité, Inserm U1224, Immunobiology and Therapy Unit, Paris, France
| | - Laura Surace
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
6
|
Chen A, Wang W, Mao Z, He Y, Chen S, Liu G, Su J, Feng P, Shi Y, Yan C, Lu J. Multimaterial 3D and 4D Bioprinting of Heterogenous Constructs for Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307686. [PMID: 37737521 DOI: 10.1002/adma.202307686] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/06/2023] [Indexed: 09/23/2023]
Abstract
Additive manufacturing (AM), which is based on the principle of layer-by-layer shaping and stacking of discrete materials, has shown significant benefits in the fabrication of complicated implants for tissue engineering (TE). However, many native tissues exhibit anisotropic heterogenous constructs with diverse components and functions. Consequently, the replication of complicated biomimetic constructs using conventional AM processes based on a single material is challenging. Multimaterial 3D and 4D bioprinting (with time as the fourth dimension) has emerged as a promising solution for constructing multifunctional implants with heterogenous constructs that can mimic the host microenvironment better than single-material alternatives. Notably, 4D-printed multimaterial implants with biomimetic heterogenous architectures can provide a time-dependent programmable dynamic microenvironment that can promote cell activity and tissue regeneration in response to external stimuli. This paper first presents the typical design strategies of biomimetic heterogenous constructs in TE applications. Subsequently, the latest processes in the multimaterial 3D and 4D bioprinting of heterogenous tissue constructs are discussed, along with their advantages and challenges. In particular, the potential of multimaterial 4D bioprinting of smart multifunctional tissue constructs is highlighted. Furthermore, this review provides insights into how multimaterial 3D and 4D bioprinting can facilitate the realization of next-generation TE applications.
Collapse
Affiliation(s)
- Annan Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Wanying Wang
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| | - Zhengyi Mao
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Yunhu He
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Shiting Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Guo Liu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Jin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Pei Feng
- State Key Laboratory of High-Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Yusheng Shi
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Chunze Yan
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Jian Lu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Hong Kong Branch of National Precious Metals Material Engineering Research, Center (NPMM), City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
7
|
Xiong H, Liao M, Zhang H, Li Y, Bai J, Zhang J, Li L, Zhang L. DARS expression in BCR/ABL1-negative myeloproliferative neoplasms and its association with the immune microenvironment. Sci Rep 2024; 14:16711. [PMID: 39030308 PMCID: PMC11271514 DOI: 10.1038/s41598-024-67067-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
DARS, encoding for aspartyl-tRNA synthetase, is implicated in the pathogenesis of various cancers, including renal cell carcinoma, glioblastoma, colon cancer, and gastric cancer. Its role in BCR/ABL1-negative myeloproliferative neoplasms (MPNs), however, remains unexplored. This study aimed to elucidate the expression of DARS in patients with MPNs (PV 23, ET 19, PMF 16) through immunohistochemical analysis and to examine the profiles of circulating immune cells and cytokines using flow cytometry. Our findings indicate a significant overexpression of DARS in all MPNs subtypes at the protein level compared to controls (P < 0.05). Notably, elevated DARS expression was linked to splenomegaly in MPNs patients. The expression of DARS showed a negative correlation with CD4+ T cells (R = - 0.451, P = 0.0004) and CD4+ T/CD8+ T cell ratio (R = - 0.3758, P = 0.0040), as well as with CD68+ tumor-associated macrophages (R = 0.4037, P = 0.0017). Conversely, it was positively correlated with IL-2 (R = 0.5419, P < 0.001), IL-5 (R = 0.3161, P = 0.0166), IL-6 (R = 0.2992, P = 0.0238), and IFN-γ (R = 0.3873, P = 0.0029). These findings underscore a significant association between DARS expression in MPNs patients and specific clinical characteristics, as well as immune cell composition. Further investigation into the interplay between DARS and the immune microenvironment in MPNs could shed light on the underlying mechanisms of MPNs pathogenesis and immune dysregulation.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Minjing Liao
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Huitao Zhang
- Department of General Practice, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yanhong Li
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jun Bai
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jinping Zhang
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China.
| | - Liansheng Zhang
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Kumar V, Stewart Iv JH. Pattern-Recognition Receptors and Immunometabolic Reprogramming: What We Know and What to Explore. J Innate Immun 2024; 16:295-323. [PMID: 38740018 PMCID: PMC11250681 DOI: 10.1159/000539278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Evolutionarily, immune response is a complex mechanism that protects the host from internal and external threats. Pattern-recognition receptors (PRRs) recognize MAMPs, PAMPs, and DAMPs to initiate a protective pro-inflammatory immune response. PRRs are expressed on the cell membranes by TLR1, 2, 4, and 6 and in the cytosolic organelles by TLR3, 7, 8, and 9, NLRs, ALRs, and cGLRs. We know their downstream signaling pathways controlling immunoregulatory and pro-inflammatory immune response. However, the impact of PRRs on metabolic control of immune cells to control their pro- and anti-inflammatory activity has not been discussed extensively. SUMMARY Immune cell metabolism or immunometabolism critically determines immune cells' pro-inflammatory phenotype and function. The current article discusses immunometabolic reprogramming (IR) upon activation of different PRRs, such as TLRs, NLRs, cGLRs, and RLRs. The duration and type of PRR activated, species studied, and location of immune cells to specific organ are critical factors to determine the IR-induced immune response. KEY MESSAGE The work herein describes IR upon TLR, NLR, cGLR, and RLR activation. Understanding IR upon activating different PRRs is critical for designing better immune cell-specific immunotherapeutics and immunomodulators targeting inflammation and inflammatory diseases.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Medical Education Building-C, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - John H Stewart Iv
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Medical Education Building-C, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Merech F, Gori S, Calo G, Hauk V, Paparini D, Rios D, Lara B, Doga L, D'Eramo L, Squassi A, Ramhorst R, Argüello RJ, Pérez Leirós C, Vota D. Monocyte immunometabolic reprogramming in human pregnancy: contribution of trophoblast cells. Am J Physiol Endocrinol Metab 2024; 326:E215-E225. [PMID: 38117266 DOI: 10.1152/ajpendo.00357.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/04/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023]
Abstract
Immunometabolism research is uncovering the relationship between metabolic features and immune cell functions in physiological and pathological conditions. Normal pregnancy entails a fine immune and metabolic regulation of the maternal-fetal interaction to assist the energetic demands of the fetus with immune homeostasis maintenance. Here, we determined the immunometabolic status of monocytes of pregnant women compared with nonpregnant controls and its impact on monocyte anti-inflammatory functions such as efferocytosis. Monocytes from pregnant women (16-20 wk) and nonpregnant age-matched controls were studied. Single cell-based metabolic assays using freshly isolated monocytes from both groups were carried out in parallel with functional assays ex vivo to evaluate monocyte efferocytic capacity. On the other hand, various in vitro metabolic assays with human monocytes or monocyte-derived macrophages were designed to explore the effect of trophoblast cells in the profiles observed. We found that pregnancy alters monocyte metabolism and function. An increased glucose dependency and enhanced efferocytosis were detected in monocytes from pregnant women at resting states, compared with nonpregnant controls. Furthermore, monocytes display a reduced glycolytic response when stimulated with lipopolysaccharide (LPS). The metabolic profiling of monocytes at this stage of pregnancy was comparable with the immunometabolic phenotypes of human monocytes treated in vitro with human first trimester trophoblast cell conditioned media. These findings suggest that immunometabolic mechanisms are involved in the functional shaping of monocytes during pregnancy with a contribution of trophoblast cells. Results provide new clues for future hypotheses regarding pregnancies complicated by metabolic disorders.NEW & NOTEWORTHY Immunometabolism stands as a novel perspective to understand the complex regulation of the immune response and to provide small molecule-based therapies. By applying this approach to study monocytes during pregnancy, we found that these cells have a unique activation pattern. They rely more on glycolysis and show increased efferocytosis/IL-10 production, but they do not have the typical proinflammatory responses. We also present evidence that trophoblast cells can shape monocytes into this distinct immunometabolic profile.
Collapse
Affiliation(s)
- Fátima Merech
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Soledad Gori
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Guillermina Calo
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Vanesa Hauk
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daniel Paparini
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daiana Rios
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Brenda Lara
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Luciana Doga
- Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Luciana D'Eramo
- Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Aldo Squassi
- Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Rafael J Argüello
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille University, CNRS, INSERM, Marseille, France
| | - Claudia Pérez Leirós
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daiana Vota
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
10
|
Dollet L, Lundell LS, Chibalin AV, Pendergrast LA, Pillon NJ, Lansbury EL, Elmastas M, Frendo-Cumbo S, Jalkanen J, de Castro Barbosa T, Cervone DT, Caidahl K, Dmytriyeva O, Deshmukh AS, Barrès R, Rydén M, Wallberg-Henriksson H, Zierath JR, Krook A. Exercise-induced crosstalk between immune cells and adipocytes in humans: Role of oncostatin-M. Cell Rep Med 2024; 5:101348. [PMID: 38151020 PMCID: PMC10829726 DOI: 10.1016/j.xcrm.2023.101348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/01/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023]
Abstract
The discovery of exercise-regulated circulatory factors has fueled interest in organ crosstalk, especially between skeletal muscle and adipose tissue, and the role in mediating beneficial effects of exercise. We studied the adipose tissue transcriptome in men and women with normal glucose tolerance or type 2 diabetes following an acute exercise bout, revealing substantial exercise- and time-dependent changes, with sustained increase in inflammatory genes in type 2 diabetes. We identify oncostatin-M as one of the most upregulated adipose-tissue-secreted factors post-exercise. In cultured human adipocytes, oncostatin-M enhances MAPK signaling and regulates lipolysis. Oncostatin-M expression arises predominantly from adipose tissue immune cell fractions, while the corresponding receptors are expressed in adipocytes. Oncostatin-M expression increases in cultured human Thp1 macrophages following exercise-like stimuli. Our results suggest that immune cells, via secreted factors such as oncostatin-M, mediate a crosstalk between skeletal muscle and adipose tissue during exercise to regulate adipocyte metabolism and adaptation.
