1
|
Xu R, Vujić N, Bianco V, Reinisch I, Kratky D, Krstic J, Prokesch A. Lipid-associated macrophages between aggravation and alleviation of metabolic diseases. Trends Endocrinol Metab 2024; 35:981-995. [PMID: 38705759 DOI: 10.1016/j.tem.2024.04.009] [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: 01/20/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024]
Abstract
Lipid-associated macrophages (LAMs) are phagocytic cells with lipid-handling capacity identified in various metabolic derangements. During disease development, they locate to atherosclerotic plaques, adipose tissue (AT) of individuals with obesity, liver lesions in steatosis and steatohepatitis, and the intestinal lamina propria. LAMs can also emerge in the metabolically demanding microenvironment of certain tumors. In this review, we discuss major questions regarding LAM recruitment, differentiation, and self-renewal, and, ultimately, their acute and chronic functional impact on the development of metabolic diseases. Further studies need to clarify whether and under which circumstances LAMs drive disease progression or resolution and how their phenotype can be modulated to ameliorate metabolic disorders.
Collapse
Affiliation(s)
- Ruonan Xu
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Valentina Bianco
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Isabel Reinisch
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Jelena Krstic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
2
|
Douglas A, Stevens B, Rendas M, Kane H, Lynch E, Kunkemoeller B, Wessendorf-Rodriguez K, Day EA, Sutton C, Brennan M, O'Brien K, Kohlgruber AC, Prendeville H, Garza AE, O'Neill LAJ, Mills KHG, Metallo CM, Veiga-Fernandes H, Lynch L. Rhythmic IL-17 production by γδ T cells maintains adipose de novo lipogenesis. Nature 2024:10.1038/s41586-024-08131-3. [PMID: 39478228 DOI: 10.1038/s41586-024-08131-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/27/2024] [Indexed: 11/06/2024]
Abstract
The circadian rhythm of the immune system helps to protect against pathogens1-3; however, the role of circadian rhythms in immune homeostasis is less well understood. Innate T cells are tissue-resident lymphocytes with key roles in tissue homeostasis4-7. Here we use single-cell RNA sequencing, a molecular-clock reporter and genetic manipulations to show that innate IL-17-producing T cells-including γδ T cells, invariant natural killer T cells and mucosal-associated invariant T cells-are enriched for molecular-clock genes compared with their IFNγ-producing counterparts. We reveal that IL-17-producing γδ (γδ17) T cells, in particular, rely on the molecular clock to maintain adipose tissue homeostasis, and exhibit a robust circadian rhythm for RORγt and IL-17A across adipose depots, which peaks at night. In mice, loss of the molecular clock in the CD45 compartment (Bmal1∆Vav1) affects the production of IL-17 by adipose γδ17 T cells, but not cytokine production by αβ or IFNγ-producing γδ (γδIFNγ) T cells. Circadian IL-17 is essential for de novo lipogenesis in adipose tissue, and mice with an adipocyte-specific deficiency in IL-17 receptor C (IL-17RC) have defects in de novo lipogenesis. Whole-body metabolic analysis in vivo shows that Il17a-/-Il17f-/- mice (which lack expression of IL-17A and IL-17F) have defects in their circadian rhythm for de novo lipogenesis, which results in disruptions to their whole-body metabolic rhythm and core-body-temperature rhythm. This study identifies a crucial role for IL-17 in whole-body metabolic homeostasis and shows that de novo lipogenesis is a major target of IL-17.
Collapse
Affiliation(s)
- Aaron Douglas
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Brenneth Stevens
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Miguel Rendas
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Harry Kane
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Evan Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Emily A Day
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Caroline Sutton
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Martin Brennan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Katie O'Brien
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Hannah Prendeville
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Amanda E Garza
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Christian M Metallo
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland.
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Molecular Biology, Princeton University, Princeton, NJ, USA.
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
3
|
Binvignat M, Sellam J, Berenbaum F, Felson DT. The role of obesity and adipose tissue dysfunction in osteoarthritis pain. Nat Rev Rheumatol 2024; 20:565-584. [PMID: 39112603 DOI: 10.1038/s41584-024-01143-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 08/29/2024]
Abstract
Obesity has a pivotal and multifaceted role in pain associated with osteoarthritis (OA), extending beyond the mechanistic influence of BMI. It exerts its effects both directly and indirectly through various modifiable risk factors associated with OA-related pain. Adipose tissue dysfunction is highly involved in OA-related pain through local and systemic inflammation, immune dysfunction, and the production of pro-inflammatory cytokines and adipokines. Adipose tissue dysfunction is intricately connected with metabolic syndrome, which independently exerts specific effects on OA-related pain, distinct from its association with BMI. The interplay among obesity, adipose tissue dysfunction and metabolic syndrome influences OA-related pain through diverse pain mechanisms, including nociceptive pain, peripheral sensitization and central sensitization. These complex interactions contribute to the heightened pain experience observed in individuals with OA and obesity. In addition, pain management strategies are less efficient in individuals with obesity. Importantly, therapeutic interventions targeting obesity and metabolic syndrome hold promise in managing OA-related pain. A deeper understanding of the intricate relationship between obesity, metabolic syndrome and OA-related pain is crucial and could have important implications for improving pain management and developing innovative therapeutic options in OA.
Collapse
Affiliation(s)
- Marie Binvignat
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Sorbonne University, INSERM UMRS_959, I3 Lab Immunology Immunopathology Immunotherapy, Paris, France
| | - Jérémie Sellam
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France.
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.
| | - Francis Berenbaum
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - David T Felson
- Boston University School of Medicine, Department of Medicine, Section of Rheumatology, Boston, MA, USA
| |
Collapse
|
4
|
Weijie Z, Meng Z, Chunxiao W, Lingjie M, Anguo Z, Yan Z, Xinran C, Yanjiao X, Li S. Obesity-induced chronic low-grade inflammation in adipose tissue: A pathway to Alzheimer's disease. Ageing Res Rev 2024; 99:102402. [PMID: 38977081 DOI: 10.1016/j.arr.2024.102402] [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: 03/11/2024] [Revised: 06/19/2024] [Accepted: 06/30/2024] [Indexed: 07/10/2024]
Abstract
Alzheimer's disease (AD) is a leading cause of cognitive impairment worldwide. Overweight and obesity are strongly associated with comorbidities, such as hypertension, diabetes, and insulin resistance (IR), which contribute substantially to the development of AD and subsequent morbidity and mortality. Adipose tissue (AT) is a highly dynamic organ composed of a diverse array of cell types, which can be classified based on their anatomic localization or cellular composition. The expansion and remodeling of AT in the context of obesity involves immunometabolic and functional shifts steered by the intertwined actions of multiple immune cells and cytokine signaling within AT, which contribute to the development of metabolic disorders, IR, and systemic markers of chronic low-grade inflammation. Chronic low-grade inflammation, a prolonged, low-dose stimulation by specific immunogens that can progress from localized sites and affect multiple organs throughout the body, leads to neurodystrophy, increased apoptosis, and disruption of homeostasis, manifesting as brain atrophy and AD-related pathology. In this review, we sought to elucidate the mechanisms by which AT contributes to the onset and progression of AD in obesity through the mediation of chronic low-grade inflammation, particularly focusing on the roles of adipokines and AT-resident immune cells.
Collapse
Affiliation(s)
- Zhai Weijie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zhao Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Wei Chunxiao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Meng Lingjie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zhao Anguo
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000 China
| | - Zhang Yan
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Cui Xinran
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xu Yanjiao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Sun Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
5
|
Savulescu-Fiedler I, Mihalcea R, Dragosloveanu S, Scheau C, Baz RO, Caruntu A, Scheau AE, Caruntu C, Benea SN. The Interplay between Obesity and Inflammation. Life (Basel) 2024; 14:856. [PMID: 39063610 PMCID: PMC11277997 DOI: 10.3390/life14070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is an important condition affecting the quality of life of numerous patients and increasing their associated risk for multiple diseases, including tumors and immune-mediated disorders. Inflammation appears to play a major role in the development of obesity and represents a central point for the activity of cellular and humoral components in the adipose tissue. Macrophages play a key role as the main cellular component of the adipose tissue regulating the chronic inflammation and modulating the secretion and differentiation of various pro- and anti-inflammatory cytokines. Inflammation also involves a series of signaling pathways that might represent the focus for new therapies and interventions. Weight loss is essential in decreasing cardiometabolic risks and the degree of associated inflammation; however, the latter can persist for long after the excess weight is lost, and can involve changes in macrophage phenotypes that can ensure the metabolic adjustment. A clear understanding of the pathophysiological processes in the adipose tissue and the interplay between obesity and chronic inflammation can lead to a better understanding of the development of comorbidities and may ensure future targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Razvan Mihalcea
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- “Prof. Dr. Matei Balș” National Institute for Infectious Diseases, 021105 Bucharest, Romania
| |
Collapse
|
6
|
Henry RJ, Barrett JP, Vaida M, Khan NZ, Makarevich O, Ritzel RM, Faden AI, Stoica BA. Interaction of high-fat diet and brain trauma alters adipose tissue macrophages and brain microglia associated with exacerbated cognitive dysfunction. J Neuroinflammation 2024; 21:113. [PMID: 38685031 PMCID: PMC11058055 DOI: 10.1186/s12974-024-03107-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 04/22/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity increases the morbidity and mortality of traumatic brain injury (TBI). Detailed analyses of transcriptomic changes in the brain and adipose tissue were performed to elucidate the interactive effects between high-fat diet-induced obesity (DIO) and TBI. Adult male mice were fed a high-fat diet (HFD) for 12 weeks prior to experimental TBI and continuing after injury. High-throughput transcriptomic analysis using Nanostring panels of the total visceral adipose tissue (VAT) and cellular components in the brain, followed by unsupervised clustering, principal component analysis, and IPA pathway analysis were used to determine shifts in gene expression patterns and molecular pathway activity. Cellular populations in the cortex and hippocampus, as well as in VAT, during the chronic phase after combined TBI-HFD showed amplification of central and peripheral microglia/macrophage responses, including superadditive changes in selected gene expression signatures and pathways. Furthermore, combined TBI and HFD caused additive dysfunction in Y-Maze, Novel Object Recognition (NOR), and Morris water maze (MWM) cognitive function tests. These novel data suggest that HFD-induced obesity and TBI can independently prime and support the development of altered states in brain microglia and VAT, including the disease-associated microglia/macrophage (DAM) phenotype observed in neurodegenerative disorders. The interaction between HFD and TBI promotes a shift toward chronic reactive microglia/macrophage transcriptomic signatures and associated pro-inflammatory disease-altered states that may, in part, underlie the exacerbation of cognitive deficits. Thus, targeting of HFD-induced reactive cellular phenotypes, including in peripheral adipose tissue immune cell populations, may serve to reduce microglial maladaptive states after TBI, attenuating post-traumatic neurodegeneration and neurological dysfunction.
Collapse
Affiliation(s)
- Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland.
| | - James P Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria Vaida
- Harrisburg University of Science and Technology, 326 Market St, Harrisburg, PA, USA
| | - Niaz Z Khan
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Oleg Makarevich
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
- VA Maryland Health Care System, Baltimore VA Medical Center, Baltimore, MD, 21201, USA
| |
Collapse
|
7
|
Oliveira BM, Sidónio B, Correia A, Pinto A, Azevedo MM, Sampaio P, Ferreira PG, Vilanova M, Teixeira L. Cytokine production by bovine adipose tissue stromal vascular fraction cells upon Neospora caninum stimulation. Sci Rep 2024; 14:8444. [PMID: 38600105 PMCID: PMC11006870 DOI: 10.1038/s41598-024-58885-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
In bovines few studies addressed the contribution of adipose tissue to the host immune response to infection. Here we evaluated the in vitro response of bovine adipose tissue stromal vascular fraction (SVF) cells to the protozoan parasite Neospora caninum, using live and freeze-killed tachyzoites. Live N. caninum induced the production of IL-6, IL-1β and IL-10 by SVF cells isolated from subcutaneous adipose tissue (SAT), while in mesenteric adipose tissue (MAT) SVF cell cultures only IL-1β and IL-10 production was increased, showing slight distinct responses between adipose tissue depots. Whereas a clear IL-8 increase was detected in peripheral blood leucocytes (PBL) culture supernatants in response to live N. caninum, no such increase was observed in SAT or MAT SVF cell cultures. Nevertheless, in response to LPS, increased IL-8 levels were detected in all cell cultures. IL-10 levels were always increased in response to stimulation (live, freeze-killed N. caninum and LPS). Overall, our results show that bovine adipose tissue SVF cells produce cytokines in response to N. caninum and can therefore be putative contributors to the host immune response against this parasite.
