1
|
Xiong B, Wang H, Song YX, Lan WY, Li J, Wang F. Natural saponins and macrophage polarization: Mechanistic insights and therapeutic perspectives in disease management. Front Pharmacol 2025; 16:1584035. [PMID: 40417220 PMCID: PMC12098594 DOI: 10.3389/fphar.2025.1584035] [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: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 05/27/2025] Open
Abstract
Macrophage polarization plays a pivotal role in immune homeostasis and disease progression across inflammatory, neoplastic, and metabolic disorders. Saponins, which are natural compounds with steroidal/triterpenoid structures, demonstrate therapeutic potential through immunomodulatory, anti-inflammatory, and anti-tumor activities. This study aims to highlight the potential of key saponins-such as ginsenosides, astragaloside IV, dioscin, platycodin D, pulsatilla saponins, and panax notoginseng saponins-in modulating macrophage polarization and enhancing conventional therapies, particularly in oncology. We conducted structured searches in PubMed, Google Scholar, and SciFinder (2013-2024) using controlled vocabulary, including "saponins," "macrophage polarization," and "therapeutic effects." Our findings demonstrate that saponins significantly modulate immune responses and improve treatment efficacy. However, clinical translation is hindered by challenges such as poor bioavailability and safety concerns, which limit systemic exposure and therapeutic utility. To overcome these barriers, innovative delivery strategies, including nanoemulsions and engineered exosomes, are essential for enhancing pharmacokinetics and therapeutic index. Future research should prioritize elucidating the molecular mechanisms underlying saponin-mediated macrophage polarization, identifying novel therapeutic targets, and optimizing drug formulations. Addressing these challenges will enable the restoration of immune balance and more effective management of diverse diseases.
Collapse
Affiliation(s)
- Beibei Xiong
- Department of Oncology, The First People’s Hospital of Shuangliu District, Chengdu, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi-Xuan Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen-Ying Lan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | - Fang Wang
- Chengdu First People’s Hospital, Chengdu, China
| |
Collapse
|
2
|
Kaylan KB, Nargis T, Figatner K, Wang JE, Pratuangtham S, Chakraborty A, Casimiro I, Nadler JL, Boxer MB, Maloney DJ, Anderson RM, Mirmira RG, Tersey SA. 12-Lipoxygenase Inhibition Improves Glycemia and Obesity-associated Inflammation in Male Human Gene Replacement Mice. Endocrinology 2025; 166:bqaf069. [PMID: 40186458 PMCID: PMC12022223 DOI: 10.1210/endocr/bqaf069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 04/07/2025]
Abstract
Obesity-associated inflammation is characterized by macrophage infiltration into peripheral tissues, contributing to the progression of prediabetes and type 2 diabetes. 12-lipoxygenase (12-LOX) catalyzes the formation of pro-inflammatory eicosanoids and promotes the migration of macrophages, yet its role in obesity-associated inflammation remains incompletely understood. Furthermore, differences between mouse and human orthologs of 12-LOX have limited efforts to study existing pharmacologic inhibitors of 12-LOX. In this study, we used a human gene replacement mouse model in which the gene encoding mouse 12-LOX (Alox15) is replaced by the human ALOX12 gene. As a model of obesity and dysglycemia, we administered male mice a high-fat diet. We subsequently investigated the effects of VLX-1005, a potent and selective small molecule inhibitor of human 12-LOX. Oral administration of VLX-1005 resulted in improved glucose homeostasis, decreased β-cell dedifferentiation, and reduced macrophage infiltration in islets and adipose tissue. Analysis of the stromal vascular fraction from adipose tissue showed a reduction in myeloid cells and cytokine expression with VLX-1005 treatment, indicating decreased adipose tissue inflammation. In a distinct mouse model in which Alox15 was selectively deleted in myeloid cells, we observed decreased β-cell dedifferentiation and reduced macrophage infiltration in both islets and adipose tissue, suggesting that the effects of VLX-1005 may relate to the inhibition of 12-LOX in macrophages. These findings highlight 12-LOX as a key factor in obesity-associated inflammation and suggest that 12-LOX inhibition could serve as a therapeutic strategy to improve glucose homeostasis and peripheral inflammation in the setting of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Kerim B Kaylan
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Titli Nargis
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Kayla Figatner
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jiayi E Wang
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarida Pratuangtham
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Advaita Chakraborty
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Isabel Casimiro
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jerry L Nadler
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | - Ryan M Anderson
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Raghavendra G Mirmira
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah A Tersey
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
3
|
Ho KT, Chu FY, Lin YK, Chin HH, Yang SC, Yang CP, Chang YH. Interleukin-4 ameliorates macrophage lipid stress through promoting cholesterol efflux and lipid homeostasis. Cytokine 2025; 188:156869. [PMID: 39954486 DOI: 10.1016/j.cyto.2025.156869] [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: 09/25/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 02/17/2025]
Abstract
Over-nutrition and lipid metabolic abnormalities are correlated with obesity and type 2 diabetes mellitus (T2DM). Individuals with long-term hyperglycemia and dyslipidemia are susceptible to life-threatening complications such as atherosclerosis. Excess amounts of modified low density lipoprotein (mLDL) attract circulating monocytes to resident at arterial wall and differentiate into pro-inflammatory M1 macrophages. M1 cells uptake mLDL through scavenger receptors-mediated endocytosis, leading to increased lipids influx, cholesterol accumulation and foam cell formation. Besides, macrophages are attracted and infiltrated into the hypertrophic adipose tissue to mediate microenvironmental lipid metabolism. Our previous studies demonstrate that anti-inflammatory interleukin-4 (IL-4) regulates lipid metabolism by inhibiting lipid accumulation and promoting lipolysis of mature adipocytes. The effects of IL-4-polarized M2 macrophages on 3T3-L1 adipogenesis and macrophage-adipocyte interaction were explored in the present study. Our results showed lipid deposits and lipid droplets (LDs)-anchored perilipin of adipocytes cultured in IL-4-polarized M2-conditioned medium (M2-CM) were decreased, while adipogenesis-driving transcription factors and critical lipid metabolic enzymes remained unaffected. It indicates that M2-secreted mediators down-regulate lipid deposits and LDs formation in late adipogenic phase rather than interfering early programming phase and lipid synthesis machinery. In addition, IL-4 reduced intracellular lipid loads by up-regulating cholesterol efflux ATP-binding cassette transporter A1 (ABCA1) and ABCG1 despite cholesterol influx CD36 was also elevated. Accordingly, IL-4 shows beneficial effects to prevent atherosclerosis via promoting catabolism of the internalized lipids and cholesterol efflux, thus efficiently reduces lipid overload and foam cell formation. These findings illustrate novel roles and protective function of IL-4 to reduce the risk of atherosclerosis incidence by efficiently promoting macrophage cholesterol efflux and lipid homeostasis.
Collapse
Affiliation(s)
- Kuo-Ting Ho
- Center for Precision Medicine, Yi-He Hospital, Quanzhou, Fujian Province, PR China; HI. Q Biomedical Laboratory, Takyun Industrial Park, Quanzhou, Fujian Province, PR China
| | - Fang-Yeh Chu
- Department of Clinical Pathology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320, Taiwan; Department of Medical Laboratory Science and Biotechnology, Yuanpei University 300, Taiwan; School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei 110, Taiwan
| | - Yi-Kai Lin
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ho-Hsun Chin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan; Laboratory Accreditation Department II, Taiwan Accreditation Foundation, Hsinchu, Taiwan
| | - Shun-Chun Yang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan; Department of Clinical Pathology, Min-Sheng General Hospital, Taoyuan 320, Taiwan
| | - Ching-Ping Yang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yih-Hsin Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| |
Collapse
|
4
|
Leinweber B, Pilorz V, Olejniczak I, Skrum L, Begemann K, Heyde I, Stenger S, Sadik CD, Oster H. Bmal1 deficiency in neutrophils alleviates symptoms induced by high-fat diet. iScience 2025; 28:112038. [PMID: 40124497 PMCID: PMC11930374 DOI: 10.1016/j.isci.2025.112038] [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: 08/24/2024] [Revised: 12/24/2024] [Accepted: 02/12/2025] [Indexed: 03/25/2025] Open
Abstract
Physiological processes, including metabolism and immune responses, are generated by the circadian clock, driven by clock genes. Disrupting circadian rhythms through a high-fat diet promotes obesity and inflammation. Studies show that deleting the clock gene, brain, and muscle ARNT-like 1 (Bmal1) in adipose tissue leads to overeating and weight gain. We now show that Bmal1 deletion in neutrophils protects against diet-induced obesity and reduces inflammatory macrophage infiltration into epididymal white adipose tissue (eWAT), despite increased food intake over 20 weeks of a high-fat diet. This protection is linked to enhanced energy expenditure, increased UCP1 expression in iBAT, improved insulin sensitivity, and altered expression of genes encoding chemokine receptors CXCR2, CXCR4, and the ligand Cxcl2 in eWAT. Our findings reveal a key role of Bmal1 in neutrophils in regulating high-fat diet-induced adipose inflammation and emphasize circadian regulation's importance in immuno-metabolic function.