Collapse
Affiliation(s)
- Lucile Dollet
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| | - Leonidas S Lundell
- Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Logan A Pendergrast
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Nicolas J Pillon
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Elizabeth L Lansbury
- Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Merve Elmastas
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Jutta Jalkanen
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Daniel T Cervone
- Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth Caidahl
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Oksana Dmytriyeva
- Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Atul S Deshmukh
- Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Romain Barrès
- Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Institute of Molecular and Cellular Pharmacology, CNRS and Université Côte d'Azur, Valbonne, France
| | - Mikael Rydén
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Inland Norway University of Applied Sciences, Lillehammer, Norway.
| |
Collapse
|
11
|
Vogel A, García González P, Argüello RJ. Measuring the Metabolic State of Tissue-Resident Macrophages via SCENITH. Methods Mol Biol 2024; 2713:363-376. [PMID: 37639136 DOI: 10.1007/978-1-0716-3437-0_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Functional reprograming of cells is linked to a process of metabolic rewiring that is adapted for such new functions or microenvironment. Macrophages are present in all tissues and exposed to different microenvironments throughout our body. Profiling energetic metabolism of tissue resident and other heterogeneous populations of macrophages in vitro and ex vivo is technologically very challenging. We have recently developed a method to functionally profile energetic metabolism with single-cell resolution, named SCENITH. This method can be performed rapidly ex vivo and does not require specialized equipment. In this book chapter, we will summarize the tissue processing, the procedure and methods, the analysis and example of results, and a series of frequently asked questions.
Collapse
Affiliation(s)
- Andrea Vogel
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Paulina García González
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Rafael J Argüello
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| |
Collapse
|
12
|
Calder PC, Bach-Faig A, Bevacqua T, Caballero Lopez CG, Chen ZY, Connolly D, Koay WL, Meydani SN, Pinar AS, Ribas-Filho D, Pierre A. Vital role for primary healthcare providers: urgent need to educate the community about daily nutritional self-care to support immune function and maintain health. BMJ Nutr Prev Health 2023; 6:392-401. [PMID: 38618551 PMCID: PMC11009526 DOI: 10.1136/bmjnph-2023-000755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/22/2023] [Indexed: 04/16/2024] Open
Abstract
The importance of self-care to improve health and social well-being is well recognised. Nevertheless, there remains a need to encourage people to better understand how their body works, and how to keep it healthy. Because of its important role, part of this understanding should be based on why the immune system must be supported. This highly complex system is essential for defending against pathogens, but also for maintaining health throughout the body by preserving homeostasis and integrity. Accordingly, the immune system requires active management for optimal functioning and to reduce the risk of chronic diseases. In addition to regular exercise, healthy sleeping patterns, cultivating mental resilience, adequate nutrition through healthy and diverse dietary habits is key to the daily support of immune function. Diet and the immune system are closely intertwined, and a poor diet will impair immunity and increase the risk of acute and chronic diseases. To help elucidate the roles of primary healthcare providers in supporting individuals to engage in self-care, an international group of experts reviewed the evidence for the roles of the immune system in maintaining health and for nutrition in daily immune support, and discussed implications for population health and clinical practice.
Collapse
Affiliation(s)
- Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Reseaech Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Anna Bach-Faig
- Faculty of Health Sciences, Open University of Catalonia, Barcelona, Spain
- Food and Nutrition Area, Barcelona Official College of Pharmacists, Barcelona, Spain
| | | | | | - Zheng-Yu Chen
- International Pharmaceutical Federation, Shanghai, China
| | | | | | - Simin N Meydani
- Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | | | - Durval Ribas-Filho
- Padre Albino Foundation, Faculty of Medicine, Catanduva, São Paulo, Brazil
| | | |
Collapse
|
13
|
Yang X, Hao J, Luo J, Lu X, Kong X. Adipose tissue‑derived extracellular vesicles: Systemic messengers in health and disease (Review). Mol Med Rep 2023; 28:189. [PMID: 37615193 PMCID: PMC10502927 DOI: 10.3892/mmr.2023.13076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023] Open
Abstract
Adipose tissue (AT) is a complicated metabolic organ consisting of a heterogeneous population of cells that exert wide‑ranging effects on the regulation of systemic metabolism and in maintaining metabolic homeostasis. Various obesity‑related complications are associated with the development of dysfunctional AT. As an essential transmitter of intercellular information, extracellular vesicles (EVs) have recently been recognized as crucial in regulating multiple physiological functions. AT‑derived extracellular vesicles (ADEVs) have been shown to facilitate cellular communication both inside and between ATs and other peripheral organs. Here, the role of EVs released from ATs in the homeostasis of metabolic and cardiovascular diseases, cancer, and neurological disorders by delivering lipids, proteins, and nucleic acids between different cells is summarized. Furthermore, the differences in the sources of ADEVs, such as adipocytes, AT macrophages, AT‑derived stem cells, and AT‑derived mesenchymal stem cells, are also discussed. This review may provide valuable information for the potential application of ADEVs in metabolic syndrome, cardiovascular diseases, cancer, and neurological disorders.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, Zheijiang 310002, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zheijiang 310002, P.R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zheijiang 310002, P.R. China
| | - Jiayue Hao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zheijiang 310058, P.R. China
| | - Jie Luo
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zheijiang 310006, P.R. China
| | - Xinliang Lu
- Bone Marrow Transplantation Center and Institute of Immunology of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Xianghui Kong
- Bone Marrow Transplantation Center and Institute of Immunology of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
14
|
Kumar V, Stewart JH. Immunometabolic reprogramming, another cancer hallmark. Front Immunol 2023; 14:1125874. [PMID: 37275901 PMCID: PMC10235624 DOI: 10.3389/fimmu.2023.1125874] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Molecular carcinogenesis is a multistep process that involves acquired abnormalities in key biological processes. The complexity of cancer pathogenesis is best illustrated in the six hallmarks of the cancer: (1) the development of self-sufficient growth signals, (2) the emergence of clones that are resistant to apoptosis, (3) resistance to the antigrowth signals, (4) neo-angiogenesis, (5) the invasion of normal tissue or spread to the distant organs, and (6) limitless replicative potential. It also appears that non-resolving inflammation leads to the dysregulation of immune cell metabolism and subsequent cancer progression. The present article delineates immunometabolic reprogramming as a critical hallmark of cancer by linking chronic inflammation and immunosuppression to cancer growth and metastasis. We propose that targeting tumor immunometabolic reprogramming will lead to the design of novel immunotherapeutic approaches to cancer.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, United States
| | - John H. Stewart
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, United States
- Louisiana State University- Louisiana Children’s Medical Center, Stanley S. Scott, School of Medicine, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, United States
| |
Collapse
|
15
|
Siggins RW, McTernan PM, Simon L, Souza-Smith FM, Molina PE. Mitochondrial Dysfunction: At the Nexus between Alcohol-Associated Immunometabolic Dysregulation and Tissue Injury. Int J Mol Sci 2023; 24:8650. [PMID: 37239997 PMCID: PMC10218577 DOI: 10.3390/ijms24108650] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Alcohol misuse, directly or indirectly as a result of its metabolism, negatively impacts most tissues, including four with critical roles in energy metabolism regulation: the liver, pancreas, adipose, and skeletal muscle. Mitochondria have long been studied for their biosynthetic roles, such as ATP synthesis and initiation of apoptosis. However, current research has provided evidence that mitochondria participate in myriad cellular processes, including immune activation, nutrient sensing in pancreatic β-cells, and skeletal muscle stem and progenitor cell differentiation. The literature indicates that alcohol impairs mitochondrial respiratory capacity, promoting reactive oxygen species (ROS) generation and disrupting mitochondrial dynamics, leading to dysfunctional mitochondria accumulation. As discussed in this review, mitochondrial dyshomeostasis emerges at a nexus between alcohol-disrupted cellular energy metabolism and tissue injury. Here, we highlight this link and focus on alcohol-mediated disruption of immunometabolism, which refers to two distinct, yet interrelated processes. Extrinsic immunometabolism involves processes whereby immune cells and their products influence cellular and/or tissue metabolism. Intrinsic immunometabolism describes immune cell fuel utilization and bioenergetics that affect intracellular processes. Alcohol-induced mitochondrial dysregulation negatively impacts immunometabolism in immune cells, contributing to tissue injury. This review will present the current state of literature, describing alcohol-mediated metabolic and immunometabolic dysregulation from a mitochondrial perspective.
Collapse
Affiliation(s)
- Robert W. Siggins
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Patrick M. McTernan
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Liz Simon
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Flavia M. Souza-Smith
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
| | - Patricia E. Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
16
|
Metabolic Regulation of T cell Activity: Implications for Metabolic-Based T-cell Therapies for Cancer. IRANIAN BIOMEDICAL JOURNAL 2023; 27:1-14. [PMID: 36624636 PMCID: PMC9971708 DOI: 10.52547/ibj.3811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Immunometabolism is an emerging field in tumor immunotherapy. Understanding the metabolic competition for access to the limited nutrients between tumor cells and immune cells can reveal the complexity of the tumor microenvironment and help develop new therapeutic approaches for cancer. Recent studies have focused on modifying the function of immune cells by manipulating their metabolic pathways. Besides, identifying metabolic events, which affect the function of immune cells leads to new therapeutic opportunities for treatment of inflammatory diseases and immune-related conditions. According to the literature, metabolic pathway such as glycolysis, tricarboxylic acid cycle, and fatty acid metabolism, significantly influence the survival, proliferation, activation, and function of immune cells and thus regulate immune responses. In this paper, we reviewed the role of metabolic processes and major signaling pathways involving in T-cell regulation and T-cell responses against tumor cells. Moreover, we summarized the new therapeutics suggested to enhance anti-tumor activity of T cells through manipulating metabolic pathways.
Collapse
|
17
|
Taylor VJ. Lactation from the inside out: Maternal homeorhetic gastrointestinal adaptations regulating energy and nutrient flow into milk production. Mol Cell Endocrinol 2023; 559:111797. [PMID: 36243202 DOI: 10.1016/j.mce.2022.111797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/30/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Lactation invokes homeorhetic processes to ramp up and supply milk synthesis components to fulfil nutritional, immunological and microbiological requirements of developing offspring, overseen by complex neuroendocrine networks. The maternal gut meets these intense metabolic demands, supported by hyperphagia and rapid adjustments to process larger food quantities. Enteroplasticity describes an inherent ability of the gastrointestinal tract to harness metabolic and structural adaptations that increase nutrient absorption. Most shifts in response to increased demands are transitory and by secreting milk, the continuous energetic drain out of the maternal body avoids development of pathological metabolic diseases. Lactation has various positive benefits for long-term maternal health but many females do not lactate for long post pregnancy and younger women are increasingly pre-disposed to excessive body mass and/or metabolic complications prior to reproducing. Inadvertently invoking intestinal adaptations to harvest and store excess nutrients has negative health implications with increased risks for both mother and offspring.