Collapse
Affiliation(s)
- Bárbara M Oliveira
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Beatriz Sidónio
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Alexandra Correia
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
| | - Ana Pinto
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal
| | - Maria M Azevedo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Paula Sampaio
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Paula G Ferreira
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal
| | - Manuel Vilanova
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal
| | - Luzia Teixeira
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313, Porto, Portugal.
- ITR-Laboratory for Integrative and Translational Research in Population Health, 4050-290, Porto, Portugal.
| |
Collapse
|
8
|
Dang Q, Li B, Jin B, Ye Z, Lou X, Wang T, Wang Y, Pan X, Hu Q, Li Z, Ji S, Zhou C, Yu X, Qin Y, Xu X. Cancer immunometabolism: advent, challenges, and perspective. Mol Cancer 2024; 23:72. [PMID: 38581001 PMCID: PMC10996263 DOI: 10.1186/s12943-024-01981-5] [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: 12/26/2023] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
For decades, great strides have been made in the field of immunometabolism. A plethora of evidence ranging from basic mechanisms to clinical transformation has gradually embarked on immunometabolism to the center stage of innate and adaptive immunomodulation. Given this, we focus on changes in immunometabolism, a converging series of biochemical events that alters immune cell function, propose the immune roles played by diversified metabolic derivatives and enzymes, emphasize the key metabolism-related checkpoints in distinct immune cell types, and discuss the ongoing and upcoming realities of clinical treatment. It is expected that future research will reduce the current limitations of immunotherapy and provide a positive hand in immune responses to exert a broader therapeutic role.
Collapse
Affiliation(s)
- Qin Dang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Borui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bing Jin
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Ting Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xuan Pan
- Department of Hepatobiliary Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chenjie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Zhao JY, Zhou LJ, Ma KL, Hao R, Li M. MHO or MUO? White adipose tissue remodeling. Obes Rev 2024; 25:e13691. [PMID: 38186200 DOI: 10.1111/obr.13691] [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: 05/05/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 01/09/2024]
Abstract
In this review, we delve into the intricate relationship between white adipose tissue (WAT) remodeling and metabolic aspects in obesity, with a specific focus on individuals with metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO). WAT is a highly heterogeneous, plastic, and dynamically secreting endocrine and immune organ. WAT remodeling plays a crucial role in metabolic health, involving expansion mode, microenvironment, phenotype, and distribution. In individuals with MHO, WAT remodeling is beneficial, reducing ectopic fat deposition and insulin resistance (IR) through mechanisms like increased adipocyte hyperplasia, anti-inflammatory microenvironment, appropriate extracellular matrix (ECM) remodeling, appropriate vascularization, enhanced WAT browning, and subcutaneous adipose tissue (SWAT) deposition. Conversely, for those with MUO, WAT remodeling leads to ectopic fat deposition and IR, causing metabolic dysregulation. This process involves adipocyte hypertrophy, disrupted vascularization, heightened pro-inflammatory microenvironment, enhanced brown adipose tissue (BAT) whitening, and accumulation of visceral adipose tissue (VWAT) deposition. The review underscores the pivotal importance of intervening in WAT remodeling to hinder the transition from MHO to MUO. This insight is valuable for tailoring personalized and effective management strategies for patients with obesity in clinical practice.
Collapse
Affiliation(s)
- Jing Yi Zhao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Juan Zhou
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kai Le Ma
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Hao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Li
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Lu Z, Ding L, Tian X, Wang Q. Single cell RNA-sequencing data generated from mouse adipose tissue during the development of obesity. Data Brief 2024; 53:110119. [PMID: 38348326 PMCID: PMC10859251 DOI: 10.1016/j.dib.2024.110119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/04/2024] [Accepted: 01/23/2024] [Indexed: 02/15/2024] Open
Abstract
In recent years, the number of obesity has increased rapidly around the world, and it has become a major public health problem endangering global health [1]. Obesity is caused by excessive calorie intake over a long period of time, and high-fat diet (HFD) is one of the important predisposing factors [2], [3], [4]. Adipose tissue (AT) is an important immune and endocrine organ in the body, and plays an important role in the body [5]. Obesity leads to AT dysfunction, AT dilation and cell hypertrophy. Dysfunctional fat cells are the main source of pro-inflammatory cytokines, which aggravate low-grade systemic inflammation and further promote the development of obesity-related diseases [6], [7], [8]. However, whether AT releases pro-inflammatory cytokines in the early stages of obesity development remains unknown. The AT microenvironment is composed of a variety of cells, including fat cells, immune cells, fibroblasts, and endothelial cells. The immune microenvironment (TIME) and its metabolic imbalance can lead to the secretion or regulation of related hormones, which causes inflammation AT [9]. TIME is very important for maintaining AT homeostasis, which is crucial for the occurrence of obesity [10,11]. This data use single-cell RNA sequencing (sNuc-Seq) to analyze the characteristics of TIME changes in the mouse epididymal adipose tissue during the development of obesity, and the changes of cell types and genes in the tissue.
Collapse
Affiliation(s)
- Zhimin Lu
- College of Sport and Health, Shandong Sport University, Jinan, Shandong 250102, China
| | - Ling Ding
- College of Sport and Health, Shandong Sport University, Jinan, Shandong 250102, China
| | | | | |
Collapse
|
11
|
Sun JY, Su Z, Yang J, Sun W, Kong X. The potential mechanisms underlying the modulating effect of perirenal adipose tissue on hypertension: Physical compression, paracrine, and neurogenic regulation. Life Sci 2024; 342:122511. [PMID: 38387699 DOI: 10.1016/j.lfs.2024.122511] [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: 12/11/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 02/24/2024]
Abstract
Hypertension, a prevalent global cardiovascular disease, affects approximately 45.4 % of adults worldwide. Despite advances in therapy, hypertension continues to pose a significant health risk due to inadequate management. It has been established that excessive adiposity contributes majorly to hypertension, accounting for 65 to 75 % of primary cases. Fat depots can be categorised into subcutaneous and visceral adipose tissue based on anatomical and physiological characteristics. The metabolic impact and the risk of hypertension are determined more significantly by visceral fat. Perirenal adipose tissue (PRAT), a viscera enveloping the kidney, is known for its superior vascularisation and abundant innervation. Although traditionally deemed as a mechanical support tissue, recent studies have indicated its contributing potential to hypertension. Hypertensive patients tend to have increased PRAT thickness compared to those without, and there is a positive correlation between PRAT thickness and elevated systolic blood pressure. This review encapsulates the anatomical characteristics and biogenesis of PRAT. We provide an overview of the potential mechanisms where PRAT may modulate blood pressure, including physical compression, paracrine effects, and neurogenic regulation. PRAT has become a promising target for hypertension management, and continuous effort is required to further explore the underlying mechanisms.
Collapse
Affiliation(s)
- Jin-Yu Sun
- Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China; Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Zhenyang Su
- Medical School of Southeast University, Nanjing 21000, China
| | - Jiaming Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Wei Sun
- Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China; Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China.
| | - Xiangqing Kong
- Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China; Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China.
| |
Collapse
|
12
|
Teixeira L, Whitmire JK, Bourgeois C. Editorial: The role of adipose tissue and resident immune cells in infections. Front Immunol 2024; 15:1360262. [PMID: 38464526 PMCID: PMC10920208 DOI: 10.3389/fimmu.2024.1360262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/13/2024] [Indexed: 03/12/2024] Open
Affiliation(s)
- Luzia Teixeira
- UMIB-Unidade Multidisciplinar de Investigação Biomédica, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Jason K. Whitmire
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Christine Bourgeois
- Université Paris-Saclay, French National Institute of Health and Medical Research (INSERM), French Alternative Energies and Atomic Energy Commission (CEA), Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), UMR1184, Le Kremlin Bicêtre, France
| |
Collapse
|
13
|
Reinisch I, Michenthaler H, Sulaj A, Moyschewitz E, Krstic J, Galhuber M, Xu R, Riahi Z, Wang T, Vujic N, Amor M, Zenezini Chiozzi R, Wabitsch M, Kolb D, Georgiadi A, Glawitsch L, Heitzer E, Schulz TJ, Schupp M, Sun W, Dong H, Ghosh A, Hoffmann A, Kratky D, Hinte LC, von Meyenn F, Heck AJR, Blüher M, Herzig S, Wolfrum C, Prokesch A. Adipocyte p53 coordinates the response to intermittent fasting by regulating adipose tissue immune cell landscape. Nat Commun 2024; 15:1391. [PMID: 38360943 PMCID: PMC10869344 DOI: 10.1038/s41467-024-45724-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/02/2024] [Indexed: 02/17/2024] Open
Abstract
In obesity, sustained adipose tissue (AT) inflammation constitutes a cellular memory that limits the effectiveness of weight loss interventions. Yet, the impact of fasting regimens on the regulation of AT immune infiltration is still elusive. Here we show that intermittent fasting (IF) exacerbates the lipid-associated macrophage (LAM) inflammatory phenotype of visceral AT in obese mice. Importantly, this increase in LAM abundance is strongly p53 dependent and partly mediated by p53-driven adipocyte apoptosis. Adipocyte-specific deletion of p53 prevents LAM accumulation during IF, increases the catabolic state of adipocytes, and enhances systemic metabolic flexibility and insulin sensitivity. Finally, in cohorts of obese/diabetic patients, we describe a p53 polymorphism that links to efficacy of a fasting-mimicking diet and that the expression of p53 and TREM2 in AT negatively correlates with maintaining weight loss after bariatric surgery. Overall, our results demonstrate that p53 signalling in adipocytes dictates LAM accumulation in AT under IF and modulates fasting effectiveness in mice and humans.
Collapse
Affiliation(s)
- Isabel Reinisch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Helene Michenthaler
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Alba Sulaj
- Institute for Diabetes and Cancer, Helmholtz Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Endocrinology, Diabetology, Metabolism and Clinical Chemistry (Internal Medicine 1), Heidelberg University Hospital, Heidelberg, Germany
| | - Elisabeth Moyschewitz
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Jelena Krstic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Markus Galhuber
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Ruonan Xu
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Zina Riahi
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Tongtong Wang
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Nemanja Vujic
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Melina Amor
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Dagmar Kolb
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
- Core Facility Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Anastasia Georgiadi
- Institute for Diabetes and Cancer, Helmholtz Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lisa Glawitsch
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Michael Schupp
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wenfei Sun
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Hua Dong
- Stem Cell Biology and Regenerative Medicine Institute, University of Stanford, Stanford, CA, USA
| | - Adhideb Ghosh
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
- Functional Genomics Center Zurich, Eidgenössische Technische Hochschule Zürich (ETH), Zurich, Switzerland
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Laura C Hinte
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Endocrinology, Diabetology, Metabolism and Clinical Chemistry (Internal Medicine 1), Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Wolfrum
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
14
|
Pries R, Kosyna FK, Depping R, Plötze-Martin K, Lange C, Meyhöfer S, Meyhöfer SM, Marquardt JU, Bruchhage KL, Steffen A. Distinguishing the impact of distinct obstructive sleep apnea syndrome (OSAS) and obesity related factors on human monocyte subsets. Sci Rep 2024; 14:340. [PMID: 38172514 PMCID: PMC10764945 DOI: 10.1038/s41598-023-49921-5] [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: 04/18/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) and obesity go hand in hand in the majority of patients and both are associated with a systemic inflammation, immune disturbance and comorbidities such as cardiovascular disease. However, the unambiguous impact of OSAS and obesity on the individual inflammatory microenvironment and the immunological consequences of human monocytes has not been distinguished yet. Therefore, aim of this study was to investigate the impact of OSAS and obesity related factors on the inflammatory microenvironment by performing flow cytometric whole blood measurements of CD14/CD16 monocyte subsets in normal weight OSAS patients, patients with obesity but without OSAS, and patients with OSAS and obesity, compared to healthy donors. Moreover, explicitly OSAS and obesity related plasma levels of inflammatory mediators adiponectin, leptin, lipocalin and metalloproteinase-9 were determined and the influence of different OSAS and obesity related factors on cytokine secretion and expression of different adhesion molecules by THP-1 monocytes was analysed. Our data revealed a significant redistribution of circulating classical and intermediate monocytes in all three patient cohorts, but differential effects in terms of monocytic adhesion molecules CD11a, CD11b, CD11c, CX3CR1, CD29, CD49d, and plasma cytokine levels. These data were reflected by differential effects of OSAS and obesity related factors leptin, TNFα and hypoxia on THP-1 cytokine secretion patterns and expression of adhesion molecules CD11b and CD49d. In summary, our data revealed differential effects of OSAS and obesity, which underlines the need for a customized therapeutic regimen with respect to the individual weighting of these overlapping diseases.