Collapse
Affiliation(s)
- Brinja Leinweber
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Violetta Pilorz
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Iwona Olejniczak
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Ludmila Skrum
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Kimberly Begemann
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Isabel Heyde
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Sarah Stenger
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Christian David Sadik
- University of Lübeck, Department of Dermatology, Allergy, and Venereology Ratzeburger Allee, 23562 Luebeck, Germany
| | - Henrik Oster
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| |
Collapse
|
5
|
Banerjee D, Patra D, Sinha A, Chakrabarty D, Patra A, Sarmah R, Dey U, Dutta R, Bhagabati SK, Mukherjee AK, Kumar A, Pal D, Dasgupta S. Macrophage foam cell-derived mediator promotes spontaneous fat lipolysis in atherosclerosis models. J Leukoc Biol 2025; 117:qiae210. [PMID: 39509245 DOI: 10.1093/jleuko/qiae210] [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: 07/28/2024] [Revised: 09/03/2024] [Indexed: 11/15/2024] Open
Abstract
Ectopic lipid accumulation in macrophages is responsible for the formation of macrophage foam cells (MFCs) which are involved in the crosstalk with the perivascular adipose tissue (PVAT) of the vascular wall that plays a pivotal role in the progression of atherosclerosis. However, the interrelationship between MFCs and PVAT implementing adipocyte dysfunction during atherosclerosis has not yet been established. We hypothesized that MFC-secreted mediator(s) is causally linked with PVAT dysfunction and the succession of atherosclerosis. To test this hypothesis, MFCs were cocultured with adipocytes, or the conditional media of MFCs (MFC-CM) were exposed to adipocytes and found a significant induction of fat lipolysis in adipocytes. The molecular filtration followed by the high-performance liquid chromatography (HPLC) fractionation and liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) analysis of MFC-CM revealed a novel mediator fetuin-A (FetA) that significantly augments toll-like receptor 4 (TLR4)-dependent fat lipolysis in adipocytes. Mechanistically, MFC-derived FetA markedly increased TLR4-dependent c-Jun N-terminal kinases (JNK)/extracellular signal-regulated kinases (ERK) activation that causes spontaneous fat lipolysis implementing adipocyte dysfunction. Thus, the present study provides the first evidence of MFC-derived FetA that induces adipocyte dysfunction by the stimulation of spontaneous fat lipolysis. Therefore, targeting the crosstalk between MFCs and adipocytes could be a newer approach to counter the progression of atherosclerosis.
Collapse
Affiliation(s)
- Dipanjan Banerjee
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Debarun Patra
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar 140001, Punjab, India
| | - Archana Sinha
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Dwaipayan Chakrabarty
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Aparup Patra
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Raktim Sarmah
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon 782103, Assam, India
| | - Upalabdha Dey
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Rajdeep Dutta
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon 782103, Assam, India
| | - Sarada K Bhagabati
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon 782103, Assam, India
| | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
- Division of Life Sciences, Institute of Advanced Studies in Science and Technology, Vigyan Path, Garchuk, Paschim Boragaon, Guwahati 781035, Assam, India
| | - Aditya Kumar
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar 140001, Punjab, India
| | - Suman Dasgupta
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India
| |
Collapse
|
6
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
7
|
Shrestha S, Jeon JH, Hong CW. Neutrophils in MASLD and MASH. BMB Rep 2025; 58:116-123. [PMID: 39757200 PMCID: PMC11955729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/23/2024] [Accepted: 06/11/2024] [Indexed: 01/07/2025] Open
Abstract
Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD) and its progressive form, Metabolic Dysfunction Associated Steatohepatitis (MASH), represent significant health concerns associated with the metabolic syndrome. These conditions are characterized by excessive hepatic fat accumulation, inflammation, and potential progression to cirrhosis and hepatocellular carcinoma. Neutrophils are innate immune cells that play a pivotal role in the development of MASLD and MASH. They can infiltrate the hepatic microenvironment in response to inflammatory cytokines and damage associated molecular patterns (DAMPs) derived from the liver and exacerbate tissue damage by releasing of reactive oxygen species (ROS), cytokines, and neutrophil extracellular traps (NETs). Moreover, neutrophils can disrupt the metabolism of hepatocytes through key factors such as neutrophil elastase (NE) and human neutrophil peptides-1 (HNP-1), leading to inflammation and fibrosis, while myeloperoxidase (MPO) and lipocalin (LCN2) are involved in inflammatory and fibrotic processes. In contrast, neutrophils contribute to liver protection and repair through mechanisms involving microRNA-223 and matrix metalloproteinase 9 (MMP9). This dual role of neutrophils highlights their significance in the pathogenesis of MASLD and MASH. This review summarizes current understanding from recent studies on the involvement of neutrophils in MASLD and MASH. Understanding complex roles of neutrophils within the liver's unique microenvironment offers insights into novel therapeutic strategies, emphasizing the need for further research to explore neutrophil-targeted interventions for managing MASLD and MASH. [BMB Reports 2025; 58(3): 116-123].
Collapse
Affiliation(s)
- Sanjeeb Shrestha
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41404, Korea
| | - Chang-Won Hong
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Cell & Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
8
|
Knights AJ, Farrell EC, Ellis OM, Song MJ, Appleton CT, Maerz T. Synovial macrophage diversity and activation of M-CSF signaling in post-traumatic osteoarthritis. eLife 2025; 12:RP93283. [PMID: 39969512 PMCID: PMC11839164 DOI: 10.7554/elife.93283] [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] [Indexed: 02/20/2025] Open
Abstract
Synovium is home to immune and stromal cell types that orchestrate inflammation following a joint injury; in particular, macrophages are central protagonists in this process. We sought to define the cellular and temporal dynamics of the synovial immune niche in a mouse model of post-traumatic osteoarthritis (PTOA), and to identify stromal-immune crosstalk mechanisms that coordinate macrophage function and phenotype. We induced PTOA in mice using a non-invasive tibial compression model of anterior cruciate ligament rupture (ACLR). Single-cell RNA-sequencing and flow cytometry were used to assess immune cell populations in healthy (Sham) and injured (7 and 28 days post-ACLR) synovium. Characterization of synovial macrophage polarization states was performed, alongside computational modeling of macrophage differentiation, as well as implicated transcriptional regulators and stromal-immune communication axes. Immune cell types are broadly represented in healthy synovium, but experience drastic expansion and speciation in PTOA, most notably in the macrophage portion. We identified several polarization states of macrophages in synovium following joint injury, underpinned by distinct transcriptomic signatures, and regulated in part by stromal-derived macrophage colony-stimulating factor signaling. The transcription factors Pu.1, Cebpα, Cebpβ, and Jun were predicted to control differentiation of systemically derived monocytes into pro-inflammatory synovial macrophages. In summary, we defined different synovial macrophage subpopulations present in healthy and injured mouse synovium. Nuanced characterization of the distinct functions, origins, and disease kinetics of macrophage subtypes in PTOA will be critical for targeting these highly versatile cells for therapeutic purposes.
Collapse
Affiliation(s)
- Alexander J Knights
- Department of Orthopaedic Surgery, University of MichiganAnn ArborUnited States
| | - Easton C Farrell
- Department of Orthopaedic Surgery, University of MichiganAnn ArborUnited States
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
| | - Olivia M Ellis
- Department of Orthopaedic Surgery, University of MichiganAnn ArborUnited States
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
| | - Michelle J Song
- Department of Orthopaedic Surgery, University of MichiganAnn ArborUnited States
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
| | - C Thomas Appleton
- Department of Physiology and Pharmacology, Western UniversityLondonCanada
- Bone and Joint Institute, Western UniversityLondonCanada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western UniversityLondonCanada
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of MichiganAnn ArborUnited States
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Department of Internal Medicine – Division of Rheumatology, University of MichiganAnn ArborUnited States
| |
Collapse
|
9
|
Yildiz R, Ganbold K, Sparman NZR, Rajbhandari P. Immune Regulatory Crosstalk in Adipose Tissue Thermogenesis. Compr Physiol 2025; 15:e70001. [PMID: 39921241 DOI: 10.1002/cph4.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Brown adipose tissue (BAT) and thermogenic beige fat within white adipose tissue (WAT), collectively known as adaptive thermogenic fat, dissipate energy as heat, offering promising therapeutic potential to combat obesity and metabolic disorders. The specific biological functions of these fat depots are determined by their unique interaction with the microenvironments, composed of immune cells, endothelial cells, pericytes, and nerve fibers. Immune cells residing in these depots play a key role in regulating energy expenditure and systemic energy homeostasis. The dynamic microenvironment of thermogenic fat depots is essential for maintaining tissue health and function. Immune cells infiltrate both BAT and beige WAT, contributing to their homeostasis and activation through intricate cellular communications. Emerging evidence underscores the importance of various immune cell populations in regulating thermogenic adipose tissue, though many remain undercharacterized. This review provides a comprehensive overview of the immune cells that regulate adaptive thermogenesis and their complex interactions within the adipose niche, highlighting their potential to influence metabolic health and contribute to therapeutic interventions for obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Ramazan Yildiz
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Khatanzul Ganbold
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Njeri Z R Sparman
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
10
|
Avery D, Morandini L, Sheakley L, Alajmi A, Bergey L, Donahue HJ, Martin RK, Olivares-Navarrete R. Obesity prolongs the pro-inflammatory response and attenuates bone healing on titanium implants. Acta Biomater 2025; 192:473-486. [PMID: 39586347 PMCID: PMC11735295 DOI: 10.1016/j.actbio.2024.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
Obesity is a metabolic disease resulting from excess body fat accumulation associated with chronic systemic inflammation. Obesity has been shown to impact the function and activity of neutrophils, macrophages, and T cells, contributing to higher circulating levels of pro-inflammatory cytokines. Biomaterial surface properties such as roughness and hydrophilicity can influence the behavior of immune cells in the peri-implant microenvironment. This study aimed to determine how obesity induced by a high-fat diet (HFD) affects the inflammatory response to modified titanium (Ti) implants and subsequent bone formation. Obese mice had significantly more neutrophils, pro-inflammatory macrophages, and T cells and fewer anti-inflammatory macrophages and mesenchymal stem cells (MSCs) in the peri-implant tissue than lean mice. Obesity also increased circulating adipokines and pro-inflammatory cytokines when compared to lean animals. Bone formation around Ti implants was reduced in obese mice compared to controls. Adoptive transfer of bone marrow cells isolated from obese mice into wild-type mice demonstrated the localized impact of obesity on immune cell function and phenotype, promoting a pro-inflammatory peri-implant microenvironment and attenuating bone formation post-implantation. These results show that obesity significantly affects the inflammatory response to modified Ti implants, prolonging the pro-inflammatory response to the implanted biomaterial and compromising bone formation. STATEMENT OF SIGNIFICANCE: Obesity has been shown to significantly alter physiological processes, including the behavior of immune cells, inducing a state of systemic chronic inflammation. Our study demonstrates that obesity-induced via a high-fat diet alters immune cell response to implanted biomaterials, with increased pro-inflammatory response and attenuated immunomodulation that results in decreased biomaterial integration.