Collapse
Affiliation(s)
- Vicky J Taylor
- School of Life, Health and Chemical Sciences (LHCS), Faculty of Science, Technology, Engineering and Mathematics (STEM), The Open University, United Kingdom.
| |
Collapse
|
18
|
Khudyakov JI, Allen KN, Crocker DE, Trost NS, Roberts AH, Pirard L, Debier C, Piotrowski ER, Vázquez-Medina JP. Comprehensive molecular and morphological resolution of blubber stratification in a deep-diving, fasting-adapted seal. Front Physiol 2022; 13:1057721. [PMID: 36589428 PMCID: PMC9795062 DOI: 10.3389/fphys.2022.1057721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Blubber is a modified subcutaneous adipose tissue in marine mammals that provides energy storage, thermoregulation, hydrodynamic locomotion, and buoyancy. Blubber displays vertical stratification by lipid content, fatty acid composition, and vascularization, leading to the assumption that deeper blubber layers are metabolically active, while superficial layers are mainly structural and thermoregulatory. However, few studies have examined functional stratification of marine mammal blubber directly, especially in pinnipeds. We characterized morphological and transcriptional differences across blubber layers in the northern elephant seal, a deep-diving and fasting-adapted phocid. We collected blubber from seals early in their fasting period and divided blubber cores into three similarly sized portions. We hypothesized that the innermost blubber portion would have higher 1) heterogeneity in adipocyte size, 2) microvascular density, and 3) expression of genes associated with metabolism and hormone signaling than outer blubber. We found that adipocyte area and variance increased from outermost (skin-adjacent) to innermost (muscle-adjacent) blubber layers, suggesting that inner blubber has a higher capacity for lipid storage and turnover than outer blubber. Inner blubber had a higher proportion of CD144+ endothelial cells, suggesting higher microvascular density. In contrast, outer blubber had a higher proportion of CD4+ immune cells than inner blubber, suggesting higher capacity for response to tissue injury. Transcriptome analysis identified 61 genes that were differentially expressed between inner and outer blubber layers, many of which have not been studied previously in marine mammals. Based on known functions of these genes in other mammals, we suggest that inner blubber has potentially higher 1) adipogenic capacity, 2) cellular diversity, and 3) metabolic and neuroendocrine signaling activity, while outer blubber may have higher 1) extracellular matrix synthesis activity and 2) responsiveness to pathogens and cell stressors. We further characterized expression of nine genes of interest identified by transcriptomics and two adipokines with higher precision across blubber layers using targeted assays. Our study provides functional insights into stratification of blubber in marine mammals and a molecular key, including CD144, CD4, HMGCS2, GABRG2, HCAR2, and COL1A2, for distinguishing blubber layers for physiological and functional studies in seals.
Collapse
Affiliation(s)
- J. I. Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States,*Correspondence: J. I. Khudyakov,
| | - K. N. Allen
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| | - D. E. Crocker
- Department of Biology, Sonoma State University, Rohnert Park, CA, United States
| | - N. S. Trost
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - A. H. Roberts
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - L. Pirard
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - C. Debier
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - E. R. Piotrowski
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - J. P. Vázquez-Medina
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
19
|
Mansouri S, Gogoi H, Patel S, Katikaneni DS, Singh A, Aybar-Torres A, de Lartigue G, Jin L. MPYS Modulates Fatty Acid Metabolism and Immune Tolerance at Homeostasis Independent of Type I IFNs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2114-2132. [PMID: 36261171 PMCID: PMC9679991 DOI: 10.4049/jimmunol.2200158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/15/2022] [Indexed: 01/04/2023]
Abstract
MPYS/STING (stimulator of IFN genes) senses cyclic dinucleotides (CDNs), generates type I IFNs, and plays a critical role in infection, inflammation, and cancer. In this study, analyzing genotype and haplotype data from the 1000 Genomes Project, we found that the R71H-G230A-R293Q (HAQ) MPYS allele frequency increased 57-fold in East Asians compared with sub-Saharan Africans. Meanwhile, the G230A-R293Q (AQ) allele frequency decreased by 98% in East Asians compared with sub-Saharan Africans. We propose that the HAQ and AQ alleles underwent a natural selection during the out-of-Africa migration. We used mouse models of HAQ and AQ to investigate the underlying mechanism. We found that the mice carrying the AQ allele, which disappeared in East Asians, had normal CDN-type I IFN responses. Adult AQ mice, however, had less fat mass than did HAQ or wild-type mice on a chow diet. AQ epididymal adipose tissue had increased regulatory T cells and M2 macrophages with protein expression associated with enhanced fatty acid oxidation. Conditional knockout mice and adoptive cell transfer indicate a macrophage and regulatory T cell-intrinsic role of MPYS in fatty acid metabolism. Mechanistically, AQ/IFNAR1-/- mice had a similar lean phenotype as for the AQ mice. MPYS intrinsic tryptophan fluorescence revealed that the R71H change increased MPYS hydrophilicity. Lastly, we found that the second transmembrane (TM) and the TM2-TM3 linker region of MPYS interact with activated fatty acid, fatty acyl-CoA. In summary, studying the evolution of the human MPYS gene revealed an MPYS function in modulating fatty acid metabolism that may be critical during the out-of-Africa migration.
Collapse
Affiliation(s)
- Samira Mansouri
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Himanshu Gogoi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Seema Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Divya S. Katikaneni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Arashdeep Singh
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL; and
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL
| | - Alexandra Aybar-Torres
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Guillaume de Lartigue
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL; and
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
20
|
Lam PH, Chen E, Chiang JJ, Miller GE. Socioeconomic disadvantage, chronic stress, and proinflammatory phenotype: an integrative data analysis across the lifecourse. PNAS NEXUS 2022; 1:pgac219. [PMID: 36329724 PMCID: PMC9615129 DOI: 10.1093/pnasnexus/pgac219] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Socioeconomic disadvantage confers risk for many chronic illnesses, and theories have highlighted chronic psychological stress and alterations to inflammatory processes as key pathways. Specifically, disadvantage can heighten chronic stress, which may promote a proinflammatory phenotype characterized by immune cells mounting exaggerated cytokine responses to challenge and being less sensitive to inhibitory signals. Importantly, lifecourse perspectives emphasize that such immune alterations should be more potent earlier in life during a sensitive period when bodily tissues are highly plastic to environmental inputs. However, examining these propositions is resource intensive, as they require cell-culturing approaches to model functional inflammatory activities, a wide age range, and longitudinal data. Here, we integrated data from five independent studies to create a diverse sample of 1,607 individuals (960 with longitudinal data; 8 to 64 years old; 359 Asian, 205 Black, and 151 Latino/a). Leveraging the resulting lifecourse data, rich interview assessments of disadvantage and stress, and ex vivo assessments of inflammation, we examined two questions: (1) Does chronic stress account for the link between disadvantage and proinflammatory phenotype? (2) Is there a developmental period during which inflammatory responses are more sensitive to disadvantage and chronic stress? Disadvantage was associated with higher chronic stress, which was linked with a proinflammatory phenotype cross-sectionally, longitudinally, and in terms of prospective change across 1.5 to 2 years. Consistent with the sensitive period hypothesis, the magnitude of these indirect associations was strongest in earlier decades and declined across the lifecourse. These findings highlight the importance of taking a lifecourse perspective in examining health disparities.
Collapse
Affiliation(s)
- Phoebe H Lam
- Department of Psychology, Northwestern University, Swift Hall, 2029 Sheridan Road, Evanston, IL 60208, USA
| | - Edith Chen
- Department of Psychology, Northwestern University, Swift Hall, 2029 Sheridan Road, Evanston, IL 60208, USA
- Institute for Policy Research, Northwestern University, 2040 Sheridan Road, Evanston, IL 60208, USA
| | - Jessica J Chiang
- Department of Psychology, Georgetown University, 306 N White-Gravenor Hall, 37th and O Streets, NW, Washington DC, 20057, USA
| | - Gregory E Miller
- Department of Psychology, Northwestern University, Swift Hall, 2029 Sheridan Road, Evanston, IL 60208, USA
- Institute for Policy Research, Northwestern University, 2040 Sheridan Road, Evanston, IL 60208, USA
| |
Collapse
|
21
|
Fernandez NC, Shinoda K. The Role of B Lymphocyte Subsets in Adipose Tissue Development, Metabolism, and Aging. Compr Physiol 2022; 12:4133-4145. [PMID: 35950657 DOI: 10.1002/cphy.c220006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adipose tissue contains resident B lymphocytes (B cells) with varying immune functions and mechanisms, depending on the adipose depot type and location. The heterogeneity of B cells and their functions affect the immunometabolism of the adipose tissue in aging and age-associated metabolic disorders. B cells exist in categorizations of subsets that have developmental or phenotypic differences with varying functionalities. Subsets can be categorized as either protective or pathogenic depending on their secretion profile or involvement in metabolic maintenance. In this article, we summarized recent finding on the B cell heterogeneity and discuss how we can utilize our current knowledge of adipose resident B lymphocytes for potential treatment for age-associated metabolic disorders. © 2022 American Physiological Society. Compr Physiol 12: 1-13, 2022.
Collapse
Affiliation(s)
- Nicole C Fernandez
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kosaku Shinoda
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Division of Endocrinology & Diabetes, Albert Einstein College of Medicine, Bronx, New York, USA
- Fleischer Institute for Diabetes and Metabolism, Bronx, New York, USA
| |
Collapse
|
22
|
Luo R, Li X, Wang D. Reprogramming Macrophage Metabolism and its Effect on NLRP3 Inflammasome Activation in Sepsis. Front Mol Biosci 2022; 9:917818. [PMID: 35847986 PMCID: PMC9276983 DOI: 10.3389/fmolb.2022.917818] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/09/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis, the most common life-threatening multi-organ dysfunction syndrome secondary to infection, lacks specific therapeutic strategy due to the limited understanding of underlying mechanisms. It is currently believed that inflammasomes play critical roles in the development of sepsis, among which NLRP3 inflammasome is involved to most extent. Recent studies have revealed that dramatic reprogramming of macrophage metabolism is commonly occurred in sepsis, and this dysregulation is closely related with the activation of NLRP3 inflammasome. In view of the fact that increasing evidence demonstrates the mechanism of metabolism reprogramming regulating NLRP3 activation in macrophages, the key enzymes and metabolites participated in this regulation should be clearer for better interpreting the relationship of NLRP3 inflammasome and sepsis. In this review, we thus summarized the detail mechanism of the metabolic reprogramming process and its important role in the NLRP3 inflammasome activation of macrophages in sepsis. This mechanism summarization will reveal the applicational potential of metabolic regulatory molecules in the treatment of sepsis.
Collapse
Affiliation(s)
- Ruiheng Luo
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xizhe Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Dan Wang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Dan Wang,
| |
Collapse
|
23
|
Abstract
Energy metabolism maintains the activation of intracellular and intercellular signal transduction, and plays a crucial role in immune response. Under environmental stimulation, immune cells change from resting to activation and trigger metabolic reprogramming. The immune system cells exhibit different metabolic characteristics when performing functions. The study of immune metabolism provides new insights into the function of immune cells, including how they differentiate, migrate and exert immune responses. Studies of immune cell energy metabolism are beginning to shed light on the metabolic mechanism of disease progression and reveal new ways to target inflammatory diseases such as autoimmune diseases, chronic viral infections, and cancer. Here, we discussed the relationship between immune cells and metabolism, and proposed the possibility of targeted metabolic process for disease treatment.