Collapse
Affiliation(s)
- Ralph Pries
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| | - Friederike Katharina Kosyna
- Institute of Physiology, Working Group Hypoxia, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Reinhard Depping
- Institute of Physiology, Working Group Hypoxia, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Kirstin Plötze-Martin
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Christian Lange
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Svenja Meyhöfer
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Lübeck, Germany
- Institute for Endocrinology & Diabetes, University Hospital of Schleswig-Holstein, Lübeck, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sebastian M Meyhöfer
- Institute for Endocrinology & Diabetes, University Hospital of Schleswig-Holstein, Lübeck, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jens U Marquardt
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Armin Steffen
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| |
Collapse
|
15
|
Fu B, Wei L, Wang C, Xiong B, Bo J, Jiang X, Zhang Y, Jia H, Dong J. Nomograms combining computed tomography-based body composition changes with clinical prognostic factors to predict survival in locally advanced cervical cancer patients. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2024; 32:427-441. [PMID: 38189735 DOI: 10.3233/xst-230212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
OBJECTIVE To explore the value of body composition changes (BCC) measured by quantitative computed tomography (QCT) for evaluating the survival of patients with locally advanced cervical cancer (LACC) underwent concurrent chemoradiotherapy (CCRT), nomograms combined BCC with clinical prognostic factors (CPF) were constructed to predict overall survival (OS) and progression-free survival (PFS). METHODS Eighty-eight patients with LACC were retrospectively selected. All patients underwent QCT scans before and after CCRT, bone mineral density (BMD), subcutaneous fat area (SFA), visceral fat area (VFA), total fat area (TFA), paravertebral muscle area (PMA) were measured from two sets of computed tomography (CT) images, and change rates of these were calculated. RESULTS Multivariate Cox regression analysis showed ΔBMD, ΔSFA, SCC-Ag, LNM were independent factors for OS (HR = 3.560, 5.870, 2.702, 2.499, respectively, all P < 0.05); ΔPMA, SCC-Ag, LNM were independent factors for PFS (HR = 2.915, 4.291, 2.902, respectively, all P < 0.05). Prognostic models of BCC combined with CPF had the highest predictive performance, and the area under the curve (AUC) for OS and PFS were 0.837, 0.846, respectively. The concordance index (C-index) of nomograms for OS and PFS were 0.834, 0.799, respectively. Calibration curves showed good agreement between the nomograms' predictive and actual OS and PFS, decision curve analysis (DCA) showed good clinical benefit of nomograms. CONCLUSION CT-based body composition changes and CPF (SCC-Ag, LNM) were associated with survival in patients with LACC. The prognostic nomograms combined BCC with CPF were able to predict the OS and PFS in patients with LACC reliably.
Collapse
Affiliation(s)
- Baoyue Fu
- Bengbu Medical College, Bengbu, Anhui, China
| | - Longyu Wei
- Bengbu Medical College, Bengbu, Anhui, China
| | - Chuanbin Wang
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | | | - Juan Bo
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | | | - Yu Zhang
- Bengbu Medical College, Bengbu, Anhui, China
| | - Haodong Jia
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Jiangning Dong
- Bengbu Medical College, Bengbu, Anhui, China
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
16
|
Meyhöfer S, Steffen A, Plötze-Martin K, Marquardt JU, Meyhöfer SM, Bruchhage KL, Pries R. Obesity-related Plasma CXCL10 Drives CX3CR1-dependent Monocytic Secretion of Macrophage Migration Inhibitory Factor. Immunohorizons 2024; 8:19-28. [PMID: 38175171 PMCID: PMC10835669 DOI: 10.4049/immunohorizons.2300114] [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: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Obesity is characterized by excessive body fat accumulation and comorbidities such as diabetes mellitus, cardiovascular disease, and obstructive sleep apnea syndrome (OSAS). Both obesity and OSAS are associated with immune disturbance, alterations of systemic inflammatory mediators, and immune cell recruitment to metabolic tissues. Chemokine CXCL10 is an important regulator of proinflammatory immune responses and is significantly increased in patients with severe obesity. This research project aims to investigate the impact of CXCL10 on human monocytes in patients with obesity. We studied the distribution of the CD14/CD16 monocyte subsets as well as their CX3CR1 expression patterns in whole-blood measurements from 92 patients with obesity and/or OSAS with regard to plasma CXCL10 values and individual clinical parameters. Furthermore, cytokine secretion by THP-1 monocytes in response to CXCL10 was analyzed. Data revealed significantly elevated plasma CXCL10 in patients with obesity with an additive effect of OSAS. CXCL10 was found to drive monocytic secretion of macrophage migration inhibitory factor via receptor protein CX3CR1, which significantly correlated with the individual body mass index. Our data show, for the first time, to our knowledge, that CX3CR1 is involved in alternative CXCL10 signaling in human monocytes in obesity-related inflammation. Obesity is a multifactorial disease, and further investigations regarding the complex interplay between obesity-related inflammatory mediators and systemic immune balances will help to better understand and improve the individual situation of our patients.
Collapse
Affiliation(s)
- Svenja Meyhöfer
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Luebeck, Germany
- Institute for Endocrinology & Diabetes, Department of Internal Medicine 1, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Armin Steffen
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Kirstin Plötze-Martin
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Jens-Uwe Marquardt
- Department of Medicine 1, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Sebastian M Meyhöfer
- Institute for Endocrinology & Diabetes, Department of Internal Medicine 1, University Hospital of Schleswig-Holstein, Luebeck, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| |
Collapse
|
17
|
Hodel K, Fonseca A, Barbosa I, Medina C, Alves B, Maciel C, Nascimento D, Oliveira-Junior G, Pedreira L, de Souza M, Godoy AL. Obesity and its Relationship with Covid-19: A Review of the Main Pharmaceutical Aspects. Curr Pharm Biotechnol 2024; 25:1651-1663. [PMID: 38258769 DOI: 10.2174/0113892010264503231108070917] [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: 06/15/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 01/24/2024]
Abstract
Important physiological changes are observed in patients with obesity, such as intestinal permeability, gastric emptying, cardiac output, and hepatic and renal function. These differences can determine variations in the pharmacokinetics of different drugs and can generate different concentrations at the site of action, which can lead to sub therapeutic or toxic concentrations. Understanding the physiological and immunological processes that lead to the clinical manifestations of COVID-19 is essential to correlate obesity as a risk factor for increasing the prevalence, severity, and lethality of the disease. Several drugs have been suggested to control COVID- 19 like Lopinavir, Ritonavir, Ribavirin, Sofosbuvir, Remdesivir, Oseltamivir, Oseltamivir phosphate, Oseltamivir carboxylate, Hydroxychloroquine, Chloroquine, Azithromycin, Teicoplanin, Tocilizumab, Anakinra, Methylprednisolone, Prednisolone, Ciclesonide and Ivermectin. Similarly, these differences between healthy people and obese people can be correlated to mechanical factors, such as insufficient doses of the vaccine for high body mass, impairing the absorption and distribution of the vaccine that will be lower than desired or can be linked to the inflammatory state in obese patients, which can influence the humoral immune response. Additionally, different aspects make the obese population more prone to persistent symptoms of the disease (long COVID), which makes understanding these mechanisms fundamental to addressing the implications of the disease. Thus, this review provides an overview of the relationship between COVID-19 and obesity, considering aspects related to pharmacokinetics, immunosuppression, immunization, and possible implications of long COVID in these individuals.
Collapse
Affiliation(s)
- Katharine Hodel
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Ananda Fonseca
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Islania Barbosa
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Caio Medina
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Brenda Alves
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Carine Maciel
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Daniel Nascimento
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Gessualdo Oliveira-Junior
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Lorena Pedreira
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Monielly de Souza
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Ana Leonor Godoy
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| |
Collapse
|
18
|
Ma S, Wang WX. Physiological trade-off of marine fish under Zn deficient and excess conditions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 901:166187. [PMID: 37586517 DOI: 10.1016/j.scitotenv.2023.166187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023]
Abstract
Fish can regulate their Zn body bioaccumulation, but the mechanisms and physiological responses at the organ level are still largely unknown. In the present study, we exposed the marine seabreams under different Zn levels (deficient, optimum and excess levels) over a period of 4 weeks and examined how fish maintained its regulation of bioaccumulation with associated physiological effects at the fish intestinal organ. Our results indicated that fish intestinal organs constantly controlled the Zip family to "rob" more Zn under Zn-deficiency (with a dietary level of 7.9 mg/kg), whereas restricted the Zn efflux to preserve the intestinal function. Under Zn-excess conditions (193.3 mg/kg), the fish intestine maintained a limited Zn homeostasis (37.8-44.6 μg/mg) by initially inhibiting the influx through the Zip family receptor, but later accelerating both influx and efflux of Zn. Based on the WGCNA method, Zn deficient dietary exposure first resulted in defense response with subsequent switching to antioxidant defense. Instead, excess Zn first triggered the immunological response, but then led to physiological toxicity (abnormal in lipid metabolism). Although Zn had multiple biological functions, it was preferentially involved in lipid metabolism under different dietary Zn doses. This study provided direct evidence for Zn regulation at the organ level and detoxification mechanisms against potential environmental toxicity in fish.
Collapse
Affiliation(s)
- Shuoli Ma
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China.
| |
Collapse
|
19
|
Ninla-aesong P, Puangsri P, Kietdumrongwong P, Jongkrijak H, Noipha K. Being overweight and obese increases suicide risk, the severity of depression, and the inflammatory response in adolescents with major depressive disorders. Front Immunol 2023; 14:1197775. [PMID: 38022570 PMCID: PMC10646409 DOI: 10.3389/fimmu.2023.1197775] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Background There is a bidirectional relationship between obesity and depression. We investigated whether the coexistence of obesity and depression increases the risk of having severe depression and a high suicide risk in adolescents with major depressive disorder (MDD). Additionally, we explored the potential mechanisms linking the coexistence of obesity and depression to worse outcomes in these patients. Methods The odds of high suicide risk and severe depression were compared among MDD patients based on different body mass index (BMI) groups. Complete blood count (CBC) parameters, inflammatory ratios (neutrophil-lymphocyte ratio [NLR], monocyte-lymphocyte ratio [MLR], and platelet-lymphocyte ratio [PLR]), and cytokine levels (IFN-γ, IL-1β, IL-6, IL-8, MCP-1, TNF-α, and TGF-β1) were evaluated across BMI groups. Additionally, Pearson correlation coefficients (r) were assessed to understand the relationships between the 8Q and 9Q scores, CBC parameters, inflammatory ratios, cytokine levels, and BMI. Results A total of 135 antidepressant-naive adolescents with MDD were included. Overweight and obese MDD patients had higher odds of having high suicide risk and severe depression than lean individuals. Furthermore, they exhibited significantly higher white blood cell (WBC), and neutrophil counts. The NLR tended to be higher in obese MDD patients than in leans. Overweight and obese MDD patients had elevated levels of interleukin (IL)-1β and IL-6 compared to lean individuals, while TGF-β1 levels appeared to decline as body weight increased. BMI showed weak positive correlations with 8Q score, WBC count, neutrophil count, monocyte count, platelet count, neutrophil percentage, and NLR, and a weak negative correlation with lymphocyte percentage. The 8Q score displayed weak positive correlations with BMI, neutrophil percentage, monocyte percentages, NLR, and MLR, and a weak negative correlation with lymphocyte percentage. Conclusion The findings suggest that coexistence of overweight or obesity with depression heightened inflammatory responses, leading to worse outcomes and increased suicide risk in adolescents MDD patients.