Collapse
Affiliation(s)
- Derek Avery
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Lais Morandini
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Luke Sheakley
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Asmaa Alajmi
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Leah Bergey
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Henry J Donahue
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States
| | - Rebecca K Martin
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
11
|
Kaylan KB, Nargis T, Figatner K, Wang JE, Pratuangtham S, Chakraborty A, Casimiro I, Nadler JL, Boxer MB, Maloney DJ, Anderson RM, Mirmira RG, Tersey SA. 12-Lipoxygenase inhibition improves glucose homeostasis and obesity-associated inflammation in human gene replacement mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632274. [PMID: 39868153 PMCID: PMC11761697 DOI: 10.1101/2025.01.10.632274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Obesity-associated inflammation is characterized by macrophage infiltration into peripheral tissues, contributing to the progression of prediabetes and type 2 diabetes (T2D). The enzyme 12-lipoxygenase (12-LOX) catalyzes the formation of pro-inflammatory eicosanoids and is known to promote the migration of macrophages, yet its role in obesity-associated inflammation remains incompletely understood. Furthermore, differences between mouse and human orthologs of 12-LOX have limited efforts to study existing pharmacologic inhibitors of 12-LOX. In this study, we utilized a human gene replacement mouse model in which the gene encoding mouse 12-LOX (Alox15) is replaced by the human ALOX12 gene. As a model of obesity and dysglycemia, we administered these mice a high-fat diet. We subsequently investigated the effects of VLX-1005, a potent and selective small molecule inhibitor of human 12-LOX. Oral administration of VLX-1005 resulted in improved glucose homeostasis, decreased β cell dedifferentiation, and reduced macrophage infiltration in islets and adipose tissue. Analysis of the stromal vascular fraction from adipose tissue showed a reduction in myeloid cells and cytokine expression with VLX-1005 treatment, indicating decreased adipose tissue inflammation. In a distinct mouse model in which Alox15 was selectively deleted in myeloid cells, we observed decreased β cell dedifferentiation and reduced macrophage infiltration in both islets and adipose tissue, suggesting that the effects of VLX-1005 may relate to the inhibition of 12-LOX in macrophages. These findings highlight 12-LOX as a key factor in obesity-associated inflammation and suggest that 12-LOX inhibition could serve as a therapeutic strategy to improve glucose homeostasis and peripheral inflammation in the setting of obesity and T2D.
Collapse
Affiliation(s)
- Kerim B. Kaylan
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Titli Nargis
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Kayla Figatner
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jiayi E. Wang
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarida Pratuangtham
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Advaita Chakraborty
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Isabel Casimiro
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jerry L. Nadler
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | - Ryan M. Anderson
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Raghavendra G. Mirmira
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah A. Tersey
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
12
|
Lempicki MD, Garrigues RJ, Zeczycki TN, Garcia BL, Harris TE, Meher AK. Matrix Metalloproteinase-2 as a novel regulator of glucose utilization by adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.09.626845. [PMID: 39713289 PMCID: PMC11661172 DOI: 10.1101/2024.12.09.626845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Glucose transporter 4 (GLUT4) expression on white adipocytes is critical for absorbing excess blood glucose, failure of which promotes hyperglycemia. Matrix metalloproteinases (MMPs) play a crucial role in remodeling the white adipose tissue (WAT) during obesity. MMPs have multiple protein substrates, and surprisingly, it is unknown if they can directly target GLUT4 on the adipocyte surface and impair glucose absorption. We identified MMP2 as the highly active gelatinase, a class of MMP, in the gonadal WAT of high-fat diet-induced obese mice. In vitro, metabolic studies in 3T3-L1 adipocytes revealed MMP2 attenuated glucose absorption and glycolysis, which were recovered by an MMP2 inhibitor. In silico structural analysis using AlphaFold identified a putative MMP2 cleavage site on the extracellular domain of GLUT4. Further, in a substrate competition assay, a peptide mimicking the MMP2 cleavage motif on GLUT4 attenuated the cleavage of an MMP substrate by MMP2. Altogether, our results suggest a novel mechanism of impaired glucose absorption by adipocytes, which may contribute to hyperglycemia during obesity.
Collapse
Affiliation(s)
- Melissa D. Lempicki
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Ryan J. Garrigues
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Tonya N. Zeczycki
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Brandon L. Garcia
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, United States
| | - Thurl E. Harris
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903, United States
| | - Akshaya K. Meher
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| |
Collapse
|
13
|
Li J, Tian Z, Zhang T, Jin J, Zhang X, Xie P, Lin H, Gu J, Wu Y, Wang X, Zhang S, Yan X, Guo D, Wang Z, Zhang Q. Single-cell view and a novel protective macrophage subset in perivascular adipose tissue in T2DM. Cell Mol Biol Lett 2024; 29:148. [PMID: 39627688 PMCID: PMC11616190 DOI: 10.1186/s11658-024-00668-5] [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: 04/11/2024] [Accepted: 11/14/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Vasculopathy underlies diabetic complications, with perivascular adipose tissue (PVAT) playing crucial roles in its development. However, the changes in the cellular composition and function of PVAT, including the specific cell subsets and mechanisms implicated in type 2 diabetes mellitus (T2DM) vasculopathy, remain unclear. METHODS To address the above issues, we performed single-cell RNA sequencing on the stromal vascular fraction (SVF) of PVAT from normal and T2DM rats. Then, various bioinformatics tools and functional experiments were used to investigate the characteristic changes in the cellular profile of diabetic PVAT SVF, their implications, and the underlying mechanisms. RESULTS Our study reveals the single-cell landscape of the SVF of PVAT, demonstrating its considerable heterogeneity and significant alterations in T2DM, including an enhanced inflammatory response and elevated proportions of macrophages and natural killer (NK) cells. Moreover, macrophages are critical hubs for cross-talk among various cell populations. Notably, we identified a decreased Pdpn+ macrophage subpopulation in the PVAT of T2DM rats and confirmed this in mice and humans. In vitro and in vivo studies demonstrated that Pdpn+ macrophages alleviated insulin resistance and modulated adipokine/cytokine expression in adipocytes via the Pla2g2d-DHA/EPA-GPR120 pathway. This subset also enhances the function of vascular endothelial and smooth muscle cells, inhibits vascular inflammation and oxidative stress, and improves vasodilatory function, thereby protecting blood vessels. CONCLUSION Pdpn+ macrophages exhibit significant vascular protective effects by alleviating insulin resistance and modulating adipokine/cytokine expression in PVAT adipocytes. This macrophage subtype may therefore play pivotal roles in mitigating vascular complications in T2DM. Our findings also underscore the critical role of immune-metabolic cross-talk in maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Jiaxuan Li
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
- Shandong Provincial Hospital, Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, 250021, China
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Chinese Ministry of Education, Shandong First Medical University, Jinan, 250021, China
| | - Zhenyu Tian
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Tongxue Zhang
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Jiajia Jin
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xinjie Zhang
- Department of Biology, University College London, London, NW1 2HE, UK
| | - Panpan Xie
- Department of Breast and Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, 252000, China
| | - Haiyan Lin
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Junfei Gu
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yingjie Wu
- Shandong Provincial Hospital, Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, 250021, China
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Chinese Ministry of Education, Shandong First Medical University, Jinan, 250021, China
| | - Xiaowei Wang
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Shucui Zhang
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xuefang Yan
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Dong Guo
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, 252000, China.
| | - Zhe Wang
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| | - Qunye Zhang
- Department of Cardiology, State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
14
|
Yang Y, Huang B, Qin Y, Wang D, Jin Y, Su L, Wang Q, Pan Y, Zhang Y, Shen Y, Hu W, Cao Z, Jin L, Zhang F. Adipocyte microRNA-802 promotes adipose tissue inflammation and insulin resistance by modulating macrophages in obesity. eLife 2024; 13:e99162. [PMID: 39589393 DOI: 10.7554/elife.99162] [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: 04/26/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024] Open
Abstract
Adipose tissue inflammation is now considered to be a key process underlying metabolic diseases in obese individuals. However, it remains unclear how adipose inflammation is initiated and maintained or the mechanism by which inflammation develops. We found that microRNA-802 (Mir802) expression in adipose tissue is progressively increased with the development of dietary obesity in obese mice and humans. The increasing trend of Mir802 preceded the accumulation of macrophages. Adipose tissue-specific knockout of Mir802 lowered macrophage infiltration and ameliorated systemic insulin resistance. Conversely, the specific overexpression of Mir802 in adipose tissue aggravated adipose inflammation in mice fed a high-fat diet. Mechanistically, Mir802 activates noncanonical and canonical NF-κB pathways by targeting its negative regulator, TRAF3. Next, NF-κB orchestrated the expression of chemokines and SREBP1, leading to strong recruitment and M1-like polarization of macrophages. Our findings indicate that Mir802 endows adipose tissue with the ability to recruit and polarize macrophages, which underscores Mir802 as an innovative and attractive candidate for miRNA-based immune therapy for adipose inflammation.