Collapse
|
24
|
Deshpande R, Lee B, Qiao Y, Grewal SS. TOR signalling is required for host lipid metabolic remodelling and survival following enteric infection in Drosophila. Dis Model Mech 2022; 15:dmm049551. [PMID: 35363274 PMCID: PMC9118046 DOI: 10.1242/dmm.049551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/22/2022] [Indexed: 12/29/2022] Open
Abstract
When infected by enteric pathogenic bacteria, animals need to initiate local and whole-body defence strategies. Although most attention has focused on the role of innate immune anti-bacterial responses, less is known about how changes in host metabolism contribute to host defence. Using Drosophila as a model system, we identify induction of intestinal target-of-rapamycin (TOR) kinase signalling as a key adaptive metabolic response to enteric infection. We find that enteric infection induces both local and systemic induction of TOR independently of the Immune deficiency (IMD) innate immune pathway, and we see that TOR functions together with IMD signalling to promote infection survival. These protective effects of TOR signalling are associated with remodelling of host lipid metabolism. Thus, we see that TOR is required to limit excessive infection-mediated wasting of host lipid stores by promoting an increase in the levels of gut- and fat body-expressed lipid synthesis genes. Our data support a model in which induction of TOR represents a host tolerance response to counteract infection-mediated lipid wasting in order to promote survival. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | | | | | - Savraj S. Grewal
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
25
|
Bapat SP, Whitty C, Mowery CT, Liang Y, Yoo A, Jiang Z, Peters MC, Zhang LJ, Vogel I, Zhou C, Nguyen VQ, Li Z, Chang C, Zhu WS, Hastie AT, He H, Ren X, Qiu W, Gayer SG, Liu C, Choi EJ, Fassett M, Cohen JN, Sturgill JL, Crotty Alexander LE, Suh JM, Liddle C, Atkins AR, Yu RT, Downes M, Liu S, Nikolajczyk BS, Lee IK, Guttman-Yassky E, Ansel KM, Woodruff PG, Fahy JV, Sheppard D, Gallo RL, Ye CJ, Evans RM, Zheng Y, Marson A. Obesity alters pathology and treatment response in inflammatory disease. Nature 2022; 604:337-342. [PMID: 35355021 PMCID: PMC9165753 DOI: 10.1038/s41586-022-04536-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 02/08/2022] [Indexed: 12/17/2022]
Abstract
Decades of work have elucidated cytokine signalling and transcriptional pathways that control T cell differentiation and have led the way to targeted biologic therapies that are effective in a range of autoimmune, allergic and inflammatory diseases. Recent evidence indicates that obesity and metabolic disease can also influence the immune system1-7, although the mechanisms and effects on immunotherapy outcomes remain largely unknown. Here, using two models of atopic dermatitis, we show that lean and obese mice mount markedly different immune responses. Obesity converted the classical type 2 T helper (TH2)-predominant disease associated with atopic dermatitis to a more severe disease with prominent TH17 inflammation. We also observed divergent responses to biologic therapies targeting TH2 cytokines, which robustly protected lean mice but exacerbated disease in obese mice. Single-cell RNA sequencing coupled with genome-wide binding analyses revealed decreased activity of nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) in TH2 cells from obese mice relative to lean mice. Conditional ablation of PPARγ in T cells revealed that PPARγ is required to focus the in vivo TH response towards a TH2-predominant state and prevent aberrant non-TH2 inflammation. Treatment of obese mice with a small-molecule PPARγ agonist limited development of TH17 pathology and unlocked therapeutic responsiveness to targeted anti-TH2 biologic therapies. These studies reveal the effects of obesity on immunological disease and suggest a precision medicine approach to target the immune dysregulation caused by obesity.
Collapse
Affiliation(s)
- Sagar P Bapat
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA.
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
- Medical Scientist Training Program, University of California, San Diego, La Jolla, CA, USA.
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Caroline Whitty
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Cody T Mowery
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
| | - Yuqiong Liang
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Arum Yoo
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Zewen Jiang
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Michael C Peters
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ling-Juan Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Ian Vogel
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Carmen Zhou
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Vinh Q Nguyen
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Zhongmei Li
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Christina Chang
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Wandi S Zhu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | - Annette T Hastie
- School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Helen He
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xin Ren
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Wenli Qiu
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah G Gayer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Chang Liu
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Eun Jung Choi
- Department of Biomedical Science, Graduate School, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Marlys Fassett
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Jarish N Cohen
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Jamie L Sturgill
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Kentucky, Lexington, KY, USA
| | - Laura E Crotty Alexander
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, CA, USA
| | - Jae Myoung Suh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Sydney Medical School, Westmead Hospital, University of Sydney, Westmead, New South Wales, Australia
| | - Annette R Atkins
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ruth T Yu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sihao Liu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Barbara S Nikolajczyk
- Department of Pharmacology and Nutritional Sciences and the Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, USA
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - K Mark Ansel
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | - Prescott G Woodruff
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
| | - John V Fahy
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
| | - Dean Sheppard
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Richard L Gallo
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chun Jimmie Ye
- Institute for Human Genetics (IHG), University of California, San Francisco, San Francisco, CA, USA
- Institute for Computational Health Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Ronald M Evans
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Ye Zheng
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Human Genetics (IHG), University of California, San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA.
| |
Collapse
|
26
|
Role of Distinct Fat Depots in Metabolic Regulation and Pathological Implications. Rev Physiol Biochem Pharmacol 2022; 186:135-176. [PMID: 35915363 DOI: 10.1007/112_2022_73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
People suffering from obesity and associated metabolic disorders including diabetes are increasing exponentially around the world. Adipose tissue (AT) distribution and alteration in their biochemical properties play a major role in the pathogenesis of these diseases. Emerging evidence suggests that AT heterogeneity and depot-specific physiological changes are vital in the development of insulin resistance in peripheral tissues like muscle and liver. Classically, AT depots are classified into white adipose tissue (WAT) and brown adipose tissue (BAT); WAT is the site of fatty acid storage, while BAT is a dedicated organ of metabolic heat production. The discovery of beige adipocyte clusters in WAT depots indicates AT heterogeneity has a more central role than hither to ascribed. Therefore, we have discussed in detail the current state of understanding on cellular and molecular origin of different AT depots and their relevance toward physiological metabolic homeostasis. A major focus is to highlight the correlation between altered WAT distribution in the body and metabolic pathogenesis in animal models and humans. We have also underscored the disparity in the molecular (including signaling) changes in various WAT tissues during diabetic pathogenesis. Exercise-mediated beneficial alteration in WAT physiology/distribution that protects against metabolic disorders is evolving. Here we have discussed the depot-specific biochemical adjustments induced by different forms of exercise. A detailed understanding of the molecular details of inter-organ crosstalk via substrate utilization/storage and signaling through chemokines provide strategies to target selected WAT depots to pharmacologically mimic the benefits of exercise countering metabolic diseases including diabetes.
Collapse
|
27
|
Chen H, Sun L, Feng L, Yin Y, Zhang W. Role of Innate lymphoid Cells in Obesity and Insulin Resistance. Front Endocrinol (Lausanne) 2022; 13:855197. [PMID: 35574038 PMCID: PMC9091334 DOI: 10.3389/fendo.2022.855197] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity, a growing chronic metabolic disease, greatly increases the risk of metabolic syndrome which includes type 2 diabetes, fatty liver and cardiovascular diseases. Obesity-associated metabolic diseases significantly contribute to mortality and reduce life expectancy. Recently, innate lymphoid cells (ILCs) have emerged as crucial regulators of metabolic homeostasis and tissue inflammation. This review focuses on the roles of ILCs in different metabolic tissues, including adipose tissue, liver, pancreas, and intestine. We briefly outline the relationship between obesity, inflammation, and insulin resistance. We then discuss how ILCs in distinct metabolic organs may function to maintain metabolic homeostasis and contribute to obesity and its associated metabolic diseases. The potential of ILCs as the therapeutic target for obesity and insulin resistance is also addressed.
Collapse
Affiliation(s)
- Hong Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Lu Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
- *Correspondence: Weizhen Zhang, ; Yue Yin,
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States
- *Correspondence: Weizhen Zhang, ; Yue Yin,
| |
Collapse
|
28
|
Trapecar M. Multiorgan microphysiological systems as tools to interrogate interorgan crosstalk and complex diseases. FEBS Lett 2021; 596:681-695. [PMID: 34923635 DOI: 10.1002/1873-3468.14260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
Metabolic and inflammatory disorders such as autoimmune and neurodegenerative diseases are increasing at alarming rates. Many of these are not tissue-specific occurrences but complex, systemic pathologies of unknown origin for which no cure exists. Such complexity obscures causal relationships among factors regulating disease progression. Emerging technologies mimicking human physiology, such as microphysiological systems (MPSs), offer new possibilities to provide clarity in systemic metabolic and inflammatory diseases. Controlled interaction of multiple MPSs and the scalability of biological complexity in MPSs, supported by continuous multiomic monitoring, might hold the key to identifying novel relationships between interorgan crosstalk, metabolism, and immunity. In this perspective, I aim to discuss the current state of modeling multiorgan physiology and evaluate current opportunities and challenges.
Collapse
Affiliation(s)
- Martin Trapecar
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
29
|
Resident and migratory adipose immune cells control systemic metabolism and thermogenesis. Cell Mol Immunol 2021; 19:421-431. [PMID: 34837070 PMCID: PMC8891307 DOI: 10.1038/s41423-021-00804-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023] Open
Abstract
Glucose is a vital source of energy for all mammals. The balance between glucose uptake, metabolism and storage determines the energy status of an individual, and perturbations in this balance can lead to metabolic diseases. The maintenance of organismal glucose metabolism is a complex process that involves multiple tissues, including adipose tissue, which is an endocrine and energy storage organ that is critical for the regulation of systemic metabolism. Adipose tissue consists of an array of different cell types, including specialized adipocytes and stromal and endothelial cells. In addition, adipose tissue harbors a wide range of immune cells that play vital roles in adipose tissue homeostasis and function. These cells contribute to the regulation of systemic metabolism by modulating the inflammatory tone of adipose tissue, which is directly linked to insulin sensitivity and signaling. Furthermore, these cells affect the control of thermogenesis. While lean adipose tissue is rich in type 2 and anti-inflammatory cytokines such as IL-10, obesity tips the balance in favor of a proinflammatory milieu, leading to the development of insulin resistance and the dysregulation of systemic metabolism. Notably, anti-inflammatory immune cells, including regulatory T cells and innate lymphocytes, protect against insulin resistance and have the characteristics of tissue-resident cells, while proinflammatory immune cells are recruited from the circulation to obese adipose tissue. Here, we review the key findings that have shaped our understanding of how immune cells regulate adipose tissue homeostasis to control organismal metabolism.