Collapse
Affiliation(s)
- Putrada Ninla-aesong
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, Thailand
- Excellence Center of Community Health Promotion, Walailak University, Nakhon Si Thammarat, Thailand
| | - Pavarud Puangsri
- Department of Medical Clinical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
| | | | - Haruthai Jongkrijak
- Walailak University Hospital, Walailak University, Nakhon Si Thammarat, Thailand
| | - Kusumarn Noipha
- Faculty of Health and Sports Science, Thaksin University, Paphayom, Phatthalung, Thailand
| |
Collapse
|
20
|
Tang C, Wang Y, Chen D, Zhang M, Xu J, Xu C, Liu J, Kan J, Jin C. Natural polysaccharides protect against diet-induced obesity by improving lipid metabolism and regulating the immune system. Food Res Int 2023; 172:113192. [PMID: 37689942 DOI: 10.1016/j.foodres.2023.113192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 09/11/2023]
Abstract
Unhealthy dietary patterns-induced obesity and obesity-related complications pose a great threat to human health all over the world. Accumulating evidence suggests that the pathophysiology of obesity and obesity-associated metabolic disorders is closely associated with dysregulation of lipid and energy metabolism, and metabolic inflammation. In this review, three potential anti-obesity mechanisms of natural polysaccharides are introduced. Firstly, natural polysaccharides protect against diet-induced obesity directly by improving lipid and cholesterol metabolism. Since the immunity also affects lipid and energy metabolism, natural polysaccharides improve lipid and energy metabolism by regulating host immunity. Moreover, diet-induced mitochondrial dysfunction, prolonged endoplasmic reticulum stress, defective autophagy and microbial dysbiosis can disrupt lipid and/or energy metabolism in a direct and/or inflammation-induced manner. Therefore, natural polysaccharides also improve lipid and energy metabolism and suppress inflammation by alleviating mitochondrial dysfunction and endoplasmic reticulum stress, promoting autophagy and regulating gut microbiota composition. Specifically, this review comprehensively summarizes underlying anti-obesity mechanisms of natural polysaccharides and provides a theoretical basis for the development of functional foods. For the first time, this review elucidates anti-obesity mechanisms of natural polysaccharides from the perspectives of their hypolipidemic, energy-regulating and immune-regulating mechanisms.
Collapse
Affiliation(s)
- Chao Tang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Yuxin Wang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Dan Chen
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Man Zhang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Jingguo Xu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Chen Xu
- Nanjing Key Laboratory of Quality and safety of agricultural product, Nanjing Xiaozhuang University, Nanjing 211171, China.
| | - Jun Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Juan Kan
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Changhai Jin
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| |
Collapse
|
21
|
Ishikawa H, Nagashima R, Kuno Y, Sasaki H, Kohda C, Iyoda M. Effects of NKT Cells on Metabolic Disorders Caused by High-Fat Diet Using CD1d-Knockout Mice. Diabetes Metab Syndr Obes 2023; 16:2855-2864. [PMID: 37744699 PMCID: PMC10517681 DOI: 10.2147/dmso.s428190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023] Open
Abstract
Purpose The purpose of this study was to investigate whether NKT cells play an important role in preventing or exacerbating diseases caused by high-fat diet (HFD) using CD1d-knockout (KO) mice which lack NKT cells. Methods Five-week-old male Balb/c (wild-type; WT) or CD1dKO mice were fed with control-diet (CTD) or HFD for 16 weeks. Results The present study revealed four main findings. First, CD1dKO mice were susceptible to obesity caused by HFD in comparison to WT mice. Second, clinical conditions of fatty liver caused by HFD were comparable between CD1dKO mice and WT mice. Third, HFD-fed WT mice showed high levels of serum biochemical markers, involved in lipid metabolisms, in comparison to WT mice fed a CTD. Notably, the serum concentrations of ALT, T-CHO, TG and HDL-C in CD1dKO mice fed a HFD were almost comparable to those of CD1dKO mice fed a CTD. Fourth, the expression of peroxisome proliferator-activated receptor (PPAR) γ, low-density lipoprotein receptor (LDLR), CD36 of epididymal adipose tissue enhanced and proprotein convertase subtilisin/kexin type (PCSK) 9 in serum decreased. Conclusion NKT cells were responsible for protection against HFD-induced obesity. However, CD1dKO mice were resistant to serum biochemical marker abnormalities after HFD feeding. One possible explanation is that the epididymal adipose tissue of CD1dKO mice could take up greater amounts of excess lipids in serum in comparison to WT mice.
Collapse
Affiliation(s)
- Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8666, Japan
| | - Hiraku Sasaki
- Department of Health Science, Faculty of Health and Sports Science, Juntendo University, Inzai, Chiba, 270-1695, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8666, Japan
| |
Collapse
|
22
|
Rohringer S, Grasl C, Ehrmann K, Hager P, Hahn C, Specht SJ, Walter I, Schneider KH, Zopf LM, Baudis S, Liska R, Schima H, Podesser BK, Bergmeister H. Biodegradable, Self-Reinforcing Vascular Grafts for In Situ Tissue Engineering Approaches. Adv Healthc Mater 2023; 12:e2300520. [PMID: 37173073 PMCID: PMC11468867 DOI: 10.1002/adhm.202300520] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/21/2023] [Indexed: 05/15/2023]
Abstract
Clinically available small-diameter synthetic vascular grafts (SDVGs) have unsatisfactory patency rates due to impaired graft healing. Therefore, autologous implants are still the gold standard for small vessel replacement. Bioresorbable SDVGs may be an alternative, but many polymers have inadequate biomechanical properties that lead to graft failure. To overcome these limitations, a new biodegradable SDVG is developed to ensure safe use until adequate new tissue is formed. SDVGs are electrospun using a polymer blend composed of thermoplastic polyurethane (TPU) and a new self-reinforcing TP(U-urea) (TPUU). Biocompatibility is tested in vitro by cell seeding and hemocompatibility tests. In vivo performance is evaluated in rats over a period for up to six months. Autologous rat aortic implants serve as a control group. Scanning electron microscopy, micro-computed tomography (µCT), histology, and gene expression analyses are applied. TPU/TPUU grafts show significant improvement of biomechanical properties after water incubation and exhibit excellent cyto- and hemocompatibility. All grafts remain patent, and biomechanical properties are sufficient despite wall thinning. No inflammation, aneurysms, intimal hyperplasia, or thrombus formation are observed. Evaluation of graft healing shows similar gene expression profiles of TPU/TPUU and autologous conduits. These new biodegradable, self-reinforcing SDVGs may be promising candidates for clinical use in the future.
Collapse
Affiliation(s)
- Sabrina Rohringer
- Center for Biomedical Research and Translational SurgeryMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
- Austrian Cluster for Tissue RegenerationDonaueschingenstraße 13Vienna1200Austria
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
| | - Christian Grasl
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
- Center for Medical Physics and Biomedical EngineeringMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
| | - Katharina Ehrmann
- Center for Biomedical Research and Translational SurgeryMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
- Austrian Cluster for Tissue RegenerationDonaueschingenstraße 13Vienna1200Austria
- Institute of Applied Synthetic ChemistryTechnical University of ViennaGetreidemarkt 9/163Vienna1060Austria
| | - Pia Hager
- Center for Biomedical Research and Translational SurgeryMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
| | - Clemens Hahn
- Center for Biomedical Research and Translational SurgeryMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
| | - Sophie J. Specht
- Center for Biomedical Research and Translational SurgeryMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
| | - Ingrid Walter
- Department of PathobiologyUniversity of Veterinary MedicineVeterinaerplatz 1Vienna1210Austria
| | - Karl H. Schneider
- Center for Biomedical Research and Translational SurgeryMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
- Austrian Cluster for Tissue RegenerationDonaueschingenstraße 13Vienna1200Austria
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
| | - Lydia M. Zopf
- Austrian Cluster for Tissue RegenerationDonaueschingenstraße 13Vienna1200Austria
- Ludwig Boltzmann Institute for TraumatologyDonaueschingenstraße 13Vienna1200Austria
| | - Stefan Baudis
- Austrian Cluster for Tissue RegenerationDonaueschingenstraße 13Vienna1200Austria
- Institute of Applied Synthetic ChemistryTechnical University of ViennaGetreidemarkt 9/163Vienna1060Austria
| | - Robert Liska
- Austrian Cluster for Tissue RegenerationDonaueschingenstraße 13Vienna1200Austria
- Institute of Applied Synthetic ChemistryTechnical University of ViennaGetreidemarkt 9/163Vienna1060Austria
| | - Heinrich Schima
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
- Center for Medical Physics and Biomedical EngineeringMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
| | - Bruno K. Podesser
- Center for Biomedical Research and Translational SurgeryMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
- Austrian Cluster for Tissue RegenerationDonaueschingenstraße 13Vienna1200Austria
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
| | - Helga Bergmeister
- Center for Biomedical Research and Translational SurgeryMedical University of ViennaWaehringer Gürtel 18‐20Vienna1090Austria
- Austrian Cluster for Tissue RegenerationDonaueschingenstraße 13Vienna1200Austria
- Ludwig Boltzmann Institute for Cardiovascular ResearchWaehringer Gürtel 18‐20Vienna1090Austria
| |
Collapse
|
23
|
Henry RJ, Barrett JP, Vaida M, Khan NZ, Makarevich O, Ritzel RM, Faden AI, Stoica BA. Interaction of high-fat diet and brain trauma alters adipose tissue macrophages and brain microglia associated with exacerbated cognitive dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.550986. [PMID: 37546932 PMCID: PMC10402152 DOI: 10.1101/2023.07.28.550986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Obesity increases the morbidity and mortality of traumatic brain injury (TBI). We performed a detailed analysis of transcriptomic changes in the brain and adipose tissue to examine the interactive effects between high-fat diet-induced obesity (DIO) and TBI in relation to central and peripheral inflammatory pathways, as well as neurological function. Adult male mice were fed a high-fat diet (HFD) for 12 weeks prior to experimental TBI and continuing after injury. Combined TBI and HFD resulted in additive dysfunction in the Y-Maze, novel object recognition (NOR), and Morris water maze (MWM) cognitive function tests. We also performed high-throughput transcriptomic analysis using Nanostring panels of cellular compartments in the brain and total visceral adipose tissue (VAT), followed by unsupervised clustering, principal component analysis, and IPA pathway analysis to determine shifts in gene expression programs and molecular pathway activity. Analysis of cellular populations in the cortex and hippocampus as well as in visceral adipose tissue during the chronic phase after combined TBI-HFD showed amplification of central and peripheral microglia/macrophage responses, including superadditive changes in select gene expression signatures and pathways. These data suggest that HFD-induced obesity and TBI can independently prime and support the development of altered states in brain microglia and visceral adipose tissue macrophages, including the disease-associated microglia/macrophage (DAM) phenotype observed in neurodegenerative disorders. The interaction between HFD and TBI promotes a shift toward chronic reactive microglia/macrophage transcriptomic signatures and associated pro-inflammatory disease-altered states that may, in part, underlie the exacerbation of cognitive deficits. Targeting of HFD-induced reactive cellular phenotypes, including in peripheral adipose tissue macrophages, may serve to reduce microglial maladaptive states after TBI, attenuating post-traumatic neurodegeneration and neurological dysfunction.