Collapse
Affiliation(s)
- Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Bin Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yimeng Qin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Danwei Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yinuo Jin
- NanJing HanKai Academy, Nanjing, China
| | - Linmin Su
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qingxin Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yumeng Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wenjun Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhengyu Cao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangfang Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
15
|
Ding L, Lu Z, Jiang X, Zhang S, Tian X, Wang Q. Obesity-derived macrophages upregulate TNF-α to induce apoptosis in glial cell via the NF-κB/PHLPP1 axis. Int Immunopharmacol 2024; 141:112962. [PMID: 39197294 DOI: 10.1016/j.intimp.2024.112962] [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: 06/03/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
Macrophages in obese adipose tissue have been shown to damage nerve fibers, however, the mechanism underlying how macrophages cause glial cell damage remains unknown. This study aimed to characterize the mechanism by which macrophages induce apoptosis in glial cell during obesity formation in mice by single-nucleus RNA sequencing (snRNA-seq). Cells obtained from paraepididymal adipose tissue in obese mice underwent snRNA-seq. Eighteen different clusters were identified, and 12 cell types were annotated, including glial cells, macrophages, and fibroblasts. There was a negative correlation between the number of glial cells and macrophages in mouse adipose tissue during the formation of obesity. The pro-apoptotic factor PHLPP1 was identified in GO Terms. The interaction between adipose tissue glial cells and macrophages was revealed via in-depth analysis, and the cell-cell communication mechanism between the TNF-α and NF-KB/PHLPP1 axes was perfected. Apoptosis of glial cell by upregulation of TNF-α via obesity-derived macrophages and activation of the NF-κB/PHLPP1 axis. We further revealed how macrophages induce apoptosis in glial cells during obesity formation, as well as different changes in the two cell populations. This study provides valuable resources and foundations for understanding the mechanistic effects of macrophages and glial cells during obesity formation, as well as diseases and potential interventions.
Collapse
Affiliation(s)
- Ling Ding
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Zhimin Lu
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Xing Jiang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Sen Zhang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Xuewen Tian
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China.
| | - Qinglu Wang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China.
| |
Collapse
|
16
|
Miracle CE, McCallister CL, Egleton RD, Salisbury TB. Mechanisms by which obesity regulates inflammation and anti-tumor immunity in cancer. Biochem Biophys Res Commun 2024; 733:150437. [PMID: 39074412 PMCID: PMC11455618 DOI: 10.1016/j.bbrc.2024.150437] [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: 05/10/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Obesity is associated with an increased risk for 13 different cancers. The increased risk for cancer in obesity is mediated by obesity-associated changes in the immune system. Obesity has distinct effects on different types of inflammation that are tied to tumorigenesis. For example, obesity promotes chronic inflammation in adipose tissue that is tumor-promoting in peripheral tissues. Conversely, obesity inhibits acute inflammation that rejects tumors. Obesity therefore promotes cancer by differentially regulating chronic versus acute inflammation. Given that obesity is chronic, the initial inflammation in adipose tissue will lead to systemic inflammation that could induce compensatory anti-inflammatory reactions in peripheral tissues to suppress chronic inflammation. The overall effect of obesity in peripheral tissues is therefore dependent on the duration and severity of obesity. Adipose tissue is a complex tissue that is composed of many cell types in addition to adipocytes. Further, adipose tissue cellularity is different at different anatomical sites throughout the body. Consequently, the sensitivity of adipose tissue to obesity is dependent on the anatomical location of the adipose depot. For example, obesity induces more inflammation in visceral than subcutaneous adipose tissue. Based on these studies, the mechanisms by which obesity promotes tumorigenesis are multifactorial and immune cell type-specific. The objective of our paper is to discuss the cellular mechanisms by which obesity promotes tumorigenesis by regulating distinct types of inflammation in adipose tissue and the tumor microenvironment.
Collapse
Affiliation(s)
- Cora E Miracle
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Chelsea L McCallister
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Richard D Egleton
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Travis B Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| |
Collapse
|
17
|
Zhu Z, Yu M, Xu M, Ji X, Zong X, Zhang Z, Shang W, Zhang L, Fang P. Baicalin suppresses macrophage JNK-mediated adipose tissue inflammation to mitigate insulin resistance in obesity. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118355. [PMID: 38762213 DOI: 10.1016/j.jep.2024.118355] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/05/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix scutellariae (the root of Scutellaria baicalensis Georgi) is a traditional Chinese medicine (TCM) used to treat a wide range of inflammation-related diseases, such as obesity, diabetes, diabetic kidney disease, and COVID-19-associated inflammatory states in the lung and kidney. Baicalin is the major anti-inflammatory component of Radix scutellariae and has shown the potential to inhibit inflammation in metabolic disorders. In this study, we explored the ability and underlying mechanisms of baicalin to modulate the macrophage to mitigate insulin resistance in obesity. MATERIALS AND METHODS Obese mice were administered baicalin (50 mg/kg/day) intraperitoneally for 3 weeks. RAW264.7 and BMDM cells were stimulated with LPS and treated with baicalin for 24 h, while 3T3-L1 and primary white adipocytes were treated with the supernatants from baicalin-treated RAW264.7 cells for 24 h. RESULTS The results showed that baicalin significantly improved glucose and insulin tolerance as well as decreased fat and adipose tissue macrophage levels in obese mice. Besides, baicalin significantly reduced serum and adipose tissue IL-1β, TNF-α and IL-6 levels in obese mice, as well as suppressed LPS-induced IL-1β, TNF-α and IL-6 expression and release in macrophages. Furthermore, treatment with the supernatant from baicalin-treated RAW264.7 cells increased the levels of PGC-1α, SIRT1, p-IRS-1 and p-AKT in adipocytes. Moreover, baicalin treatment dramatically downregulated macrophage p-p38, p-JNK, and Ac-p65Lys310 levels while increasing SIRT1 both in vivo and in vitro. Importantly, JNK inhibitor SP600125 blocked most of the effects of baicalin on SIRT1, Ac-p65Lys310 and pro-inflammatory factors in macrophages. CONCLUSION Therefore, these results demonstrated for the first time that baicalin exerts its anti-inflammatory effects in obese adipose tissue macrophages mainly through suppressing JNK/SIRT1/p65 signaling. These findings amplified the mechanisms of baicalin and its potential to attenuate insulin resistance.
Collapse
Affiliation(s)
- Ziyue Zhu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mei Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mengfan Xu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xin Ji
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Xicui Zong
- Hanlin College, Nanjing University of Chinese Medicine, Taizhou, 225300, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China.
| | - Wenbin Shang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Li Zhang
- Hanlin College, Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
18
|
Lin C, Chen Y, Ge Y, Niu H, Zhang X, Jiang F, Wu C. A Bibliometric and Knowledge-Map Analysis of Macrophage Polarization in Insulin Resistance From 1999 to 2023. Immun Inflamm Dis 2024; 12:e70048. [PMID: 39465505 PMCID: PMC11513609 DOI: 10.1002/iid3.70048] [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: 03/28/2024] [Revised: 07/14/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Despite numerous studies confirming the association between insulin resistance (IR) and macrophage polarization, there is a lack of bibliometric analysis in this area. Therefore, our objective is to conduct a comprehensive analysis of published literature and identify potential future research trends using bibliometrics. METHOD Publications on the topic of macrophage polarization in IR were gathered from the Web of Science Core Collection database (WoSCC) spanning the years 1999-2023. Bibliometric analysis and visualization were conducted using VOSviewers, CiteSpace, the R package "bibliometrix" and Tableau Public. RESULT A total of 3435 articles published between 1999 and 2023 were included in the analysis. These articles originated from 75 countries, with the United States and China leading in contributions. The top five research institutions are the University of California, San Diego, Harvard University, the University of Michigan, Shanghai Jiao Tong University, and Huazhong University of Science and Technology. In this research domain, Diabetes is the most frequently published journal, and the Journal of Clinical Investigation is the most co-cited. Among the 19,398 authors contributing to these publications, Lumeng CN. not only authored the most papers but also received the highest number of co-citations. "Insulin resistance" emerges as a primary keyword in the analysis of emerging research hotspots. CONCLUSION For the first time, bibliometric methods have been employed to conduct a comprehensive summary of papers relevant to macrophage polarization in IR. This study aims to identify the current research direction and future research hotspots, offering valuable guidance and insights for scholars in the field.