Collapse
|
30
|
McLaughlin T, Schnittger I, Nagy A, Zanley E, Xu Y, Song Y, Nieman K, Tremmel JA, Dey D, Boyd J, Sacks H. Relationship Between Coronary Atheroma, Epicardial Adipose Tissue Inflammation, and Adipocyte Differentiation Across the Human Myocardial Bridge. J Am Heart Assoc 2021; 10:e021003. [PMID: 34726081 PMCID: PMC8751937 DOI: 10.1161/jaha.121.021003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Background Inflammation in epicardial adipose tissue (EAT) may contribute to coronary atherosclerosis. Myocardial bridge is a congenital anomaly in which the left anterior descending coronary artery takes a "tunneled" course under a bridge of myocardium: while atherosclerosis develops in the proximal left anterior descending coronary artery, the bridged portion is spared, highlighting the possibility that geographic separation from inflamed EAT is protective. We tested the hypothesis that inflammation in EAT was related to atherosclerosis by comparing EAT from proximal and bridge depots in individuals with myocardial bridge and varying degrees of atherosclerotic plaque. Methods and Results Maximal plaque burden was quantified by intravascular ultrasound, and inflammation was quantified by pericoronary EAT signal attenuation (pericoronary adipose tissue attenuation) from cardiac computed tomography scans. EAT overlying the proximal left anterior descending coronary artery and myocardial bridge was harvested for measurement of mRNA and microRNA (miRNA) using custom chips by Nanostring; inflammatory cytokines were measured in tissue culture supernatants. Pericoronary adipose tissue attenuation was increased, indicating inflammation, in proximal versus bridge EAT, in proportion to atherosclerotic plaque. Individuals with moderate-high versus low plaque burden exhibited greater expression of inflammation and hypoxia genes, and lower expression of adipogenesis genes. Comparison of gene expression in proximal versus bridge depots revealed differences only in participants with moderate-high plaque: inflammation was higher in proximal and adipogenesis lower in bridge EAT. Secreted inflammatory cytokines tended to be higher in proximal EAT. Hypoxia-inducible factor 1a was highly associated with inflammatory gene expression. Seven miRNAs were differentially expressed by depot: 3192-5P, 518D-3P, and 532-5P were upregulated in proximal EAT, whereas miR 630, 575, 16-5P, and 320E were upregulated in bridge EAT. miR 630 correlated directly with plaque burden and inversely with adipogenesis genes. miR 3192-5P, 518D-3P, and 532-5P correlated inversely with hypoxia/oxidative stress, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PCG1a), adipogenesis, and angiogenesis genes. Conclusions Inflammation is specifically elevated in EAT overlying atherosclerotic plaque, suggesting that EAT inflammation is caused by atherogenic molecular signals, including hypoxia-inducible factor 1a and/or miRNAs in an "inside-to-out" relationship. Adipogenesis was suppressed in the bridge EAT, but only in the presence of atherosclerotic plaque, supporting cross talk between the vasculature and EAT. miR 630 in EAT, expressed differentially according to burden of atherosclerotic plaque, and 3 other miRNAs appear to inhibit key genes related to adipogenesis, angiogenesis, hypoxia/oxidative stress, and thermogenesis in EAT, highlighting a role for miRNA in mediating cross talk between the coronary vasculature and EAT.
Collapse
Affiliation(s)
- Tracey McLaughlin
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Ingela Schnittger
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Anna Nagy
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Elizabeth Zanley
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Yue Xu
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Yanqiu Song
- Cardiovascular Institute Tianjin Chest Hospital Tianjin China
| | - Koen Nieman
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Jennifer A Tremmel
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Damini Dey
- Department of Biomedical Sciences and Medicine Cedars-Sinai Medical Center Biomedical Imaging Research Institute Los Angeles CA
| | - Jack Boyd
- Department of Cardiothoracic Surgery Stanford University School of Medicine Stanford CA
| | - Harold Sacks
- Division of Endocrinology Department of Medicine David Geffen School of Medicine at UCLA Los Angeles CA
| |
Collapse
|
31
|
Spiljar M, Steinbach K, Rigo D, Suárez-Zamorano N, Wagner I, Hadadi N, Vincenti I, Page N, Klimek B, Rochat MA, Kreutzfeldt M, Chevalier C, Stojanović O, Bejuy O, Colin D, Mack M, Cansever D, Greter M, Merkler D, Trajkovski M. Cold exposure protects from neuroinflammation through immunologic reprogramming. Cell Metab 2021; 33:2231-2246.e8. [PMID: 34687652 PMCID: PMC8570411 DOI: 10.1016/j.cmet.2021.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 07/24/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023]
Abstract
Autoimmunity is energetically costly, but the impact of a metabolically active state on immunity and immune-mediated diseases is unclear. Ly6Chi monocytes are key effectors in CNS autoimmunity with an elusive role in priming naive autoreactive T cells. Here, we provide unbiased analysis of the immune changes in various compartments during cold exposure and show that this energetically costly stimulus markedly ameliorates active experimental autoimmune encephalomyelitis (EAE). Cold exposure decreases MHCII on monocytes at steady state and in various inflammatory mouse models and suppresses T cell priming and pathogenicity through the modulation of monocytes. Genetic or antibody-mediated monocyte depletion or adoptive transfer of Th1- or Th17-polarized cells for EAE abolishes the cold-induced effects on T cells or EAE, respectively. These findings provide a mechanistic link between environmental temperature and neuroinflammation and suggest competition between cold-induced metabolic adaptations and autoimmunity as energetic trade-off beneficial for the immune-mediated diseases.
Collapse
Affiliation(s)
- Martina Spiljar
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Karin Steinbach
- Department of Pathology and Immunology, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland
| | - Dorothée Rigo
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nicolas Suárez-Zamorano
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland
| | - Noushin Hadadi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ilena Vincenti
- Department of Pathology and Immunology, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland
| | - Nicolas Page
- Department of Pathology and Immunology, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland
| | - Bogna Klimek
- Department of Pathology and Immunology, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland
| | - Mary-Aude Rochat
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland
| | - Claire Chevalier
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ozren Stojanović
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Olivia Bejuy
- CIBM Centre for BioMedical Imaging, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Didier Colin
- Small Animal Preclinical Imaging Platform, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Matthias Mack
- Department of Internal Medicine II - Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Dilay Cansever
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospitals, Geneva, Switzerland.
| | - Mirko Trajkovski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire (CMU), University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
32
|
Abstract
This study explored the associations between specific profiles of biological dysregulation and mental health outcomes in a national, community sample of healthy adults in the United States. A latent class analysis of data from the Midlife Development in the United States study (n = 1,757) was conducted to determine classes of biological dysregulation. Multinomial logistic regressions of class membership were employed to determine associations with measures related to depression, including whether or not individuals had sought treatment, Center for Epidemiological Studies Depression Scale, and both the generalized distress and anhedonia subscales of the Mood and Anxiety Symptoms Questionnaire. Four classes of dysregulation emerged: baseline/low dysregulation, metabolic and inflammatory dysregulation, parasympathetic dysregulation, and SAM pathway dysregulation. Individuals who met the criteria for depression measures were more likely to be in the metabolic and immune dysregulation and parasympathetic dysregulation groups as compared to the baseline group. The results suggest that mental health outcomes, such as depression, are differentially associated with specific profiles of biological dysregulation. A more nuanced approach to profiles of dysregulation could better inform treatment decisions.Lay summaryHigher levels of allostatic load, which represents the cumulative wear and tear of exposure to stress, are associated with increased rates of depression and anhedonia. Specifically, parasympathetic dysregulation and immunometabolic dysregulation are associated with negative mental health outcomes.
Collapse
Affiliation(s)
- Jason T Carbone
- School of Social Work, Integrative Biosciences (IBio) Center, Wayne State University, Detroit, MI, USA
| |
Collapse
|
33
|
Zhang G, Ma L, Bai L, Li M, Guo T, Tian B, He Z, Fu Q. Inflammatory microenvironment-targeted nanotherapies. J Control Release 2021; 334:114-126. [PMID: 33887284 DOI: 10.1016/j.jconrel.2021.04.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022]
Abstract
Inflammatory microenvironments (IMEs) are common pathological characteristics and drive the development of multiple chronic diseases. Thus, IME-targeted therapies exhibit potential for the treatment of inflammatory diseases. Nanoplatforms have significant advantages in improving the efficiency of anti-inflammatory treatments. Owing to their improved therapeutic effects and reduced side effects, IME-targeted nanotherapies have recently drawn interest from the research community. This review introduces IMEs and discusses the application of IME-targeted nanotherapies for inflammatory diseases. The development of rational targeting strategies tailored to IMEs in damaged tissues can help promote therapies for chronic diseases.
Collapse
Affiliation(s)
- Guangshuai Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Lixue Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Lijun Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Mo Li
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Tiange Guo
- Laboratory Animal Department, General Hospital of Northern Theater Command, No. 83, Wenhua Road, Shenyang 110016, China
| | - Baocheng Tian
- School of Pharmacy, Binzhou Medical University, No. 346, Guanhai Road, Yantai 264003, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
34
|
Villarreal-Calderon JR, Cuellar-Tamez R, Castillo EC, Luna-Ceron E, García-Rivas G, Elizondo-Montemayor L. Metabolic shift precedes the resolution of inflammation in a cohort of patients undergoing bariatric and metabolic surgery. Sci Rep 2021; 11:12127. [PMID: 34108550 PMCID: PMC8190106 DOI: 10.1038/s41598-021-91393-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Bariatric and metabolic surgery has shown to promote weight loss and reduce systemic inflammation. However, the sequence and timing of events regarding metabolic improvement and inflammation resolution has been rarely explored. Furthermore, data on inflammatory markers of Th17 and Th1 cell responses after bariatric surgery is scarce. We conducted a prospective study in subjects with obesity that underwent bariatric and metabolic surgery, with follow-ups at 3 and 6 months. Anthropometric and metabolic markers such as insulin levels, HOMA-IR, and lipid parameters declined significantly 3 months after surgery; while hs-CRP, TNF-α, IL-1β, IL-6, and IL-8 serum concentrations decreased 6 months after the procedure. Concentrations of Th1 signature and driver cytokines, particularly IFN-γ, IL-12, and IL-18, and of Th17 driver IL-23 also decreased significantly after 6 months. Significant positive correlations between triglyceride levels and hs-CRP, IL-1β, and IFN-γ concentrations, and between Apo B and IFN-γ levels were observed 6 months after bariatric and metabolic surgery. In addition, BMI was associated with hs-CRP and TNF-α concentrations. Fat mass correlated with hs-CRP, TNF-α, and IL-12. Analysis of the temporality of metabolic and inflammatory events suggests that improvement in the metabolic status occurs before resolution of systemic inflammation and may be a requisite for the later event.