Collapse
Affiliation(s)
- Rebecca J. Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - James P. Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria Vaida
- Harrisburg University of Science and Technology, 326 Market St, Harrisburg, PA, USA
| | - Niaz Z. Khan
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Oleg Makarevich
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rodney M. Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A. Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
- VA Maryland Health Care System, Baltimore VA Medical Center, Baltimore, MD 21201, USA
| |
Collapse
|
24
|
Bubnovskaya L, Ganusevich I, Merentsev S, Osinsky D. CANCER-ASSOCIATED ADIPOCYTES AND PROGNOSTIC VALUE OF PREOPERATIVE NEUTROPHIL-LYMPHOCYTE RATIO IN GASTRIC CANCER. Exp Oncol 2023; 45:88-98. [PMID: 37417278 DOI: 10.15407/exp-oncology.2023.01.088] [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: 06/26/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND The neutrophil-to-lymphocyte ratio (NLR) turned out to be a routinely available marker capable to reflect the systemic inflammatory response created by a tumor. Gastric cancer (GC) grows in the anatomical vicinity of adipose tissue, which is also associated with low-grade inflammation. AIM To investigate the usefulness of the combined use of preoperative NLR and density of intratumoral cancer-associated adipocytes (CAAs) for predicting the disease outcome in GC patients. MATERIALS AND METHODS A total of 151 patients with GC were eligible for retrospective analysis between 2009 and 2015.NLR preoperative values were calculated. Perilipin expression in tumor tissue was examined immunohistochemically. RESULTS Low preoperative NLR is the most reliable prognostic factor for the favorable outcome for patients with low density of intratumoral CAAs. Patients with a high density of CCAs are at high risk of lethal outcomes independently of the value of preoperative NLR. CONCLUSION The results have clearly shown an association between preoperative NLR and the density of CAAs in the primary tumor of GC patients. The prognostic value of NLR is essentially modified by means of the individual density of intratumoral CAAs in GC patients.The elevated NLR could be of significant predictive potential for a negative prognosis for patients with tumors characterized by the high density of CAAs independently of BMI.
Collapse
Affiliation(s)
- L Bubnovskaya
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv 03022, Ukraine
| | - I Ganusevich
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv 03022, Ukraine
| | - S Merentsev
- City Clinical Oncological Center, Kyiv 03115, Ukraine
| | - D Osinsky
- City Clinical Oncological Center, Kyiv 03115, Ukraine
| |
Collapse
|
25
|
Li JH, Hepworth MR, O'Sullivan TE. Regulation of systemic metabolism by tissue-resident immune cell circuits. Immunity 2023; 56:1168-1186. [PMID: 37315533 PMCID: PMC10321269 DOI: 10.1016/j.immuni.2023.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023]
Abstract
Recent studies have demonstrated that tissue homeostasis and metabolic function are dependent on distinct tissue-resident immune cells that form functional cell circuits with structural cells. Within these cell circuits, immune cells integrate cues from dietary contents and commensal microbes in addition to endocrine and neuronal signals present in the tissue microenvironment to regulate structural cell metabolism. These tissue-resident immune circuits can become dysregulated during inflammation and dietary overnutrition, contributing to metabolic diseases. Here, we review the evidence describing key cellular networks within and between the liver, gastrointestinal tract, and adipose tissue that control systemic metabolism and how these cell circuits become dysregulated during certain metabolic diseases. We also identify open questions in the field that have the potential to enhance our understanding of metabolic health and disease.
Collapse
Affiliation(s)
- Joey H Li
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA; Medical Scientist Training Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| |
Collapse
|
26
|
Mc Gettigan N, Allen K, Saeidi R, O’ Toole A, Boland K. A systematic review of the effect of structured exercise on inflammation and body composition in inflammatory bowel disease. Int J Colorectal Dis 2023; 38:143. [PMID: 37227593 PMCID: PMC10212817 DOI: 10.1007/s00384-023-04437-2] [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] [Accepted: 05/09/2023] [Indexed: 05/26/2023]
Abstract
PURPOSE Given the substantial risk of treatment failure in inflammatory bowel disease (IBD), adjuvant therapies may play a role in disease management. We aim to carry out a systematic review to examine the effects of structured exercise on the inflammatory response in patients with IBD. Our secondary aim is to examine the effect of structured exercise programmes on body composition given both an increase in visceral obesity and the presence of sarcopenia have deleterious effects on outcomes in IBD. METHODS A systematic review was carried out following the Methodological Expectations of Cochrane Intervention Reviews (MECIR) manual and the Cochrane Handbook for Systematic Reviews of Interventions. Title/Abstract and MeSH Terms were used to search for relevant studies. RESULTS In total, 1516 records were screened for eligibility, and 148 records were reviewed for eligibility, of which 16 were included and a further 7 studies were identified from hand searching references. Four studies included body composition outcomes, and 14 studies reviewed the inflammatory response to exercise. CONCLUSION Further studies of adequate duration are required to include patients with more active disease to demonstrate an inflammatory response to exercise. Body composition measurements including muscle mass and visceral adiposity may play a key role in response to medical therapy in IBD and should be included as exploratory outcomes in future studies. A meta-analysis was not carried out due to the significant heterogeneity amongst studies.
Collapse
Affiliation(s)
- Neasa Mc Gettigan
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Gastroenterology, Beaumont Hospital, Dublin, Ireland
| | - Kathryn Allen
- Department of Gastroenterology, Beaumont Hospital, Dublin, Ireland
| | - Reza Saeidi
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Gastroenterology, Beaumont Hospital, Dublin, Ireland
| | - Aoibhlinn O’ Toole
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Gastroenterology, Beaumont Hospital, Dublin, Ireland
| | - Karen Boland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Gastroenterology, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
27
|
Duong LK, Corbali HI, Riad TS, Ganjoo S, Nanez S, Voss T, Barsoumian HB, Welsh J, Cortez MA. Lipid metabolism in tumor immunology and immunotherapy. Front Oncol 2023; 13:1187279. [PMID: 37205182 PMCID: PMC10185832 DOI: 10.3389/fonc.2023.1187279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023] Open
Abstract
Lipids are a diverse class of biomolecules that have been implicated in cancer pathophysiology and in an array of immune responses, making them potential targets for improving immune responsiveness. Lipid and lipid oxidation also can affect tumor progression and response to treatment. Although their importance in cellular functions and their potential as cancer biomarkers have been explored, lipids have yet to be extensively investigated as a possible form of cancer therapy. This review explores the role of lipids in cancer pathophysiology and describes how further understanding of these macromolecules could prompt novel treatments for cancer.
Collapse
Affiliation(s)
- Lisa K. Duong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Halil Ibrahim Corbali
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Thomas S. Riad
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shonik Ganjoo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Selene Nanez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Tiffany Voss
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B. Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
28
|
Maniyadath B, Zhang Q, Gupta RK, Mandrup S. Adipose tissue at single-cell resolution. Cell Metab 2023; 35:386-413. [PMID: 36889280 PMCID: PMC10027403 DOI: 10.1016/j.cmet.2023.02.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 03/09/2023]
Abstract
Adipose tissue exhibits remarkable plasticity with capacity to change in size and cellular composition under physiological and pathophysiological conditions. The emergence of single-cell transcriptomics has rapidly transformed our understanding of the diverse array of cell types and cell states residing in adipose tissues and has provided insight into how transcriptional changes in individual cell types contribute to tissue plasticity. Here, we present a comprehensive overview of the cellular atlas of adipose tissues focusing on the biological insight gained from single-cell and single-nuclei transcriptomics of murine and human adipose tissues. We also offer our perspective on the exciting opportunities for mapping cellular transitions and crosstalk, which have been made possible by single-cell technologies.
Collapse
Affiliation(s)
- Babukrishna Maniyadath
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Qianbin Zhang
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rana K Gupta
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
29
|
Barthelemy J, Bogard G, Wolowczuk I. Beyond energy balance regulation: The underestimated role of adipose tissues in host defense against pathogens. Front Immunol 2023; 14:1083191. [PMID: 36936928 PMCID: PMC10019896 DOI: 10.3389/fimmu.2023.1083191] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 03/06/2023] Open
Abstract
Although the adipose tissue (AT) is a central metabolic organ in the regulation of whole-body energy homeostasis, it is also an important endocrine and immunological organ. As an endocrine organ, AT secretes a variety of bioactive peptides known as adipokines - some of which have inflammatory and immunoregulatory properties. As an immunological organ, AT contains a broad spectrum of innate and adaptive immune cells that have mostly been studied in the context of obesity. However, overwhelming evidence supports the notion that AT is a genuine immunological effector site, which contains all cell subsets required to induce and generate specific and effective immune responses against pathogens. Indeed, AT was reported to be an immune reservoir in the host's response to infection, and a site of parasitic, bacterial and viral infections. In addition, besides AT's immune cells, preadipocytes and adipocytes were shown to express innate immune receptors, and adipocytes were reported as antigen-presenting cells to regulate T-cell-mediated adaptive immunity. Here we review the current knowledge on the role of AT and AT's immune system in host defense against pathogens. First, we will summarize the main characteristics of AT: type, distribution, function, and extraordinary plasticity. Second, we will describe the intimate contact AT has with lymph nodes and vessels, and AT immune cell composition. Finally, we will present a comprehensive and up-to-date overview of the current research on the contribution of AT to host defense against pathogens, including the respiratory viruses influenza and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Isabelle Wolowczuk
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
30
|
Diverse effects of obesity on antitumor immunity and immunotherapy. Trends Mol Med 2023; 29:112-123. [PMID: 36473793 DOI: 10.1016/j.molmed.2022.11.004] [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: 09/07/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 12/07/2022]
Abstract
Currently, obesity is one of the biggest health burdens facing society because it causes several comorbidities, such as type 2 diabetes, atherosclerosis, and heart disease. Obesity is also linked to multiple types of cancer. Obesity is the second most common preventable cause of cancer after smoking; the rates of obesity are increasing worldwide, as are the rates of obesity-associated cancer. Multiple factors link obesity to cancer, such as increased levels of growth hormones and adipokines, gut dysbiosis, altered tumor metabolism, and chronic low-grade inflammation. More recently, obesity has been shown to also affect the immune response against cancer. In this review we discuss the interplay between obesity, the immune system, and cancer.
Collapse
|
31
|
Tognolli K, Silva V, Sousa-Filho CPB, Cardoso CAL, Gorjão R, Otton R. Green tea beneficial effects involve changes in the profile of immune cells in the adipose tissue of obese mice. Eur J Nutr 2023; 62:321-336. [PMID: 35994086 DOI: 10.1007/s00394-022-02963-3] [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: 10/11/2021] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE During obesity, the adipose tissue is usually infiltrated by immune cells which are related to hallmarks of obesity such as systemic inflammation and insulin resistance (IR). Green tea (GT) has been widely studied for its anti-inflammatory actions, including the modulation in the proliferation and activity of immune cells, in addition to preventing cardiovascular and metabolic diseases. METHODS The aim of the present study was to analyze the population of immune cells present in the subcutaneous and epididymal white adipose tissue (WAT) of mice kept at thermoneutrality (TN) and fed with a high-fat diet (HFD) for 16 weeks, supplemented or not with GT extract (500 mg/kg/12 weeks). RESULTS The HFD in association with TN has induced chronic inflammation, and IR in parallel with changes in the profile of immune cells in the subcutaneous and epidydimal WAT, increasing pro-inflammatory cytokines release, inflammatory cells infiltration, and fibrotic aspects in WAT. On the other hand, GT prevented body weight gain, in addition to avoiding IR and inflammation, and the consequent tissue fibrosis, maintaining a lower concentration of cytokines and a profile of immune cells similar to the control mice, preventing the harmful modulations induced by both HFD and TN. CONCLUSIONS GT beneficial effects in WAT abrogated the deleterious effects triggered by HFD and TN, maintaining all immune cells and fibrotic markers at the same level as in lean mice. These results place WAT immune cells population as a potential target of GT action, also highlighting the positive effects of GT in obese mice housed at TN.