Collapse
Affiliation(s)
- Chuning Lin
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Yuan Chen
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Yao Ge
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Huimin Niu
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Xinyi Zhang
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Feng Jiang
- Department of NeonatologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiJiangsu ProvinceChina
| | - Chuyan Wu
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| |
Collapse
|
19
|
Soedono S, Julietta V, Nawaz H, Cho KW. Dynamic Roles and Expanding Diversity of Adipose Tissue Macrophages in Obesity. J Obes Metab Syndr 2024; 33:193-212. [PMID: 39324219 PMCID: PMC11443328 DOI: 10.7570/jomes24030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024] Open
Abstract
Adipose tissue macrophages (ATMs) are key regulators of adipose tissue (AT) inflammation and insulin resistance in obesity, and the traditional M1/M2 characterization of ATMs is inadequate for capturing their diversity in obese conditions. Single-cell transcriptomic profiling has revealed heterogeneity among ATMs that goes beyond the old paradigm and identified new subsets with unique functions. Furthermore, explorations of their developmental origins suggest that multiple differentiation pathways contribute to ATM variety. These advances raise concerns about how to define ATM functions, how they are regulated, and how they orchestrate changes in AT. This review provides an overview of the current understanding of ATMs and their updated categorization in both mice and humans during obesity. Additionally, diverse ATM functions and contributions in the context of obesity are discussed. Finally, potential strategies for targeting ATM functions as therapeutic interventions for obesity-induced metabolic diseases are addressed.
Collapse
Affiliation(s)
- Shindy Soedono
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Vivi Julietta
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Hadia Nawaz
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| |
Collapse
|
20
|
Yuzbashian E, Berg E, de Campos Zani SC, Chan CB. Cow's Milk Bioactive Molecules in the Regulation of Glucose Homeostasis in Human and Animal Studies. Foods 2024; 13:2837. [PMID: 39272602 PMCID: PMC11395457 DOI: 10.3390/foods13172837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Obesity disrupts glucose metabolism, leading to insulin resistance (IR) and cardiometabolic diseases. Consumption of cow's milk and other dairy products may influence glucose metabolism. Within the complex matrix of cow's milk, various carbohydrates, lipids, and peptides act as bioactive molecules to alter human metabolism. Here, we summarize data from human studies and rodent experiments illustrating how these bioactive molecules regulate insulin and glucose homeostasis, supplemented with in vitro studies of the mechanisms behind their effects. Bioactive carbohydrates, including lactose, galactose, and oligosaccharides, generally reduce hyperglycemia, possibly by preventing gut microbiota dysbiosis. Milk-derived lipids of the milk fat globular membrane improve activation of insulin signaling pathways in animal trials but seem to have little impact on glycemia in human studies. However, other lipids produced by ruminants, including polar lipids, odd-chain, trans-, and branched-chain fatty acids, produce neutral or contradictory effects on glucose metabolism. Bioactive peptides derived from whey and casein may exert their effects both directly through their insulinotropic effects or renin-angiotensin-aldosterone system inhibition and indirectly by the regulation of incretin hormones. Overall, the results bolster many observational studies in humans and suggest that cow's milk intake reduces the risk of, and can perhaps be used in treating, metabolic disorders. However, the mechanisms of action for most bioactive compounds in milk are still largely undiscovered.
Collapse
Affiliation(s)
- Emad Yuzbashian
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Emily Berg
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | | | - Catherine B Chan
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
21
|
Anwar C, Lin JR, Tsai ML, Ho CT, Lai CS. Calebin A attenuated inflammation in RAW264.7 macrophages and adipose tissue to improve hepatic glucose metabolism and hyperglycemia in high-fat diet-fed obese mice. Eur J Pharmacol 2024; 978:176789. [PMID: 38945287 DOI: 10.1016/j.ejphar.2024.176789] [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: 04/03/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
The increased incidence of obesity, which become a global health problem, requires more functional food products with minor side and excellent effects. Calebin A (CbA) is a non-curcuminoid compound, which is reported to be an effective treatment for lipid metabolism and thermogenesis. However, its ability and mechanism of action in improving obesity-associated hyperglycemia remain unclear. This study was designed to explore the effect and mechanism of CbA in hyperglycemia via improvement of inflammation and glucose metabolism in the adipose tissue and liver in high-fat diet (HFD)-fed mice. After 10 weeks fed HFD, obese mice supplemented with CbA (25 and 100 mg/kg) for another 10 weeks showed a remarkable reducing adiposity and blood glucose. CbA modulated M1/M2 macrophage polarization, ameliorated inflammatory cytokines, and restored adiponectin as well as Glut 4 expression in the adipose tissue. In the in vitro study, CbA attenuated pro-inflammatory markers while upregulated anti-inflammatory IL-10 in LPS + IFNγ-generated M1 phenotype macrophages. In the liver, CbA attenuated steatosis, inflammatory infiltration, and protein levels of inflammatory TNF-α and IL-6. Moreover, CbA markedly upregulated Adiponectin receptor 1, AMPK, and insulin downstream Akt signaling to improve glycogen content and increase Glut2 protein. These findings indicated that CbA may be a novel therapeutic approach to treat obesity and hyperglycemia phenotype targeting on adipose inflammation and hepatic insulin signaling.
Collapse
Affiliation(s)
- Choirul Anwar
- Institute of Aquatic Science and Technology, Collage of Hydrosphere Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Jing-Ru Lin
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Mei-Ling Tsai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, 08901, USA.
| | - Ching-Shu Lai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan.
| |
Collapse
|
22
|
Hasan S, Ghani N, Zhao X, Good J, Huang A, Wrona HL, Liu J, Liu CJ. Dietary pyruvate targets cytosolic phospholipase A2 to mitigate inflammation and obesity in mice. Protein Cell 2024; 15:661-685. [PMID: 38512816 PMCID: PMC11365557 DOI: 10.1093/procel/pwae014] [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: 11/21/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Obesity has a multifactorial etiology and is known to be a state of chronic low-grade inflammation, known as meta-inflammation. This state is associated with the development of metabolic disorders such as glucose intolerance and nonalcoholic fatty liver disease. Pyruvate is a glycolytic metabolite and a crucial node in various metabolic pathways. However, its role and molecular mechanism in obesity and associated complications are obscure. In this study, we reported that pyruvate substantially inhibited adipogenic differentiation in vitro and its administration significantly prevented HFD-induced weight gain, white adipose tissue inflammation, and metabolic dysregulation. To identify the target proteins of pyruvate, drug affinity responsive target stability was employed with proteomics, cellular thermal shift assay, and isothermal drug response to detect the interactions between pyruvate and its molecular targets. Consequently, we identified cytosolic phospholipase A2 (cPLA2) as a novel molecular target of pyruvate and demonstrated that pyruvate restrained diet-induced obesity, white adipose tissue inflammation, and hepatic steatosis in a cPLA2-dependent manner. Studies with global ablation of cPLA2 in mice showed that the protective effects of pyruvate were largely abrogated, confirming the importance of pyruvate/cPLA2 interaction in pyruvate attenuation of inflammation and obesity. Overall, our study not only establishes pyruvate as an antagonist of cPLA2 signaling and a potential therapeutic option for obesity but it also sheds light on the mechanism of its action. Pyruvate's prior clinical use indicates that it can be considered a safe and viable alternative for obesity, whether consumed as a dietary supplement or as part of a regular diet.
Collapse
Affiliation(s)
- Sadaf Hasan
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10016, United States
| | - Nabil Ghani
- Department of Medicine, Division of Internal Medicine, Saint Peter’s University Hospital, Rutgers University, New Brunswick, NJ 08901, United States
| | - Xiangli Zhao
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10016, United States
- Department of Orthopedics & Rehabilitation, Yale University School of Medicine, New Haven, CT 06510, United States
| | - Julia Good
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10016, United States
| | - Amanda Huang
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10016, United States
- Cornell University, Ithaca, New York, NY, United States
| | - Hailey Lynn Wrona
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10016, United States
- Department of Biomedical Engineering, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, United States
| | - Jody Liu
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10016, United States
- New York University, NY 14853, United States
| | - Chuan-ju Liu
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10016, United States
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, United States
- Department of Orthopedics & Rehabilitation, Yale University School of Medicine, New Haven, CT 06510, United States
| |
Collapse
|
23
|
Lempicki MD, Gray JA, Abuna G, Murata RM, Divanovic S, McNamara CA, Meher AK. BAFF neutralization impairs the autoantibody-mediated clearance of dead adipocytes and aggravates obesity-induced insulin resistance. Front Immunol 2024; 15:1436900. [PMID: 39185417 PMCID: PMC11341376 DOI: 10.3389/fimmu.2024.1436900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
B cell-activating factor (BAFF) is a critical TNF-family cytokine that regulates homeostasis and peripheral tolerance of B2 cells. BAFF overproduction promotes autoantibody generation and autoimmune diseases. During obesity, BAFF is predominantly produced by white adipose tissue (WAT), and IgG autoantibodies against adipocytes are identified in the WAT of obese humans. However, it remains to be determined if the autoantibodies formed during obesity affect WAT remodeling and systemic insulin resistance. Here, we show that IgG autoantibodies are generated in high-fat diet (HFD)-induced obese mice that bind to apoptotic adipocytes and promote their phagocytosis by macrophages. Next, using murine models of obesity in which the gonadal WAT undergoes remodeling, we found that BAFF neutralization depleted IgG autoantibodies, increased the number of dead adipocytes, and exacerbated WAT inflammation and insulin resistance. RNA sequencing of the stromal vascular fraction from the WAT revealed decreased expression of immunoglobulin light-chain and heavy-chain variable genes suggesting a decreased repertoire of B cells after BAFF neutralization. Further, the B cell activation and the phagocytosis pathways were impaired in the WAT of BAFF-neutralized mice. In vitro, plasma IgG fractions from BAFF-neutralized mice reduced the phagocytic clearance of apoptotic adipocytes. Altogether, our study suggests that IgG autoantibodies developed during obesity, at least in part, dampens exacerbated WAT inflammation and systemic insulin resistance.