Collapse
Affiliation(s)
- Jose Romeo Villarreal-Calderon
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 64710, Monterrey, Mexico.,Tecnologico de Monterrey, Centro de Investigación en Obesidad y Nutrición Clínica, 64710, Monterrey, Mexico.,Tecnologico de Monterrey. Cardiovascular Medicine and Metabolomics Research Group, Hospital Zambrano Hellion, TecSalud, 66278, San Pedro Garza García, Mexico
| | - Ricardo Cuellar-Tamez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 64710, Monterrey, Mexico.,Tecnologico de Monterrey. Cardiovascular Medicine and Metabolomics Research Group, Hospital Zambrano Hellion, TecSalud, 66278, San Pedro Garza García, Mexico
| | - Elena C Castillo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 64710, Monterrey, Mexico.,Tecnologico de Monterrey. Cardiovascular Medicine and Metabolomics Research Group, Hospital Zambrano Hellion, TecSalud, 66278, San Pedro Garza García, Mexico
| | - Eder Luna-Ceron
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 64710, Monterrey, Mexico.,Tecnologico de Monterrey. Cardiovascular Medicine and Metabolomics Research Group, Hospital Zambrano Hellion, TecSalud, 66278, San Pedro Garza García, Mexico
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 64710, Monterrey, Mexico. .,Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, 66278, San Pedro Garza García, Mexico. .,Tecnologico de Monterrey. Cardiovascular Medicine and Metabolomics Research Group, Hospital Zambrano Hellion, TecSalud, 66278, San Pedro Garza García, Mexico.
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 64710, Monterrey, Mexico. .,Tecnologico de Monterrey, Centro de Investigación en Obesidad y Nutrición Clínica, 64710, Monterrey, Mexico. .,Tecnologico de Monterrey. Cardiovascular Medicine and Metabolomics Research Group, Hospital Zambrano Hellion, TecSalud, 66278, San Pedro Garza García, Mexico.
| |
Collapse
|
35
|
Zhou Z, Tao Y, Zhao H, Wang Q. Adipose Extracellular Vesicles: Messengers From and to Macrophages in Regulating Immunometabolic Homeostasis or Disorders. Front Immunol 2021; 12:666344. [PMID: 34108967 PMCID: PMC8183682 DOI: 10.3389/fimmu.2021.666344] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is comprised of heterogenous cell populations that regulate both energy metabolism and immune reactions. Macrophages play critical roles in regulating immunometabolic homeostasis or disorders through cooperation with adipocytes, adipose tissue-derived stem cells (ADSCs) or other cells in adipose tissue. Extracellular vesicles (EVs) are recently recognized as efficient messengers for intercellular communication. Emerging evidences have demonstrated that adipose EVs are actively involved in the mutual interactions of macrophages, adipocytes and ADSCs, which produce considerable influences on immunometabolism under healthy or obese conditions. Here, we will elaborate the production and the characteristics of adipose EVs that are related to macrophages under different metabolic demands or stresses, whilst discuss the roles of these EVs in regulating local or systemic immunometabolic homeostasis or disorders in the context of adipocyte-macrophage dialogue and ADSC-macrophage interaction. Particularly, we provide a profile of dynamic adipose microenvironments based on macrophages. Adipose EVs act as the messengers between ADSCs and macrophages to maintain the balance of metabolism and immunity, while drive a vicious cycle between hypertrophic adipocytes and inflammatory macrophages to cause immunometabolic imbalance. This review may provide valuable information about the physio- or pathological roles of adipose EVs and the application of adipose EVs in the diagnosis and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Zixin Zhou
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Tao
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Zhao
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
36
|
Michailidou Z, Gomez-Salazar M, Alexaki VI. Innate Immune Cells in the Adipose Tissue in Health and Metabolic Disease. J Innate Immun 2021; 14:4-30. [PMID: 33849008 DOI: 10.1159/000515117] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/09/2021] [Indexed: 11/19/2022] Open
Abstract
Metabolic disorders, such as obesity, type 2 diabetes mellitus, and nonalcoholic fatty liver disease, are characterized by chronic low-grade tissue and systemic inflammation. During obesity, the adipose tissue undergoes immunometabolic and functional transformation. Adipose tissue inflammation is driven by innate and adaptive immune cells and instigates insulin resistance. Here, we discuss the role of innate immune cells, that is, macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid type 2 cells, dendritic cells, and mast cells, in the adipose tissue in the healthy (lean) and diseased (obese) state and describe how their function is shaped by the obesogenic microenvironment, and humoral, paracrine, and cellular interactions. Moreover, we particularly outline the role of hypoxia as a central regulator in adipose tissue inflammation. Finally, we discuss the long-lasting effects of adipose tissue inflammation and its potential reversibility through drugs, caloric restriction, or exercise training.
Collapse
Affiliation(s)
- Zoi Michailidou
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Mario Gomez-Salazar
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
37
|
Bechara R, McGeachy MJ, Gaffen SL. The metabolism-modulating activity of IL-17 signaling in health and disease. J Exp Med 2021; 218:211951. [PMID: 33822846 PMCID: PMC8025242 DOI: 10.1084/jem.20202191] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/08/2021] [Accepted: 02/25/2021] [Indexed: 12/22/2022] Open
Abstract
IL-17 was discovered nearly 30 yr ago, but it has only been recently appreciated that a key function of this cytokine is to orchestrate cellular and organismal metabolism. Indeed, metabolic regulation is integrated into both the physiological and the pathogenic aspects of IL-17 responses. Thus, understanding the interplay between IL-17 and downstream metabolic processes could ultimately inform therapeutic opportunities for diseases involving IL-17, including some not traditionally linked to this cytokine pathway. Here, we discuss the emerging pathophysiological roles of IL-17 related to cellular and organismal metabolism, including metabolic regulation of IL-17 signal transduction.
Collapse
Affiliation(s)
- Rami Bechara
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Mandy J McGeachy
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Sarah L Gaffen
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
38
|
Cruz-Pineda WD, Parra-Rojas I, Rodríguez-Ruíz HA, Illades-Aguiar B, Matia-García I, Garibay-Cerdenares OL. The regulatory role of insulin in energy metabolism and leukocyte functions. J Leukoc Biol 2021; 111:197-208. [PMID: 33724523 PMCID: PMC9291603 DOI: 10.1002/jlb.2ru1220-847r] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
Insulin is the hormone responsible for maintaining glucose homeostasis in the body, in addition to participating in lipid metabolism, protein synthesis, and the inhibition of gluconeogenesis. These functions are well characterized in the classic organ target cells that are responsible for general energy regulation: the liver, skeletal muscle, and adipose tissue. However, these actions are not restricted to these tissues because insulin has been shown to affect most cells in the body. This review describes the role of insulin in leukocyte signaling pathways, metabolism and functions, and how insulin resistance could affect this signaling and deteriorate leukocyte metabolism and function, in addition to showing evidence that suggests leukocytes may substantially contribute to the development of systemic insulin resistance.
Collapse
Affiliation(s)
- Walter David Cruz-Pineda
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Isela Parra-Rojas
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Hugo Alberto Rodríguez-Ruíz
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico.,Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Inés Matia-García
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Olga Lilia Garibay-Cerdenares
- CONACyT-Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico.,Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| |
Collapse
|
39
|
Kunz N, Kemper C. Complement Has Brains-Do Intracellular Complement and Immunometabolism Cooperate in Tissue Homeostasis and Behavior? Front Immunol 2021; 12:629986. [PMID: 33717157 PMCID: PMC7946832 DOI: 10.3389/fimmu.2021.629986] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
The classical liver-derived and serum-effective complement system is well appreciated as a key mediator of host protection via instruction of innate and adaptive immunity. However, recent studies have discovered an intracellularly active complement system, the complosome, which has emerged as a central regulator of the core metabolic pathways fueling human immune cell activity. Induction of expression of components of the complosome, particularly complement component C3, during transmigration from the circulation into peripheral tissues is a defining characteristic of monocytes and T cells in tissues. Intracellular complement activity is required to induce metabolic reprogramming of immune cells, including increased glycolytic flux and OXPHOS, which drive the production of the pro-inflammatory cytokine IFN-γ. Consequently, reduced complosome activity translates into defects in normal monocyte activation, faulty Th1 and cytotoxic T lymphocyte responses and loss of protective tissue immunity. Intriguingly, neurological research has identified an unexpected connection between the physiological presence of innate and adaptive immune cells and certain cytokines, including IFN-γ, in and around the brain and normal brain function. In this opinion piece, we will first review the current state of research regarding complement driven metabolic reprogramming in the context of immune cell tissue entry and residency. We will then discuss how published work on the role of IFN-γ and T cells in the brain support a hypothesis that an evolutionarily conserved cooperation between the complosome, cell metabolism and IFN-γ regulates organismal behavior, as well as immunity.