Collapse
Affiliation(s)
- Kaue Tognolli
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil
| | - Victoria Silva
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil
| | - Celso Pereira Batista Sousa-Filho
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil
| | | | - Renata Gorjão
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil
| | - Rosemari Otton
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, Regente Feijó Avenue, 1295, Sao Paulo, SP, 03342-000, Brazil.
| |
Collapse
|
32
|
Sun K, Li X, Scherer PE. Extracellular Matrix (ECM) and Fibrosis in Adipose Tissue: Overview and Perspectives. Compr Physiol 2023; 13:4387-4407. [PMID: 36715281 PMCID: PMC9957663 DOI: 10.1002/cphy.c220020] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation. It is characterized by an overaccumulation of extracellular matrix (ECM) during unhealthy expansion of adipose tissue in response to over nutrition. In obese adipose-depots, hypoxia stimulates multiple pro-fibrotic signaling pathways in different cell populations, thereby inducing the overproduction of the ECM components, including collagens, noncollagenous proteins, and additional enzymatic components of ECM synthesis. As a consequence, local fibrosis develops. The result of fibrosis-induced mechanical stress not only triggers cell necrosis and inflammation locally in adipose tissue but also leads to system-wide lipotoxicity and insulin resistance. A better understanding of the mechanisms underlying the obesity-induced fibrosis will help design therapeutic approaches to reduce or reverse the pathological changes associated with obese adipose tissue. Here, we aim to summarize the major advances in the field, which include newly identified fibrotic factors, cell populations that contribute to the fibrosis in adipose tissue, as well as novel mechanisms underlying the development of fibrosis. We further discuss the potential therapeutic strategies to target fibrosis in adipose tissue for the treatment of obesity-linked metabolic diseases and cancer. © 2023 American Physiological Society. Compr Physiol 13:4387-4407, 2023.
Collapse
Affiliation(s)
- Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Philipp E. Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
33
|
Cai Z, He B. Adipose tissue aging: An update on mechanisms and therapeutic strategies. Metabolism 2023; 138:155328. [PMID: 36202221 DOI: 10.1016/j.metabol.2022.155328] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
Aging is a complex biological process characterized by a progressive loss of physiological integrity and increased vulnerability to age-related diseases. Adipose tissue plays central roles in the maintenance of whole-body metabolism homeostasis and has recently attracted significant attention as a biological driver of aging and age-related diseases. Here, we review the most recent advances in our understanding of the molecular and cellular mechanisms underlying age-related decline in adipose tissue function. In particular, we focus on the complex inter-relationship between metabolism, immune, and sympathetic nervous system within adipose tissue during aging. Moreover, we discuss the rejuvenation strategies to delay aging and extend lifespan, including senescent cell ablation (senolytics), dietary intervention, physical exercise, and heterochronic parabiosis. Understanding the pathological mechanisms that underlie adipose tissue aging will be critical for the development of new intervention strategies to slow or reverse aging and age-related diseases.
Collapse
Affiliation(s)
- Zhaohua Cai
- Heart Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Ben He
- Heart Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China.
| |
Collapse
|
34
|
Zhang YX, Ou MY, Yang ZH, Sun Y, Li QF, Zhou SB. Adipose tissue aging is regulated by an altered immune system. Front Immunol 2023; 14:1125395. [PMID: 36875140 PMCID: PMC9981968 DOI: 10.3389/fimmu.2023.1125395] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023] Open
Abstract
Adipose tissue is a widely distributed organ that plays a critical role in age-related physiological dysfunctions as an important source of chronic sterile low-grade inflammation. Adipose tissue undergoes diverse changes during aging, including fat depot redistribution, brown and beige fat decrease, functional decline of adipose progenitor and stem cells, senescent cell accumulation, and immune cell dysregulation. Specifically, inflammaging is common in aged adipose tissue. Adipose tissue inflammaging reduces adipose plasticity and pathologically contributes to adipocyte hypertrophy, fibrosis, and ultimately, adipose tissue dysfunction. Adipose tissue inflammaging also contributes to age-related diseases, such as diabetes, cardiovascular disease and cancer. There is an increased infiltration of immune cells into adipose tissue, and these infiltrating immune cells secrete proinflammatory cytokines and chemokines. Several important molecular and signaling pathways mediate the process, including JAK/STAT, NFκB and JNK, etc. The roles of immune cells in aging adipose tissue are complex, and the underlying mechanisms remain largely unclear. In this review, we summarize the consequences and causes of inflammaging in adipose tissue. We further outline the cellular/molecular mechanisms of adipose tissue inflammaging and propose potential therapeutic targets to alleviate age-related problems.
Collapse
Affiliation(s)
- Yi-Xiang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min-Yi Ou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zi-Han Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang-Bai Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Papotti B, Opstad TB, Åkra S, Tønnessen T, Braathen B, Hansen CH, Arnesen H, Solheim S, Seljeflot I, Ronda N. Macrophage polarization markers in subcutaneous, pericardial, and epicardial adipose tissue are altered in patients with coronary heart disease. Front Cardiovasc Med 2023; 10:1055069. [PMID: 36937936 PMCID: PMC10017535 DOI: 10.3389/fcvm.2023.1055069] [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: 09/27/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Background Epicardial and pericardial adipose tissue (EAT and PAT) surround and protect the heart, with EAT directly sharing the microcirculation with the myocardium, possibly presenting a distinct macrophage phenotype that might affect the inflammatory environment in coronary heart disease (CHD). This study aims to investigate the expression of genes in different AT compartments driving the polarization of AT macrophages toward an anti-inflammatory (L-Galectin 9; CD206) or pro-inflammatory (NOS2) phenotype. Methods EAT, PAT, and subcutaneous (SAT) biopsies were collected from 52 CHD patients undergoing coronary artery bypass grafting, and from 22 CTRLs undergoing aortic valve replacement. L-Galectin9 (L-Gal9), CD206, and NOS2 AT gene expression and circulating levels were analyzed through RT-PCR and ELISA, respectively. Results L-Gal9, CD206, and NOS2 gene expression was similar in all AT compartments in CHD and CTRLs, as were also L-Gal9 and CD206 circulating levels, while NOS2 serum levels were higher in CHD (p = 0.012 vs. CTRLs). In CTRLs, NOS2 expression was lower in EAT vs. SAT (p = 0.007), while in CHD patients CD206 expression was lower in both SAT and EAT as compared to PAT (p = 0.003, p = 0.006, respectively), suggestive of a possible macrophage reprogramming toward a pro-inflammatory phenotype in EAT. In CHD patients, NOS2 expression in SAT correlated to that in PAT and EAT (p = 0.007, both), CD206 expression correlated positively to L-Gal9 (p < 0.001) only in EAT, and CD206 expression associated with that of macrophage identifying markers in all AT compartments (p < 0.001, all). In CHD patients, subjects with LDL-C above 1.8 mmol/L showed significantly higher NOS2 expression in PAT and EAT as compared to subjects with LDL-C levels below (p < 0.05), possibly reflecting increased cardiac AT pro-inflammatory activation. In SAT and PAT, CD206 expression associated with BMI in both CHD and CTRLs (p < 0.05, all), and with L-Gal9 in EAT, however only in CTRLs (p = 0.002). Conclusion CHD seems to be accompanied by an altered cardiac, and especially epicardial AT macrophage polarization. This may represent an important pathophysiological mechanism and a promising field of therapy targeting the excessive AT inflammation, in need of further investigation.
Collapse
Affiliation(s)
- Bianca Papotti
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
- Department of Food and Drug, University of Parma, Parma, Italy
- *Correspondence: Bianca Papotti,
| | - Trine Baur Opstad
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sissel Åkra
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Theis Tønnessen
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Bjørn Braathen
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Charlotte Holst Hansen
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Harald Arnesen
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Svein Solheim
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Ingebjørg Seljeflot
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parma, Italy
| |
Collapse
|
36
|
Essential Minerals and Metabolic Adaptation of Immune Cells. Nutrients 2022; 15:nu15010123. [PMID: 36615781 PMCID: PMC9824256 DOI: 10.3390/nu15010123] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
Modern lifestyles deviated considerably from the ancestral routines towards major shifts in diets and increased sedentarism. The trace elements status of the human body is no longer adequately supported by micronutrient-inferior farmed meats and crop commodities produced by the existing agricultural food systems. This is particular evident in the increased obesogenic adipogenesis and low-grade inflammation that fails to resolve with time. The metabolically restrictive environment of the inflamed tissues drives activation and proliferation of transient and resident populations of immune cells in favor of pro-inflammatory phenotypes, as well as a part of the enhanced autoimmune response. As different stages of the immune activation and resolution depend on the availability of specific minerals to maintain the structural integrity of skin and mucus membranes, activation and migration of immune cells, activation of the complement system, and the release of pro-inflammatory cytokines and chemokines, this review discusses recent advances in our understanding of the contribution of select minerals in optimizing the responses of innate and adaptive immune outcomes. An abbreviated view on the absorption, transport, and delivery of minerals to the body tissues as related to metabolic adaptation is considered.
Collapse
|
37
|
Kolb H. Obese visceral fat tissue inflammation: from protective to detrimental? BMC Med 2022; 20:494. [PMID: 36575472 PMCID: PMC9795790 DOI: 10.1186/s12916-022-02672-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/21/2022] [Indexed: 12/28/2022] Open
Abstract
Obesity usually is accompanied by inflammation of fat tissue, with a prominent role of visceral fat. Chronic inflammation in obese fat tissue is of a lower grade than acute immune activation for clearing the tissue from an infectious agent. It is the loss of adipocyte metabolic homeostasis that causes activation of resident immune cells for supporting tissue functions and regaining homeostasis. Initially, the excess influx of lipids and glucose in the context of overnutrition is met by adipocyte growth and proliferation. Eventual lipid overload of hypertrophic adipocytes leads to endoplasmic reticulum stress and the secretion of a variety of signals causing increased sympathetic tone, lipolysis by adipocytes, lipid uptake by macrophages, matrix remodeling, angiogenesis, and immune cell activation. Pro-inflammatory signaling of adipocytes causes the resident immune system to release increased amounts of pro-inflammatory and other mediators resulting in enhanced tissue-protective responses. With chronic overnutrition, these protective actions are insufficient, and death of adipocytes as well as senescence of several tissue cell types is seen. This structural damage causes the expression or release of immunostimulatory cell components resulting in influx and activation of monocytes and many other immune cell types, with a contribution of stromal cells. Matrix remodeling and angiogenesis is further intensified as well as possibly detrimental fibrosis. The accumulation of senescent cells also may be detrimental via eventual spread of senescence state from affected to neighboring cells by the release of microRNA-containing vesicles. Obese visceral fat inflammation can be viewed as an initially protective response in order to cope with excess ambient nutrients and restore tissue homeostasis but may contribute to tissue damage at a later stage.
Collapse
Affiliation(s)
- Hubert Kolb
- Faculty of Medicine, University of Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany. .,West-German Centre of Diabetes and Health, Düsseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Düsseldorf, Germany.
| |
Collapse
|
38
|
Abstract
Oncostatin M (OSM) is a member of the glycoprotein 130 cytokine family that is involved in chronic inflammation and increased in adipose tissue under obesity and insulin resistance. OSM was shown to inhibit adipogenesis, suppress browning, and contribute to insulin resistance in cultured white adipocytes. In contrast, OSM may have a metabolically favourable role on adipocytes in mouse models of obesity and insulin resistance. However, a putative role of OSM in modulating lipolysis has not been investigated in detail to date. To address this, cultured white adipocytes of mouse or human origin were exposed to 10 or 100 ng/ml of OSM for various time periods. In murine 3T3-L1 cells, OSM stimulation directly activated hormone-sensitive lipase (HSL) and other players of the lipolytic machinery, and dose-dependently increased free fatty acid and glycerol release. In parallel, OSM attenuated insulin-mediated suppression of lipolysis and induced phosphorylation of serine-residues on the insulin receptor substrate-1 (IRS1) protein. Key experiments were verified in a second murine and a human adipocyte cell line. Inhibiton of extracellular signal-regulated kinase (ERK)-1/2 activation, abolished OSM-mediated HSL phosphorylation and lipolysis. In conclusion, OSM signalling directly promotes lipolysis in white adipocytes in an ERK1/2-dependent manner.