Collapse
Affiliation(s)
- Melissa D. Lempicki
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Jake A. Gray
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Gabriel Abuna
- School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Ramiro M. Murata
- School of Dental Medicine, East Carolina University, Greenville, NC, United States
| | - Senad Divanovic
- Department of Pediatrics University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Coleen A. McNamara
- Cardiovascular Research Center, Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Akshaya K. Meher
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
24
|
Tang J, Ma Y, Li M, Liu X, Wang Y, Zhang J, Shu H, Liu Z, Zhang C, Fu L, Hu J, Zhang Y, Jia Z, Feng Y. FADD regulates adipose inflammation, adipogenesis, and adipocyte survival. Cell Death Discov 2024; 10:323. [PMID: 39009585 PMCID: PMC11250791 DOI: 10.1038/s41420-024-02089-x] [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: 03/13/2024] [Revised: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
Adipose tissue, aside from adipocytes, comprises various abundant immune cells. The accumulation of low-grade chronic inflammation in adipose tissue serves as a primary cause and hallmark of insulin resistance. In this study, we investigate the physiological roles of FADD in adipose tissue inflammation, adipogenesis, and adipocyte survival. High levels of Fadd mRNA were observed in mitochondrial-rich organs, particularly brown adipose tissue. To explore its metabolic functions, we generated global Fadd knockout mice, resulting in embryonic lethality, while heterozygous knockout (Fadd+/-) mice did not show any significant changes in body weight or composition. However, Fadd+/- mice exhibited reduced respiratory exchange ratio (RER) and serum cholesterol levels, along with heightened global and adipose inflammatory responses. Furthermore, AT masses and expression levels of adipogenic and lipogenic genes were decreased in Fadd+/- mice. In cellular studies, Fadd inhibition disrupted adipogenic differentiation and suppressed the expression of adipogenic and lipogenic genes in cultured adipocytes. Additionally, Fadd overexpression caused adipocyte death in vitro with decreased RIPK1 and RIPK3 expression, while Fadd inhibition downregulated RIPK3 in iWAT in vivo. These findings collectively underscore the indispensable role of FADD in adipose inflammation, adipogenesis, and adipocyte survival.
Collapse
Affiliation(s)
- Jianlei Tang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Endocrinology Department of the Second People's Hospital of Lianyungang City, Lianyungang, China
| | - Yue Ma
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China
| | - Meilin Li
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China
| | - Xiangpeng Liu
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China
| | - Yuting Wang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Zhang
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China
| | - Hui Shu
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China
| | - Zhiwei Liu
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China
| | - Chi Zhang
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China
| | - Lei Fu
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
- Suzhou Medical School, Soochow University, Suzhou, China.
| | - Yong Zhang
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China.
| | - Zhihao Jia
- Cambridge-Suda Genomic Resource Center, Suzhou Medical School, Soochow University, Suzhou, China.
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
- Suzhou Medical School, Soochow University, Suzhou, China.
| |
Collapse
|
25
|
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
|
26
|
Yue C, Tang Y, Chang M, Wang Y, Peng H, Wang X, Wang Z, Zang X, Ben H, Yu G. Dietary supplementation with short- and long-chain structured lipids alleviates obesity via regulating hepatic lipid metabolism, inflammation and gut microbiota in high-fat-diet-induced obese mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:5089-5103. [PMID: 38288873 DOI: 10.1002/jsfa.13344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Obesity is closely associated with lipid accumulation, inflammation and intestinal microbiota dysbiosis. Short- and long-chain type structured lipids (SLCTs) are kinds of low-calorie structured lipids and demonstrate anti-obesity and hypolipidemia bioactivity. The objective of this study is to investigate the potential effects of dietary supplementation of SLCTs rich in short-chain fatty acids and polyunsaturated fatty acids on high-fat-diet-induced obesity and gut microbiota modulation in C57BL/6J mice. RESULTS Results showed that SLCTs supplementation ameliorated body weight, dyslipidemia, liver lipid accumulation, liver injury and systemic inflammation in obese mice. As expected, immunohistochemical analysis showed that SLCTs significantly increased the expression of proliferator-activated receptor alpha and decreased the expression of Toll-like receptor 4 in liver tissue. Furthermore, SLCTs supplementation significantly downregulated the expression level of liver inflammation-related genes while upregulating the expression level of liver lipid metabolism-related genes. Additionally, SLCTs supplementation markedly enhanced the diversity of gut microbiota, reduced the Firmicutes/Bacteroidetes ratio and increased the diversity and richness of beneficial intestinal microorganisms, such as Bacteroides, Lactobacillus, Lachnospiraceae NK4A136 group, Alloprevotella and Ruminococcaceae UCG-014. CONCLUSION Our work suggested that SLCTs may have the potential to reduce obesity associated with a high-fat diet by regulating liver metabolism, inflammation and gut microbiota. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chonghui Yue
- College of Food & Bioengineering, Henan University of Science and Technology, Luoyang, China
- Henan Engineering Research Center of Food Material, Henan University of Science & Technology, Luoyang, China
- College of Food Science, Northeast Agricultural University, Harbin, China
| | - Yu Tang
- College of Food & Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Menghan Chang
- College of Food & Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yueyue Wang
- College of Food & Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Huainan Peng
- College of Food & Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Xin Wang
- College of Food Science, Northeast Agricultural University, Harbin, China
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Ziyu Wang
- College of Food & Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Xiaodan Zang
- College of Public Health, Food Quality and Safety, Mudanjiang Medical University, Mudanjiang, China
| | - Hongyan Ben
- College of Food Science, Northeast Agricultural University, Harbin, China
| | - Guoping Yu
- College of Food Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
27
|
Dahdah N, Tercero-Alcázar C, Malagón MM, Garcia-Roves PM, Guzmán-Ruiz R. Interrelation of adipose tissue macrophages and fibrosis in obesity. Biochem Pharmacol 2024; 225:116324. [PMID: 38815633 DOI: 10.1016/j.bcp.2024.116324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Obesity is characterized by adipose tissue expansion, extracellular matrix remodelling and unresolved inflammation that contribute to insulin resistance and fibrosis. Adipose tissue macrophages represent the most abundant class of immune cells in adipose tissue inflammation and could be key mediators of adipocyte dysfunction and fibrosis in obesity. Although macrophage activation states are classically defined by the M1/M2 polarization nomenclature, novel studies have revealed a more complex range of macrophage phenotypes in response to external condition or the surrounding microenvironment. Here, we discuss the plasticity of adipose tissue macrophages (ATMs) in response to their microenvironment in obesity, with special focus on macrophage infiltration and polarization, and their contribution to adipose tissue fibrosis. A better understanding of the role of ATMs as regulators of adipose tissue remodelling may provide novel therapeutic strategies against obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Norma Dahdah
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Carmen Tercero-Alcázar
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María M Malagón
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Pablo Miguel Garcia-Roves
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Rocío Guzmán-Ruiz
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology and Immunology, IMIBIC, Reina Sofía University Hospital, University of Córdoba, 14004 Córdoba, Spain.
| |
Collapse
|
28
|
Zhang G, Wang S, Ma P, Li S, Sun X, Zhao Y, Pan J. Increased regional body fat is associated with depressive symptoms: a cross-sectional analysis of NHANES data obtained during 2011-2018. BMC Psychiatry 2024; 24:336. [PMID: 38702637 PMCID: PMC11067210 DOI: 10.1186/s12888-024-05782-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
AIMS The findings from previous epidemiological studies of the association between regional body fat and depressive symptoms have been unclear. We aimed to determine the association between the body fat in different regions and depressive symptoms based on data from the National Health and Nutrition Examination Survey (NHANES). METHODS This study included 3393 participants aged ≥ 20 years from the NHANES performed during 2011-2018. Depressive symptoms were assessed using the Patient Health Questionnaire-9. The fat mass (FM) was measured in different regions using dual-energy X-ray absorptiometry to determine the total FM, trunk FM, arm FM, and leg FM. The FM index (FMI) was obtained by dividing the FM in kilograms by the square of the body height in meters. Weighted data were calculated in accordance with analytical guidelines. Linear logistic regression models were used to quantify the association between regional FMI and depressive symptoms. Univariate and stratified analyses were also performed. RESULTS The participants in this study comprised 2066 males and 1327 females. There were 404 (11.91%) participants with depressive symptoms, who were aged 40.89 ± 11.74 years and had a body mass index of 30.07 ± 7.82 kg/m². A significant association was found between total FMI and depressive symptoms. In the fully adjusted multivariate regression model, a higher total FMI (odds ratio = 2.18, 95% confidence interval [CI] = 1.08-4.39) was related to a higher risk of depressive symptoms, while increased total FMI (β = 1.55, 95% CI = 0.65-2.44, p = 0.001), trunk FMI (β = 0.57, 95% CI = 0.04-1.10, p = 0.036), and arm FMI (β = 0.96, 95% CI = 0.33-1.59, p = 0.004) were significantly associated with PHQ-9 (Patient Health Questionnaire-9) scores, whereas the leg FMI was not (p = 0.102). The weighted association between total FMI and depressive symptoms did not differ significantly between most of the subpopulations (all p values for interaction > 0.05). The risk of having depression was higher in individuals who were non-Hispanic Whites, smokers, drinkers, obese, and had diabetes and thyroid problems (p < 0.05). CONCLUSION These findings suggest that the population with a higher regional FMI is more likely to have depressive symptoms, especially in those who also have an increased total FMI. The association is more pronounced in individuals who are smokers, drinkers, obese, and have diabetes and thyroid problems.