Collapse
Affiliation(s)
- Natalia Kunz
- Complement and Inflammation Research Section (CIRS), National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Claudia Kemper
- Complement and Inflammation Research Section (CIRS), National Heart, Lung and Blood Institute, Bethesda, MD, United States.,Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
40
|
Amadori M, Spelta C. The Autumn Low Milk Yield Syndrome in High Genetic Merit Dairy Cattle: The Possible Role of a Dysregulated Innate Immune Response. Animals (Basel) 2021; 11:ani11020388. [PMID: 33546430 PMCID: PMC7913622 DOI: 10.3390/ani11020388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 01/29/2023] Open
Abstract
Simple Summary Milk yield worldwide is dominated by few cosmopolitan dairy cattle breeds producing high production levels in the framework of hygiene standards that have dramatically improved over the years. Yet, there is evidence that such achievements have gone along with substantial animal health and welfare problems for many years, exemplified by reduced life expectancy and high herd replacement rates. Also, these animals are very susceptible to diverse environmental stressors, among which hot summer climate plays a central role in the occurrence of diverse disease cases underlying early cull from the herd. Milk production is also affected by heat stress, both directly and indirectly, as shown by low milk yield in the following autumn period. This article highlights the low milk yield syndrome and sets it into a conceptual framework, based on the crucial role of the innate immune system in the response to non-infectious stressors and in adaptation physiology at large. Abstract The analysis of milk yield data shows that high genetic merit dairy cows do not express their full production potential in autumn. Therefore, we focused on metabolic stress and inflammatory response in the dry and peripartum periods as possible causes thereof. It was our understanding that some cows could not cope with the stress imposed by their physiological and productive status by means of adequate adaptation strategies. Accordingly, this study highlights the noxious factors with a potential to affect cows in the above transition period: hot summer climate, adverse genetic traits, poor coping with unfavorable environmental conditions, outright production diseases and consequences thereof. In particular, the detrimental effects in the dry period of overcrowding, photoperiod change and heat stress on mammary gland development and milk production are highlighted in the context of the autumn low milk yield syndrome. The latter could be largely accounted for by a “memory” effect on the innate immune system induced in summer by diverse stressors after dry-off, according to strong circumstantial and indirect experimental evidence. The “memory” effect is based on distinct epigenetic changes of innate immunity genes, as already shown in cases of bovine mastitis. Following a primary stimulation, the innate immune system would be able to achieve a state known as “trained immunity”, a sort of “education” which modifies the response to the same or similar stressors upon a subsequent exposure. In our scenario, the “education” of the innate immune system would induce a major shift in the metabolism of inflammatory cells following their reprogramming. This would entail a higher basal consumption of glucose, in competition with the need for the synthesis of milk. Also, there is strong evidence that the inflammatory response generated in the dry period leads to a notable reduction of dry matter intake after calving, and to a reduced efficiency of oxidative phosphorylation in mitochondria. On the whole, an effective control of the stressors in the dry period is badly needed for better disease control and optimal production levels in dairy cattle.
Collapse
Affiliation(s)
- Massimo Amadori
- RNIV, Italian Society of Veterinary Immunology, 25125 Brescia, Italy
- Correspondence:
| | - Chiara Spelta
- Private Veterinary Practitioner, 27100 Pavia, Italy;
| |
Collapse
|
41
|
Zinöcker MK, Svendsen K, Dankel SN. The homeoviscous adaptation to dietary lipids (HADL) model explains controversies over saturated fat, cholesterol, and cardiovascular disease risk. Am J Clin Nutr 2021; 113:277-289. [PMID: 33471045 DOI: 10.1093/ajcn/nqaa322] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
SFAs play the leading role in 1 of the greatest controversies in nutrition science. Relative to PUFAs, SFAs generally increase circulating concentrations of LDL cholesterol, a risk factor for atherosclerotic cardiovascular disease (ASCVD). However, the purpose of regulatory mechanisms that control the diet-induced lipoprotein cholesterol dynamics is rarely discussed in the context of human adaptive biology. We argue that better mechanistic explanations can help resolve lingering controversies, with the potential to redefine aspects of research, clinical practice, dietary advice, public health management, and food policy. In this paper we propose a novel model, the homeoviscous adaptation to dietary lipids (HADL) model, which explains changes in lipoprotein cholesterol as adaptive homeostatic adjustments that serve to maintain cell membrane fluidity and hence optimal cell function. Due to the highly variable intake of fatty acids in humans and other omnivore species, we propose that circulating lipoproteins serve as a buffer to enable the rapid redistribution of cholesterol molecules between specific cells and tissues that is necessary with changes in dietary fatty acid supply. Hence, circulating levels of LDL cholesterol may change for nonpathological reasons. Accordingly, an SFA-induced raise in LDL cholesterol in healthy individuals could represent a normal rather than a pathologic response. These regulatory mechanisms may become disrupted secondarily to pathogenic processes in association with insulin resistance and the presence of other ASCVD risk factors, as supported by evidence showing diverging lipoprotein responses in healthy individuals as opposed to those with metabolic disorders such as insulin resistance and obesity. Corresponding with the model, we suggest alternative contributing factors to the association between elevated LDL cholesterol concentrations and ASCVD, involving dietary factors beyond SFAs, such as an increased endotoxin load from diet-gut microbiome interactions and subsequent chronic low-grade inflammation that interferes with fine-tuned signaling pathways.
Collapse
Affiliation(s)
| | - Karianne Svendsen
- Department of Nutrition, University of Oslo, Oslo, Norway.,The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Simon Nitter Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
42
|
Manti M, Stener-Victorin E, Benrick A. Skeletal Muscle Immunometabolism in Women With Polycystic Ovary Syndrome: A Meta-Analysis. Front Physiol 2020; 11:573505. [PMID: 33192572 PMCID: PMC7642984 DOI: 10.3389/fphys.2020.573505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/25/2020] [Indexed: 12/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine and metabolic disorder affecting up to 15% of women at reproductive age. The main features of PCOS are hyperandrogenism and irregular menstrual cycles together with metabolic dysfunctions including hyperinsulinemia and insulin resistance and a 4-fold increased risk of developing type 2 diabetes. Despite the high prevalence the pathophysiology of the syndrome is unclear. Insulin resistance in women with PCOS likely affect the skeletal muscle and recently it was demonstrated that changes in DNA methylation affects the gene expression in skeletal muscle that in part can explain their metabolic abnormalities. The objective of this work was to combine gene expression array data from different datasets to improve statistical power and thereby identify novel biomarkers that can be further explored. In this narrative review, we performed a meta-analysis of skeletal muscle arrays available from Gene Expression Omnibus and from publications. The eligibility criteria were published articles in English, and baseline (no treatment) skeletal muscle samples from women with PCOS and controls. The R package Metafor was used for integration of the datasets. One hundred and fourteen unique transcripts were differentially expressed in skeletal muscle from women with PCOS vs. controls (q < 0.05), 87% of these transcripts have not been previously identified as altered in PCOS muscle. ING2, CDKAL1, and AKTIP had the largest differential increase in expression, and TSHZ2, FKBP2, and OCEL1 had the largest decrease in expression. Two genes, IRX3 and CDKAL1 were consistently upregulated (q < 0.05) in the individual analyses and meta-analysis. Based on the meta-analysis, we identified several dysregulated immunometabolic pathways as a part of the molecular mechanisms of insulin resistance in the skeletal muscle of women with PCOS. The transcriptomic data need to be verified by functional analyses as well as proteomics to advance our understanding of PCOS specific insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Maria Manti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Anna Benrick
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,School of Health Sciences, University of Skövde, Skövde, Sweden
| |
Collapse
|
43
|
Abstract
SARS-CoV2 infection or COVID-19 has created panic around the world since its first origin in December 2019 in Wuhan city, China. The COVID-19 pandemic has infected more than 46.4 million people, with 1,199,727 deaths. The immune system plays a crucial role in the severity of COVID-19 and the development of pneumonia-induced acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). Along with providing protection, both innate and T cell-based adaptive immune response dysregulate during severe SARS-CoV2 infection. This dysregulation is more pronounced in older population and patients with comorbidities (Diabetes, hypertension, obesity, other pulmonary and autoimmune diseases). However, COVID-19 patients develop protective antibodies (Abs) against SARS-CoV2, but they do not long for last. The induction of the immune response against the pathogen also requires metabolic energy that generates through the process of immunometabolism. The change in the metabolic stage of immune cells from homeostasis to an inflammatory or infectious environment is called immunometabolic reprogramming. The article describes the cellular immunology (macrophages, T cells, B cells, dendritic cells, NK cells and pulmonary epithelial cells (PEC) and vascular endothelial cells) and the associated immune response during COVID-19. Immunometabolism may serve as a cell-specific therapeutic approach to target COVID-19.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
44
|
Macrophage Stimulated by Low Ambient Temperature Hasten Tumor Growth via Glutamine Production. Biomedicines 2020; 8:biomedicines8100381. [PMID: 32993179 PMCID: PMC7600495 DOI: 10.3390/biomedicines8100381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/08/2020] [Accepted: 09/24/2020] [Indexed: 12/25/2022] Open
Abstract
Ambient temperature can regulate the immune response and affect tumor growth. Although thermoneutral caging reduces tumor growth via immune activation, little attention has been paid to the tumorigenic effect of low temperature. In the present study, tumor growth was higher at low ambient temperature (4 °C for 8 h/d) than at the standard housing temperature (22 °C) in allograft models. Low temperature-stimulated tumor growth in mice was reduced by monocyte depletion using clodronate liposomes. Proliferation was considerably greater in cancer cells treated with 33 °C-cultured RAW264.7 cell-conditioned media (33CM) than in cells treated with 37 °C-cultured RAW264.7 cell-conditioned media (37CM). Additionally, glutamine levels were markedly higher in 33CM-treated cells than in 37CM-treated cells. We further confirmed that the addition of glutamine into 37CM enhanced its effects on cancer cell proliferation and glutamine uptake inhibition ameliorated the accelerated proliferation induced by 33CM. Consistently, the inhibition of glutamine uptake in the allograft model exposed to low temperature, effectively reduced tumor volume and weight. Collectively, these data suggest that the secretion and utilization of glutamine by macrophages and cancer cells, respectively, are key regulators of low temperature-enhanced cancer progression in the tumor microenvironment.
Collapse
|
45
|
Goll JB, Li S, Edwards JL, Bosinger SE, Jensen TL, Wang Y, Hooper WF, Gelber CE, Sanders KL, Anderson EJ, Rouphael N, Natrajan MS, Johnson RA, Sanz P, Hoft D, Mulligan MJ. Transcriptomic and Metabolic Responses to a Live-Attenuated Francisella tularensis Vaccine. Vaccines (Basel) 2020; 8:vaccines8030412. [PMID: 32722194 PMCID: PMC7563297 DOI: 10.3390/vaccines8030412] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/29/2020] [Accepted: 06/14/2020] [Indexed: 12/15/2022] Open
Abstract
The immune response to live-attenuated Francisella tularensis vaccine and its host evasion mechanisms are incompletely understood. Using RNA-Seq and LC–MS on samples collected pre-vaccination and at days 1, 2, 7, and 14 post-vaccination, we identified differentially expressed genes in PBMCs, metabolites in serum, enriched pathways, and metabolites that correlated with T cell and B cell responses, or gene expression modules. While an early activation of interferon α/β signaling was observed, several innate immune signaling pathways including TLR, TNF, NF-κB, and NOD-like receptor signaling and key inflammatory cytokines such as Il-1α, Il-1β, and TNF typically activated following infection were suppressed. The NF-κB pathway was the most impacted and the likely route of attack. Plasma cells, immunoglobulin, and B cell signatures were evident by day 7. MHC I antigen presentation was more actively up-regulated first followed by MHC II which coincided with the emergence of humoral immune signatures. Metabolomics analysis showed that glycolysis and TCA cycle-related metabolites were perturbed including a decline in pyruvate. Correlation networks that provide hypotheses on the interplay between changes in innate immune, T cell, and B cell gene expression signatures and metabolites are provided. Results demonstrate the utility of transcriptomics and metabolomics for better understanding molecular mechanisms of vaccine response and potential host–pathogen interactions.