Collapse
Affiliation(s)
- Pim P. van Krieken
- Division of Pediatric Endocrinology and Diabetology, University Children’s Hospital, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital, University of Zurich, Zurich, Switzerland
| | - Julian Roos
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | | | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children’s Hospital, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital, University of Zurich, Zurich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children’s Hospital, University of Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Qian X, Meng X, Zhang S, Zeng W. Neuroimmune regulation of white adipose tissues. FEBS J 2022; 289:7830-7853. [PMID: 34564950 DOI: 10.1111/febs.16213] [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: 05/05/2021] [Revised: 08/21/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
The white adipose tissues (WAT) are located in distinct depots throughout the body. They serve as an energy reserve, providing fatty acids for other tissues via lipolysis when needed, and function as an endocrine organ to regulate systemic metabolism. Their activities are coordinated through intercellular communications among adipocytes and other cell types such as residential and infiltrating immune cells, which are collectively under neuronal control. The adipocytes and immune subtypes including macrophages/monocytes, eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2s), T and B cells, dendritic cells (DCs), and natural killer (NK) cells display cellular and functional diversity in response to the energy states and contribute to metabolic homeostasis and pathological conditions. Accumulating evidence reveals that neuronal innervations control lipid deposition and mobilization via regulating lipolysis, adipocyte size, and cellularity. Vice versa, the neuronal innervations and activity are influenced by cellular factors in the WAT. Though the literature describing adipose tissue cells is too extensive to cover in detail, we strive to highlight a selected list of neuronal and immune components in this review. The cell-to-cell communications and the perspective of neuroimmune regulation are emphasized to enlighten the potential therapeutic opportunities for treating metabolic disorders.
Collapse
Affiliation(s)
- Xinmin Qian
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xia Meng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Shan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
40
|
Correale J, Marrodan M. Multiple sclerosis and obesity: The role of adipokines. Front Immunol 2022; 13:1038393. [PMID: 36457996 PMCID: PMC9705772 DOI: 10.3389/fimmu.2022.1038393] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/28/2022] [Indexed: 11/24/2023] Open
Abstract
Multiple Sclerosis (MS), a chronic inflammatory disease of the central nervous system that leads to demyelination and neurodegeneration has been associated with various environmental and lifestyle factors. Population-based studies have provided evidence showing the prevalence of MS is increasing worldwide. Because a similar trend has been observed for obesity and metabolic syndrome, interest has grown in possible underlying biological mechanisms shared by both conditions. Adipokines, a family of soluble factors produced by adipose tissue that participate in a wide range of biological functions, contribute to a low state of chronic inflammation observed in obesity, and influence immune function, metabolism, and nutritional state. In this review, we aim to describe epidemiological and biological factors common to MS and obesity, as well as provide an update on current knowledge of how different pro- and anti-inflammatory adipokines participate as immune response mediators in MS, as well as in the animal model for MS, namely, experimental autoimmune encephalomyelitis (EAE). Multiple Sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) leading to demyelination, and neurodegeneration. Although its pathogenesis is not yet fully understood, there is considerable evidence to suggest MS arises from complex interactions between individual genetic susceptibility and external environmental factors. In recent decades, population-based studies have provided evidence indicating the prevalence of MS is increasing worldwide, in parallel with the rise in obesity and metabolic syndrome. This synchronous increment in the incidence of both MS and obesity has led to a search for potential biological mechanisms linking both conditions. Notably, a large number of studies have established significant correlation between obesity and higher prevalence, or worse prognosis, of several immune-mediated conditions. Fat tissue has been found to produce a variety of soluble factors named adipokines. These mediators, secreted by both adipocytes as well as diverse immune cells, participate in a wide range of biological functions, further strengthening the concept of a link between immune function, metabolism, and nutritional state. Because obesity causes overproduction of pro-inflammatory adipokines (namely leptin, resistin and visfatin) and reduction of anti-inflammatory adipokines (adiponectin and apelin), adipose tissue dysregulation would appear to contribute to a state of chronic, low-grade inflammation favoring the development of disease. In this review, we present a summary of current knowledge related to the pathological effects of different adipokines, prevalent in obese MS patients.
Collapse
Affiliation(s)
- Jorge Correale
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires/Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | |
Collapse
|
41
|
Becker YLC, Duvvuri B, Fortin PR, Lood C, Boilard E. The role of mitochondria in rheumatic diseases. Nat Rev Rheumatol 2022; 18:621-640. [PMID: 36175664 DOI: 10.1038/s41584-022-00834-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2022] [Indexed: 11/09/2022]
Abstract
The mitochondrion is an intracellular organelle thought to originate from endosymbiosis between an ancestral eukaryotic cell and an α-proteobacterium. Mitochondria are the powerhouses of the cell, and can control several important processes within the cell, such as cell death. Conversely, dysregulation of mitochondria possibly contributes to the pathophysiology of several autoimmune diseases. Defects in mitochondria can be caused by mutations in the mitochondrial genome or by chronic exposure to pro-inflammatory cytokines, including type I interferons. Following the release of intact mitochondria or mitochondrial components into the cytosol or the extracellular space, the bacteria-like molecular motifs of mitochondria can elicit pro-inflammatory responses by the innate immune system. Moreover, antibodies can target mitochondria in autoimmune diseases, suggesting an interplay between the adaptive immune system and mitochondria. In this Review, we discuss the roles of mitochondria in rheumatic diseases such as systemic lupus erythematosus, antiphospholipid syndrome and rheumatoid arthritis. An understanding of the different contributions of mitochondria to distinct rheumatic diseases or manifestations could permit the development of novel therapeutic strategies and the use of mitochondria-derived biomarkers to inform pathogenesis.
Collapse
Affiliation(s)
- Yann L C Becker
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada
- Département de microbiologie et immunologie, Université Laval, Québec, QC, Canada
| | - Bhargavi Duvvuri
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Paul R Fortin
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada
- Division of Rheumatology, Department of Medicine, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA.
| | - Eric Boilard
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada.
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada.
- Département de microbiologie et immunologie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
42
|
Bertola A, Gallerand A, Ivanov S. Immune cell involvement in brown adipose tissue functions. DISCOVERY IMMUNOLOGY 2022; 1:kyac007. [PMID: 38566905 PMCID: PMC10917225 DOI: 10.1093/discim/kyac007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/15/2022] [Accepted: 10/28/2022] [Indexed: 04/04/2024]
Abstract
Brown adipose tissue (BAT) contains many immune cells. The presence of macrophages, monocytes, dendritic cells, T cells, B cells, and mast cells was documented in BAT. However, in comparison to white adipose tissue, relatively little is known on the impact of immune cells on BAT function. By directly interacting with BAT stromal cells, or by secreting pro- and anti-inflammatory mediators, immune cells modulate BAT activation and subsequently influence on adaptative thermogenesis and heat generation. In the current manuscript, we will focus on the diversity and functions of BAT immune cells.
Collapse
|
43
|
Wang G, Song A, Bae M, Wang QA. Adipose Tissue Plasticity in Aging. Compr Physiol 2022; 12:4119-4132. [PMID: 36214190 DOI: 10.1002/cphy.c220005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
As a dynamic endocrine organ, white adipose tissue (WAT) stores lipids and plays a critical role in maintaining whole-body energy homeostasis and insulin sensitivity. A large group of the population over 65 years old suffer from increased WAT mass, especially in the visceral location. Visceral adiposity accelerates aging through promoting age-associated chronic conditions, significantly shortening life expectancy. Unlike WAT, brown adipose tissue (BAT) functions as an effective energy sink that burns and disposes of excess lipids and glucose upon activation of thermogenesis. Unfortunately, the thermogenic activity of BAT declines during aging. New appreciation of cellular and functional remodeling of WAT and BAT during aging has emerged in recent years. Efforts are underway to explore the potential underlying mechanisms behind these age-associated alterations in WAT and BAT and the impact of these alterations on whole-body metabolism. Lastly, it is intriguing to translate our knowledge obtained from animal models to the clinic to prevent and treat age-associated metabolic disorders. © 2022 American Physiological Society. Compr Physiol 12: 4119-4132, 2022.
Collapse
Affiliation(s)
- Guan Wang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Anying Song
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Marie Bae
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, California, USA
| | - Qiong A Wang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, California, USA.,Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| |
Collapse
|
44
|
Presti EL, Nuzzo D, Al Mahmeed W, Al-Rasadi K, Al-Alawi K, Banach M, Banerjee Y, Ceriello A, Cesur M, Cosentino F, Firenze A, Galia M, Goh SY, Janez A, Kalra S, Kapoor N, Kempler P, Lessan N, Lotufo P, Papanas N, Rizvi AA, Sahebkar A, Santos RD, Stoian AP, Toth PP, Viswanathan V, Rizzo M. Molecular and pro-inflammatory aspects of COVID-19: The impact on cardiometabolic health. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166559. [PMID: 36174875 PMCID: PMC9510069 DOI: 10.1016/j.bbadis.2022.166559] [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: 08/08/2022] [Revised: 09/03/2022] [Accepted: 09/21/2022] [Indexed: 12/15/2022]
Abstract
Obesity, type 2 diabetes (T2DM), hypertension (HTN), and Cardiovascular Disease (CVD) often cluster together as “Cardiometabolic Disease” (CMD). Just under 50% of patients with CMD increased the risk of morbidity and mortality right from the beginning of the COVID-19 pandemic as it has been reported in most countries affected by the SARS-CoV2 virus. One of the pathophysiological hallmarks of COVID-19 is the overactivation of the immune system with a prominent IL-6 response, resulting in severe and systemic damage involving also cytokines such as IL2, IL4, IL8, IL10, and interferon-gamma were considered strong predictors of COVID-19 severity. Thus, in this mini-review, we try to describe the inflammatory state, the alteration of the adipokine profile, and cytokine production in the obese state of infected and not infected patients by SARS-CoV2 with the final aim to find possible influences of COVID-19 on CMD and CVD. The immunological-based discussion of the molecular processes could inspire the study of promising targets for managing CMD patients and its complications during COVID-19.