Collapse
Affiliation(s)
- GuiMei Zhang
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, 510632, P.R. China
| | - Sisi Wang
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, 510632, P.R. China
| | - Ping Ma
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, 510632, P.R. China
| | - Shuna Li
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xizhe Sun
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, 510632, P.R. China
| | - Yang Zhao
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, 510632, P.R. China
| | - Jiyang Pan
- Department of Psychiatry, Sleep Medicine Centre, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, 510632, P.R. China.
| |
Collapse
|
29
|
Zhao D, Wang Y, Wu S, Ji X, Gong K, Zheng H, Zhu M. Research progress on the role of macrophages in acne and regulation by natural plant products. Front Immunol 2024; 15:1383263. [PMID: 38736879 PMCID: PMC11082307 DOI: 10.3389/fimmu.2024.1383263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
Acne vulgaris is one of the most common skin diseases. The current understanding of acne primarily revolves around inflammatory responses, sebum metabolism disorders, aberrant hormone and receptor expression, colonization by Cutibacterium acnes, and abnormal keratinization of follicular sebaceous glands. Although the precise mechanism of action remains incompletely understood, it is plausible that macrophages exert an influence on these pathological features. Macrophages, as a constituent of the human innate immune system, typically manifest distinct phenotypes across various diseases. It has been observed that the polarization of macrophages toward the M1 phenotype plays a pivotal role in the pathogenesis of acne. In recent years, extensive research on acne has revealed an increasing number of natural remedies exhibiting therapeutic efficacy through the modulation of macrophage polarization. This review investigates the role of cutaneous macrophages, elucidates their potential significance in the pathogenesis of acne, a prevalent chronic inflammatory skin disorder, and explores the therapeutic mechanisms of natural plant products targeting macrophages. Despite these insights, the precise role of macrophages in the pathogenesis of acne remains poorly elucidated. Subsequent investigations in this domain will further illuminate the pathogenesis of acne and potentially offer guidance for identifying novel therapeutic targets for this condition.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yun Wang
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shuhui Wu
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiaotian Ji
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Ke Gong
- Department of Traditional Chinese Medicine, Cangzhou Central Hospital, Cangzhou, China
| | - Huie Zheng
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Mingfang Zhu
- Department of Dermatology, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
30
|
Day KS, Rempel L, Rossi FMV, Theret M. Origins and functions of eosinophils in two non-mucosal tissues. Front Immunol 2024; 15:1368142. [PMID: 38585275 PMCID: PMC10995313 DOI: 10.3389/fimmu.2024.1368142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Eosinophils are a type of granulocyte named after the presence of their eosin-stained granules. Traditionally, eosinophils have been best known to play prominent roles in anti-parasitic responses and mediating allergic reactions. Knowledge of their behaviour has expanded with time, and they are now recognized to play integral parts in the homeostasis of gastrointestinal, respiratory, skeletal muscle, adipose, and connective tissue systems. As such, they are implicated in a myriad of pathologies, and have been the target of several medical therapies. This review focuses on the lifespan of eosinophils, from their origins in the bone marrow, to their tissue-resident role. In particular, we wish to highlight the functions of eosinophils in non-mucosal tissues with skeletal muscle and the adipose tissues as examples, and to discuss the current understanding of their participation in diseased states in these tissues.
Collapse
Affiliation(s)
- Katie S. Day
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Lucas Rempel
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Marine Theret
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
31
|
Abstract
Obesity is epidemic and of great concern because of its comorbid and costly inflammatory-driven complications. Extensive investigations in mice have elucidated highly coordinated, well-balanced interactions between adipocytes and immune cells in adipose tissue that maintain normal systemic metabolism in the lean state, while in obesity, proinflammatory changes occur in nearly all adipose tissue immune cells. Many of these changes are instigated by adipocytes. However, less is known about obesity-induced adipose-tissue immune cell alterations in humans. Upon high-fat diet feeding, the adipocyte changes its well-known function as a metabolic cell to assume the role of an immune cell, orchestrating proinflammatory changes that escalate inflammation and progress during obesity. This transformation is particularly prominent in humans. In this review, we (a) highlight a leading and early role for adipocytes in promulgating inflammation, (b) discuss immune cell changes and the time course of these changes (comparing humans and mice when possible), and (c) note how reversing proinflammatory changes in most types of immune cells, including adipocytes, rescues adipose tissue from inflammation and obese mice from insulin resistance.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Pennsylvania State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA;
| | - Tuo Deng
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Dharti Shantaram
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
| | - Willa A Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
| |
Collapse
|
32
|
Qian S, Shi Y, Senfeld J, Peng Q, Shen J. The P2Y 2 receptor mediates terminal adipocyte differentiation and insulin resistance: Evidence for a dual G-protein coupling mode. J Biol Chem 2024; 300:105589. [PMID: 38141758 PMCID: PMC10828443 DOI: 10.1016/j.jbc.2023.105589] [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: 07/15/2023] [Revised: 11/29/2023] [Accepted: 12/12/2023] [Indexed: 12/25/2023] Open
Abstract
Several P2Y nucleotide receptors have been shown to be involved in the early stage of adipocyte differentiation in vitro and insulin resistance in obese mice; however, the exact receptor subtype(s) and its underlying molecular mechanism in relevant human cells are unclear. Here, using human primary visceral preadipocytes as a model, we found that during preadipocyte-to-mature adipocyte differentiation, the P2Y2 nucleotide receptor (P2Y2R) was the most upregulated subtype among the eight known P2Y receptors and the only one further dramatically upregulated after inflammatory TNFα treatment. Functional studies indicated that the P2Y2R induced intracellular Ca2+, ERK1/2, and JNK signaling but not the p38 pathway. In addition, stimulation of the P2Y2R suppressed basal and insulin-induced phosphorylation of AKT, accompanied by decreased GLUT4 membrane translocation and glucose uptake in mature adipocytes, suggesting a role of P2Y2R in insulin resistance. Mechanistically, we found that activation of P2Y2R did not increase lipolysis but suppressed PIP3 generation. Interestingly, activation of P2Y2R triggered Gi-protein coupling, and pertussis toxin pretreatment largely inhibited P2Y2R-mediated ERK1/2 signaling and cAMP suppression. Further, treatment of the cells with AR-C 118925XX, a selective P2Y2R antagonist, significantly inhibited adipogenesis, and P2Y2R knockout decreased mouse body weight gain with smaller eWAT mass infiltrated with fewer macrophages as compared to WT mice in response to a Western diet. Thus, we revealed that terminal adipocyte differentiation and inflammation selectively upregulate P2Y2R expression and that P2Y2R mediates insulin resistance by suppressing the AKT signaling pathway, highlighting P2Y2R as a potential new drug target to combat obesity and type-2 diabetes.
Collapse
Affiliation(s)
- Shenqi Qian
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama, USA; Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Shi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama, USA
| | - Jared Senfeld
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama, USA
| | - Qianman Peng
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama, USA
| | - Jianzhong Shen
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama, USA.