Collapse
Affiliation(s)
- Johannes B. Goll
- The Emmes Company, Rockville, MD 20850, USA; (J.B.G.); (T.L.J.); (W.F.H.); (C.E.G.)
| | - Shuzhao Li
- Departments of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.L.); (Y.W.)
| | - James L. Edwards
- Department of Chemistry, Saint Louis University, St Louis, MO 63103, USA; (J.L.E.); (K.L.S.)
| | - Steven E. Bosinger
- Yerkes National Primate Research Center, Secret Path, Atlanta, GA 30329, USA;
- Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA; (N.R.); (M.S.N.)
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Decatur, GA 30030, USA
| | - Travis L. Jensen
- The Emmes Company, Rockville, MD 20850, USA; (J.B.G.); (T.L.J.); (W.F.H.); (C.E.G.)
| | - Yating Wang
- Departments of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.L.); (Y.W.)
| | - William F. Hooper
- The Emmes Company, Rockville, MD 20850, USA; (J.B.G.); (T.L.J.); (W.F.H.); (C.E.G.)
| | - Casey E. Gelber
- The Emmes Company, Rockville, MD 20850, USA; (J.B.G.); (T.L.J.); (W.F.H.); (C.E.G.)
| | - Katherine L. Sanders
- Department of Chemistry, Saint Louis University, St Louis, MO 63103, USA; (J.L.E.); (K.L.S.)
| | - Evan J. Anderson
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Nadine Rouphael
- Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA; (N.R.); (M.S.N.)
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Muktha S. Natrajan
- Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA; (N.R.); (M.S.N.)
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Robert A. Johnson
- Biomedical Advanced Research and Development Authority, U. S. Department of Health and Human Services, Washington, DC 20201, USA;
| | - Patrick Sanz
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA;
| | - Daniel Hoft
- Division of Infectious Diseases, Allergy and Immunology, Saint Louis University Health Sciences Center, St. Louis, MO 63104, USA;
| | - Mark J. Mulligan
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, 30322, USA
- Division of Infectious Diseases and Immunology, Department of Medicine, and New York University (NYU) Langone Vaccine Center, NYU School of Medicine, New York, NY 10016, USA
- Correspondence: ; Tel.: +1-212-263-9410; Fax: +1-646-501-4645
| |
Collapse
|
46
|
Câmara DAD, Shibli JA, Müller EA, De-Sá-Junior PL, Porcacchia AS, Blay A, Lizier NF. Adipose Tissue-Derived Stem Cells: The Biologic Basis and Future Directions for Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E3210. [PMID: 32708508 PMCID: PMC7420246 DOI: 10.3390/ma13143210] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/15/2020] [Indexed: 01/12/2023]
Abstract
Mesenchymal stem cells (MSCs) have been isolated from a variety of tissues using different methods. Active research have confirmed that the most accessible site to collect them is the adipose tissue; which has a significantly higher concentration of MSCs. Moreover; harvesting from adipose tissue is less invasive; there are no ethical limitations and a lower risk of severe complications. These adipose-derived stem cells (ASCs) are also able to increase at higher rates and showing telomerase activity, which acts by maintaining the DNA stability during cell divisions. Adipose-derived stem cells secret molecules that show important function in other cells vitality and mechanisms associated with the immune system, central nervous system, the heart and several muscles. They release cytokines involved in pro/anti-inflammatory, angiogenic and hematopoietic processes. Adipose-derived stem cells also have immunosuppressive properties and have been reported to be "immune privileged" since they show negative or low expression of human leukocyte antigens. Translational medicine and basic research projects can take advantage of bioprinting. This technology allows precise control for both scaffolds and cells. The properties of cell adhesion, migration, maturation, proliferation, mimicry of cell microenvironment, and differentiation should be promoted by the printed biomaterial used in tissue engineering. Self-renewal and potency are presented by MSCs, which implies in an open-source for 3D bioprinting and regenerative medicine. Considering these features and necessities, ASCs can be applied in the designing of tissue engineering products. Understanding the heterogeneity of ASCs and optimizing their properties can contribute to making the best therapeutic use of these cells and opening new paths to make tissue engineering even more useful.
Collapse
Affiliation(s)
| | - Jamil Awad Shibli
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., Jundiaí 13212-213, Brazil;
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos 07040-170, Brazil;
| | - Eduardo Alexandre Müller
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos 07040-170, Brazil;
| | | | - Allan Saj Porcacchia
- Department of Psychobiology, Federal University of São Paulo, São Paulo 04021-001, Brazil;
| | - Alberto Blay
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., Jundiaí 13212-213, Brazil;
| | - Nelson Foresto Lizier
- Nicell-Pesquisa e Desenvolvimento Científico LTDA, São Paulo 04006-000, Brazil;
- Department of Psychobiology, Federal University of São Paulo, São Paulo 04021-001, Brazil;
| |
Collapse
|
47
|
OGT suppresses S6K1-mediated macrophage inflammation and metabolic disturbance. Proc Natl Acad Sci U S A 2020; 117:16616-16625. [PMID: 32601203 DOI: 10.1073/pnas.1916121117] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Enhanced inflammation is believed to contribute to overnutrition-induced metabolic disturbance. Nutrient flux has also been shown to be essential for immune cell activation. Here, we report an unexpected role of nutrient-sensing O-linked β-N-acetylglucosamine (O-GlcNAc) signaling in suppressing macrophage proinflammatory activation and preventing diet-induced metabolic dysfunction. Overnutrition stimulates an increase in O-GlcNAc signaling in macrophages. O-GlcNAc signaling is down-regulated during macrophage proinflammatory activation. Suppressing O-GlcNAc signaling by O-GlcNAc transferase (OGT) knockout enhances macrophage proinflammatory polarization, promotes adipose tissue inflammation and lipolysis, increases lipid accumulation in peripheral tissues, and exacerbates tissue-specific and whole-body insulin resistance in high-fat-diet-induced obese mice. OGT inhibits macrophage proinflammatory activation by catalyzing ribosomal protein S6 kinase beta-1 (S6K1) O-GlcNAcylation and suppressing S6K1 phosphorylation and mTORC1 signaling. These findings thus identify macrophage O-GlcNAc signaling as a homeostatic mechanism maintaining whole-body metabolism under overnutrition.
Collapse
|
48
|
Shao Y, Saredy J, Yang WY, Sun Y, Lu Y, Saaoud F, Drummer C, Johnson C, Xu K, Jiang X, Wang H, Yang X. Vascular Endothelial Cells and Innate Immunity. Arterioscler Thromb Vasc Biol 2020; 40:e138-e152. [PMID: 32459541 PMCID: PMC7263359 DOI: 10.1161/atvbaha.120.314330] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In addition to the roles of endothelial cells (ECs) in physiological processes, ECs actively participate in both innate and adaptive immune responses. We previously reported that, in comparison to macrophages, a prototypic innate immune cell type, ECs have many innate immune functions that macrophages carry out, including cytokine secretion, phagocytic function, antigen presentation, pathogen-associated molecular patterns-, and danger-associated molecular patterns-sensing, proinflammatory, immune-enhancing, anti-inflammatory, immunosuppression, migration, heterogeneity, and plasticity. In this highlight, we introduce recent advances published in both ATVB and many other journals: (1) several significant characters classify ECs as novel immune cells not only in infections and allograft transplantation but also in metabolic diseases; (2) several new receptor systems including conditional danger-associated molecular pattern receptors, nonpattern receptors, and homeostasis associated molecular patterns receptors contribute to innate immune functions of ECs; (3) immunometabolism and innate immune memory determine the innate immune functions of ECs; (4) a great induction of the immune checkpoint receptors in ECs during inflammations suggests the immune tolerogenic functions of ECs; and (5) association of immune checkpoint inhibitors with cardiovascular adverse events and cardio-oncology indicates the potential contributions of ECs as innate immune cells.
Collapse
Affiliation(s)
- Ying Shao
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Jason Saredy
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - William Y. Yang
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Yu Sun
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Yifan Lu
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Fatma Saaoud
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Charles Drummer
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Candice Johnson
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Keman Xu
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Xiaohua Jiang
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Hong Wang
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| | - Xiaofeng Yang
- Centers of Inflammation, Translational & Clinical Lung Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
- Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140
| |
Collapse
|
49
|
Review: Following the smoke signals: inflammatory signaling in metabolic homeostasis and homeorhesis in dairy cattle. Animal 2020; 14:s144-s154. [PMID: 32024563 DOI: 10.1017/s1751731119003203] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inflammatory cascades are a critical component of the immune response to infection or tissue damage, involving an array of signals, including water-soluble metabolites, lipid mediators and several classes of proteins. Early investigation of these signaling pathways focused largely on immune cells and acute disease models. However, more recent findings have highlighted critical roles of both immune cells and inflammatory mediators on tissue remodeling and metabolic homeostasis in healthy animals. In dairy cattle, inflammatory signals in various tissues and in circulation change rapidly and dramatically, starting just prior to and at the onset of lactation. Furthermore, several observations in healthy cows point to homeostatic control of inflammatory tone, which we define as a regulatory process to balance immune tolerance with activation to keep downstream effects under control. Recent evidence suggests that peripartum inflammatory changes influence whole-body nutrient flux of dairy cows over the course of days and months. Inflammatory mediators can suppress appetite, even at levels that do not induce acute responses (e.g. fever), thereby decreasing nutrient availability. On the other hand, inhibition of inflammatory signaling with non-steroidal anti-inflammatory drug (NSAID) treatment suppresses hepatic gluconeogenesis, leading to hypoglycemia in some cases. Over the long term, though, peripartum NSAID treatment substantially increases peak and whole-lactation milk synthesis by multiparous cows. Inflammatory regulation of nutrient flux may provide a homeorhetic mechanism to aid cows in adapting to rapid changes in metabolic demand at the onset of lactation, but excessive systemic inflammation has negative effects on metabolic homeostasis through inhibition of appetite and promotion of immune cell activity. Thus, in this review, we provide perspectives on the overlapping regulation of immune responses and metabolism by inflammatory mediators, which may provide a mechanistic underpinning for links between infectious and metabolic diseases in transition dairy cows. Moreover, we point to novel approaches to the management of this challenging phase of the production cycle.
Collapse
|
50
|
Tamura Y, Tomiya S, Takegaki J, Kouzaki K, Tsutaki A, Nakazato K. Apple polyphenols induce browning of white adipose tissue. J Nutr Biochem 2020; 77:108299. [DOI: 10.1016/j.jnutbio.2019.108299] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/09/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
|