Collapse
Affiliation(s)
- Elena Lo Presti
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Domenico Nuzzo
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Wael Al Mahmeed
- Heart and Vascular Institute, Cleveland Clinic, Abu Dhabi, United Arab Emirates
| | | | - Kamila Al-Alawi
- Department of Training and Studies, Royal Hospital, Ministry of Health, Muscat, Oman
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Yajnavalka Banerjee
- Department of Biochemistry, Mohamed Bin Rashid University, Dubai, United Arab Emirates
| | | | - Mustafa Cesur
- Clinic of Endocrinology, Ankara Güven Hospital, Ankara, Turkey
| | - Francesco Cosentino
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, University of Stockholm, Sweden
| | - Alberto Firenze
- Unit of Research and International Cooperation, University Hospital of Palermo, Italy
| | - Massimo Galia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bind), University of Palermo, Italy
| | - Su-Yen Goh
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Slovenia
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital & BRIDE, Karnal, India
| | - Nitin Kapoor
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, India; Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter Kempler
- Department of Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Nader Lessan
- The Research Institute, Imperial College London Diabetes Centre, Abu Dhabi, United Arab Emirates
| | - Paulo Lotufo
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, Brazil
| | - Nikolaos Papanas
- Diabetes Center, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece
| | - Ali A Rizvi
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raul D Santos
- Heart Institute (InCor) University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil; Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Anca P Stoian
- Faculty of Medicine, Diabetes, Nutrition and Metabolic Diseases, Carol Davila University, Bucharest, Romania
| | - Peter P Toth
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, Italy.
| | | |
Collapse
|
45
|
Yao J, Wu D, Qiu Y. Adipose tissue macrophage in obesity-associated metabolic diseases. Front Immunol 2022; 13:977485. [PMID: 36119080 PMCID: PMC9478335 DOI: 10.3389/fimmu.2022.977485] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Adipose tissue macrophage (ATM) has been appreciated for its critical contribution to obesity-associated metabolic diseases in recent years. Here, we discuss the regulation of ATM on both metabolic homeostatsis and dysfunction. In particular, the macrophage polarization and recruitment as well as the crosstalk between ATM and adipocyte in thermogenesis, obesity, insulin resistance and adipose tissue fibrosis have been reviewed. A better understanding of how ATM regulates adipose tissue remodeling may provide novel therapeutic strategies against obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Jingfei Yao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Dongmei Wu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yifu Qiu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- *Correspondence: Yifu Qiu,
| |
Collapse
|
46
|
Ma Y, Jun H, Wu J. Immune cell cholinergic signaling in adipose thermoregulation and immunometabolism. Trends Immunol 2022; 43:718-727. [PMID: 35931611 PMCID: PMC9727785 DOI: 10.1016/j.it.2022.07.006] [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: 06/08/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022]
Abstract
Research focusing on adipose immunometabolism has been expanded from inflammation in white fat during obesity development to immune cell function regulating thermogenic fat, energy expenditure, and systemic metabolism. This opinion discusses our current understanding of how resident immune cells within the thermogenic fat niche may regulate whole-body energy homeostasis. Furthermore, various types of immune cells can synthesize acetylcholine (ACh) and regulate important physiological functions. We highlight a unique subset of cholinergic macrophages within subcutaneous adipose tissue, termed cholinergic adipose macrophages (ChAMs); these macrophages interact with beige adipocytes through cholinergic receptor nicotinic alpha 2 subunit (CHRNA2) signaling to induce adaptive thermogenesis. We posit that these newly identified thermoregulatory macrophages may broaden our view of immune system functions for maintaining metabolic homeostasis and potentially treating obesity and metabolic disorders.
Collapse
Affiliation(s)
- Yingxu Ma
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA; Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Heejin Jun
- Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - Jun Wu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Mc Gettigan N, Hanley M, Skelly F, Dowling J, Dunne R, Morrin MM, McCaffrey N, O'Toole A, Boland K. Impact of a physician-led exercise programme on quality of life, muscle mass and clinical response in inflammatory bowel disease patients during induction with disease-modifying therapy: a study protocol. BMJ Open Gastroenterol 2022. [DOI: 10.1136/bmjgast-2022-000959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
IntroductionBiologic and small-molecule therapies have revolutionised the treatment of moderate-to-severe inflammatory bowel disease (IBD). A significant proportion of patients experience early or delayed treatment failure. Patients with IBD with greater visceral obesity are less likely to respond to biologics. Sarcopenia has been identified as a predictor of disease severity and need for rescue therapy in acute severe ulcerative colitis. The aim of this study is to assess the feasibility of a physician-derived exercise programme in patients with IBD commencing biologic or small-molecule therapy in addition to the effect on physical fitness, body composition and objective measures of quality of life, fatigue scores and disease activity.Methods and analysisThis is a randomised controlled feasibility study comparing the effects of a physician-derived exercise programme and standard medical care (biologic/small-molecule therapy) with standard care alone in patients with moderate to severe IBD. Patients with IBD in the intervention group will undergo a structured exercise programme for 20 weeks. Both IBD groups will carry out body composition, disease activity and quality-of-life assessments at baseline, week 12 and week 26. The primary objective is to assess the feasibility of the physician-derived exercise programme in patients with IBD commencing disease-modifying therapies. Secondary endpoints include a change in cardiorespiratory fitness, disease activity/inflammation, fatigue, health-related quality of life outcomes and body composition between the two IBD groups. Exploratory endpoints include validation of anterior thigh ultrasound for sarcopenia screening, assessment of proinflammatory cytokines and markers of immunometabolism.Ethics and disseminationThis study has received ethical approval from the Beaumont Hospital Ethics committee on 22 October 2021 (reference number 21/21). Data generated or analysed during this study will be published as an article and supplementary appendix in relevant medical journals. The data will also be presented at national and international conferences.Trial registration numberNCT05174754.
Collapse
|
48
|
Almengló C, Fu X, Flores-Arias MT, Fernández ÁL, Viñuela JE, Martínez-Cereijo JM, Durán D, Rodríguez-Mañero M, González-Juanatey JR, Eiras S. Synergism between obesity and HFpEF on neutrophils phenotype and its regulation by adipose tissue-molecules and SGLT2i dapagliflozin. J Cell Mol Med 2022; 26:4416-4427. [PMID: 35818731 PMCID: PMC9357605 DOI: 10.1111/jcmm.17466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/30/2022] [Accepted: 06/15/2022] [Indexed: 11/27/2022] Open
Abstract
The adiposity invokes innate immune activity, coronary microvascular dysfunction and consequently heart failure preserved ejection fraction (HFpEF). Our aim was to study the neutrophils profile on obesity and cardiovascular disease and its regulation by adipose tissue-secretome and dapagliflozin. We have isolated neutrophils from patients undergoing open heart surgery (19 women and 51 men). Its migration activity was performed with culture-transwell, transcriptional studies of proteolytic enzymes, adhesion molecules or receptors were analysed by real-time PCR and proteomics (from 20 patients) analysis by TripleTOF mass spectrometer. Differentiated HL-60 (dHL-60) was used as a preclinical model on microfluidic for endothelial cells attaching assays and genes regulation with epicardial and subcutaneous fat secretomes from patients (3 women and 9 men) or dapagliflozin 1-10 μM treatments. The transcriptional and proteomics studies have determined higher levels of adhesion molecules in neutrophils from patients with obesity. The adhesion molecule CD11b levels were higher in those patients with the combined obesity and HFpEF factors (1.70 ± 0.06 a.u. without obesity, 1.72 ± 0.04 a.u. obesity or HFpEF without obesity and 1.79 ± 0.08 a.u. obesity and HFpEF; p < .01). While fat-secretome induces its upregulation, dapagliflozin can modulated it. Because CD11b upregulation is associated with higher neutrophils migration and adhesion into endothelial cells, dapagliflozin might modulate this mechanism on patients with obesity and HFpEF.
Collapse
Affiliation(s)
- Cristina Almengló
- Cardiology Group, Health Research Institute, Santiago de Compostela, Spain
| | - Xiaoran Fu
- Translational Cardiology Group, Health Research Institute, Santiago de Compostela, Spain
| | - María Teresa Flores-Arias
- Photonics4 Life Research Group, Applied Physics Department, Facultade de Física and Facultade de Óptica e Optometría, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ángel L Fernández
- Heart Surgery Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV Madrid, Madrid, Spain
| | - Juan E Viñuela
- Translational Cardiology Group, Health Research Institute, Santiago de Compostela, Spain.,Immunology Laboratory, Health Research Institute, Santiago de Compostela, Spain
| | - José M Martínez-Cereijo
- Heart Surgery Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Darío Durán
- Heart Surgery Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain.,CIBERCV Madrid, Madrid, Spain
| | - Moisés Rodríguez-Mañero
- Translational Cardiology Group, Health Research Institute, Santiago de Compostela, Spain.,CIBERCV Madrid, Madrid, Spain.,Cardiovascular Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - José R González-Juanatey
- Cardiology Group, Health Research Institute, Santiago de Compostela, Spain.,CIBERCV Madrid, Madrid, Spain.,Cardiovascular Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Sonia Eiras
- Translational Cardiology Group, Health Research Institute, Santiago de Compostela, Spain.,CIBERCV Madrid, Madrid, Spain
| |
Collapse
|
49
|
Widyahening IS, Vidiawati D, Pakasi TA, Soewondo P, Ahsan A. Noncommunicable diseases risk factors and the risk of COVID-19 among university employees in Indonesia. PLoS One 2022; 17:e0263146. [PMID: 35666734 PMCID: PMC9170090 DOI: 10.1371/journal.pone.0263146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Noncommunicable diseases (NCDs) are still a major public health problem in Indonesia. Studies have shown that risk factors of NCDs are associated with coronavirus disease 2019 (COVID-19) severity and mortality. However, it is unclear whether NCD risk factors are also risks for new COVID-19 cases. This study aimed to obtain an NCD risk profile among university employees and its associations with contracting COVID-19. Methods A cross-sectional study was conducted in October 2021. Participants were administrative employees of Universitas Indonesia (UI), Depok City, West Java. Assessment of NCD risk factors was based on the World Health Organization STEPwise approach to NCD risk factor surveillance (WHO STEPS). Demographic, working, and medical-history data were obtained electronically by using a Google Form. Physical and laboratory examinations were done in the Integrated Post for NCDs. Risks were expressed as adjusted odds ratio (ORadj) and 95% confidence interval (CI) in multivariate analyses. Results A total of 613 employees were enrolled. Men were predominant (54.8%), and about 36% of them work in shift as security personnel. About 66.7% were overweight or obese and 77.8% had hypertension. There were 138 (22.8%) employees who had COVID-19. Nearly all (95.6%) had been fully vaccinated against COVID-19. At-risk waist circumference (ORadj 1.72, 95% CI 1.15–2.56, p = 0.008) and total cholesterol level of 200–239 mg/dL (ORadj 2.30, 95% CI 1.19–4.44, p = 0.013) were independent risk factors, but shift work (ORadj 0.52, 95% CI 0.34–0.80, p = 0.003) was protective against COVID-19. Conclusion The prevalence of NCD risk factors among university administrative employees was high, increasing the risk of contracting COVID-19. A behavioral intervention program to manage NCD risk factors at the university level is urgently needed according to the Health Promoting University framework.
Collapse
Affiliation(s)
- Indah Suci Widyahening
- Department of Community Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta Pusat, Indonesia
- Southeast Asian Ministers of Education Organization—Regional Centre for Food and Nutrition (SEAMEO-RECFON)—Pusat Kajian Gizi Regional (PKGR), Universitas Indonesia, Jakarta, Indonesia
- * E-mail:
| | - Dhanasari Vidiawati
- Department of Community Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta Pusat, Indonesia
| | - Trevino A. Pakasi
- Department of Community Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta Pusat, Indonesia
| | - Pradana Soewondo
- Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia–Cipto Mangunkusumo Hospital, Jakarta Pusat, Indonesia
| | - Abdillah Ahsan
- Department of Economics, Faculty of Economics and Business, Universitas Indonesia, Depok, Indonesia
| |
Collapse
|
50
|
Chen Y, Zhang J, Cui W, Silverstein RL. CD36, a signaling receptor and fatty acid transporter that regulates immune cell metabolism and fate. J Exp Med 2022; 219:213166. [PMID: 35438721 PMCID: PMC9022290 DOI: 10.1084/jem.20211314] [Citation(s) in RCA: 145] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/13/2022] Open
Abstract
CD36 is a type 2 cell surface scavenger receptor widely expressed in many immune and non-immune cells. It functions as both a signaling receptor responding to DAMPs and PAMPs, as well as a long chain free fatty acid transporter. Recent studies have indicated that CD36 can integrate cell signaling and metabolic pathways through its dual functions and thereby influence immune cell differentiation and activation, and ultimately help determine cell fate. Its expression along with its dual functions in both innate and adaptive immune cells contribute to pathogenesis of common diseases, including atherosclerosis and tumor progression, which makes CD36 and its downstream effectors potential therapeutic targets. This review comprehensively examines the dual functions of CD36 in a variety of immune cells, especially macrophages and T cells. We also briefly discuss CD36 function in non-immune cells, such as adipocytes and platelets, which impact the immune system via intercellular communication. Finally, outstanding questions in this field are provided for potential directions of future studies.
Collapse
Affiliation(s)
- Yiliang Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI.,Versiti, Blood Research Institute, Milwaukee, WI
| | - Jue Zhang
- Versiti, Blood Research Institute, Milwaukee, WI
| | - Weiguo Cui
- Versiti, Blood Research Institute, Milwaukee, WI.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Roy L Silverstein
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI.,Versiti, Blood Research Institute, Milwaukee, WI
| |
Collapse
|