| |
Collapse
|
33
|
Engin AB. Message Transmission Between Adipocyte and Macrophage in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:273-295. [PMID: 39287855 DOI: 10.1007/978-3-031-63657-8_9] [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: 09/19/2024]
Abstract
Obesity is characterized by the chronic low-grade activation of the innate immune system. In this respect, macrophage-elicited metabolic inflammation and adipocyte-macrophage interaction have primary importance in obesity. Large quantity of macrophages is accumulated by different mechanisms in obese adipose tissue. Hypertrophic adipocyte-derived chemotactic monocyte chemoattractant protein-1 (MCP-1)/C-C chemokine receptor 2 (CCR2) pathway promotes more macrophage accumulation into the obese adipose tissue. However, obesity-induced changes in adipose tissue macrophage density are mainly dependent on increases in the triple-positive cluster of differentiation (CD)11b+ F4/80+ CD11c+ adipose tissue macrophage subpopulation. As epigenetic regulators, microRNAs (miRNAs) are one of the most important mediators of obesity. miRNAs are expressed by adipocytes as well as macrophages and regulate inflammation with the expression of target genes. A paracrine loop involving free fatty acids and tumor necrosis factor-alpha (TNF-α) between adipocytes and macrophages establishes a vicious cycle that aggravates inflammatory changes in the adipose tissue. Adipocyte-specific caspase-1 and production of interleukin-1beta (IL-1β) by macrophages; both adipocyte and macrophage induction by toll-like receptor-4 (TLR4) through nuclear factor-kappaB (NF-κB) activation; free fatty acid-induced and TLR-mediated activation of c-Jun N-terminal kinase (JNK)-related pro-inflammatory pathways in CD11c+ immune cells; are effective in mutual message transmission between adipocyte and macrophage and in the development of adipose tissue inflammation. Thus, the metabolic status of adipocytes and their released exosomes are important determinants of macrophage inflammatory output. However, old adipocytes are removed by macrophages through trogocytosis or sending an "eat me" signal. As a single miRNA can be able to regulate a variety of target genes and signaling pathways, reciprocal transfer of miRNAs between adipocytes and macrophages via miRNA-loaded exosomes reorganizes the different stages of obesity. Changes in the expression of circulating miRNAs because of obesity progression or anti-obesity treatment indicate that miRNAs could be used as potential biomarkers. Therefore, it is believed that targeting macrophage-associated miRNAs with anti-obesity miRNA-loaded nano-carriers may be successful in the attenuation of both obesity and adipose tissue inflammation in clinical practice. Moreover, miRNA-containing exosomes and transferable mitochondria between the adipocyte and macrophage are investigated as new therapeutic targets for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
34
|
Abstract
The signals and structure of the tissues in which leukocytes reside critically mould leukocyte function and development and have challenged our fundamental understanding of how to define and categorize tissue-resident immune cells. One specialized tissue niche that has a powerful effect on immune cell function is adipose tissue. The field of adipose tissue leukocyte biology has expanded dramatically and has revealed how tissue niches can shape immune cell function and reshape them in a setting of metabolic stress, such as obesity. Most notably, adipose tissue macrophages and T cells are under intense investigation due to their contributions to adipose tissue in the lean and obese states. Both adipose tissue macrophages and T cells have features associated with the metabolic function of adipose tissue that are distinct from features of macrophages and T cells that are classically characterized in other tissues. This Review provides state-of-the-art understanding of adipose tissue macrophages and T cells and discusses how their unique niche can help us to better understand diversity in leukocyte responses.
Collapse
Affiliation(s)
- Ramiah D Jacks
- Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carey N Lumeng
- Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
35
|
Xu C, Sarver DC, Lei X, Sahagun A, Zhong J, Na CH, Rudich A, Wong GW. CTRP6 promotes the macrophage inflammatory response, and its deficiency attenuates LPS-induced inflammation. J Biol Chem 2024; 300:105566. [PMID: 38103643 PMCID: PMC10789631 DOI: 10.1016/j.jbc.2023.105566] [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: 05/20/2023] [Revised: 11/27/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023] Open
Abstract
Macrophages play critical roles in inflammation and tissue homeostasis, and their functions are regulated by various autocrine, paracrine, and endocrine factors. We have previously shown that CTRP6, a secreted protein of the C1q family, targets both adipocytes and macrophages to promote obesity-linked inflammation. However, the gene programs and signaling pathways directly regulated by CTRP6 in macrophages remain unknown. Here, we combine transcriptomic and phosphoproteomic analyses to show that CTRP6 activates inflammatory gene programs and signaling pathways in mouse bone marrow-derived macrophages (BMDMs). Treatment of BMDMs with CTRP6 upregulated proinflammatory, and suppressed the antiinflammatory, gene expression. We also showed that CTRP6 activates p44/42-MAPK, p38-MAPK, and NF-κB signaling pathways to promote inflammatory cytokine secretion from BMDMs, and that pharmacologic inhibition of these signaling pathways markedly attenuated the effects of CTRP6. Pretreatment of BMDMs with CTRP6 also sensitized and potentiated the BMDMs response to lipopolysaccharide (LPS)-induced inflammatory signaling and cytokine secretion. Consistent with the metabolic phenotype of proinflammatory macrophages, CTRP6 treatment induced a shift toward aerobic glycolysis and lactate production, reduced oxidative metabolism, and elevated mitochondrial reactive oxygen species production in BMDMs. Importantly, in accordance with our in vitro findings, BMDMs from CTRP6-deficient mice were less inflammatory at baseline and showed a marked suppression of LPS-induced inflammatory gene expression and cytokine secretion. Finally, loss of CTRP6 in mice also dampened LPS-induced inflammation and hypothermia. Collectively, our findings suggest that CTRP6 regulates and primes the macrophage response to inflammatory stimuli and thus may have a role in modulating tissue inflammatory tone in different physiological and disease contexts.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xia Lei
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Ageline Sahagun
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jun Zhong
- Delta Omics Inc, Baltimore, Maryland, USA
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Assaf Rudich
- Faculty of Health Sciences, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
36
|
Martemucci G, Fracchiolla G, Muraglia M, Tardugno R, Dibenedetto RS, D’Alessandro AG. Metabolic Syndrome: A Narrative Review from the Oxidative Stress to the Management of Related Diseases. Antioxidants (Basel) 2023; 12:2091. [PMID: 38136211 PMCID: PMC10740837 DOI: 10.3390/antiox12122091] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Metabolic syndrome (MS) is a growing disorder affecting thousands of people worldwide, especially in industrialised countries, increasing mortality. Oxidative stress, hyperglycaemia, insulin resistance, inflammation, dysbiosis, abdominal obesity, atherogenic dyslipidaemia and hypertension are important factors linked to MS clusters of different pathologies, such as diabesity, cardiovascular diseases and neurological disorders. All biochemical changes observed in MS, such as dysregulation in the glucose and lipid metabolism, immune response, endothelial cell function and intestinal microbiota, promote pathological bridges between metabolic syndrome, diabesity and cardiovascular and neurodegenerative disorders. This review aims to summarise metabolic syndrome's involvement in diabesity and highlight the link between MS and cardiovascular and neurological diseases. A better understanding of MS could promote a novel strategic approach to reduce MS comorbidities.
Collapse
Affiliation(s)
- Giovanni Martemucci
- Department of Agricultural and Environmental Sciences, University of Bari Aldo Moro, 70126 Bari, Italy;
| | - Giuseppe Fracchiolla
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Marilena Muraglia
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Roberta Tardugno
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Roberta Savina Dibenedetto
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | | |
Collapse
|
37
|
Hildreth AD, Padilla ET, Gupta M, Wong YY, Sun R, Legala AR, O'Sullivan TE. Adipose cDC1s contribute to obesity-associated inflammation through STING-dependent IL-12 production. Nat Metab 2023; 5:2237-2252. [PMID: 37996702 DOI: 10.1038/s42255-023-00934-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Obesity is associated with chronic low-grade white adipose tissue (WAT) inflammation that can contribute to the development of insulin resistance in mammals. Previous studies have identified interleukin (IL)-12 as a critical upstream regulator of WAT inflammation and metabolic dysfunction during obesity. However, the cell types and mechanisms that initiate WAT IL-12 production remain unclear. Here we show that conventional type 1 dendritic cells (cDC1s) are the cellular source of WAT IL-12 during obesity through analysis of mouse and human WAT single-cell transcriptomic datasets, IL-12 reporter mice and IL-12p70 protein levels by enzyme-linked immunosorbent assay. We demonstrate that cDC1s contribute to obesity-associated inflammation by increasing group 1 innate lymphocyte interferon-γ production and inflammatory macrophage accumulation. Inducible depletion of cDC1s increased WAT insulin sensitivity and systemic glucose tolerance during diet-induced obesity. Mechanistically, endocytosis of apoptotic bodies containing self-DNA by WAT cDC1s drives stimulator of interferon genes (STING)-dependent IL-12 production. Together, these results suggest that WAT cDC1s act as critical regulators of adipose tissue inflammation and metabolic dysfunction during obesity.
Collapse
Affiliation(s)
- Andrew D Hildreth
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eddie T Padilla
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Meha Gupta
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yung Yu Wong
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ryan Sun
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Akshara R Legala
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
38
|
Araújo M, Moreira D, Mesquita I, Ferreira C, Mendes-Frias A, Barros-Carvalho S, Dinis-Oliveira RJ, Duarte-Oliveira C, Cunha C, Carvalho A, Saha B, Cordeiro-da-Silva A, Estaquier J, Silvestre R. Intramacrophage lipid accumulation compromises T cell responses and is associated with impaired drug therapy against visceral leishmaniasis. Immunology 2023; 170:510-526. [PMID: 37635289 DOI: 10.1111/imm.13686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023] Open
Abstract
Under perturbing conditions such as infection with Leishmania, a protozoan parasite living within the phagosomes in mammalian macrophages, cellular and organellar structures, and metabolism are dynamically regulated for neutralizing the pressure of parasitism. However, how modulations of the host cell metabolic pathways support Leishmania infection remains unknown. Herein, we report that lipid accumulation heightens the susceptibility of mice to L. donovani infection and promotes resistance to first-line anti-leishmanial drugs. Despite being pro-inflammatory, the in vitro generated uninfected lipid-laden macrophages (LLMs) or adipose-tissue macrophages (ATMs) display lower levels of reactive oxygen and nitrogen species. Upon infection, LLMs secrete higher IL-10 and lower IL-12p70 cytokines, inhibiting CD4+ T cell activation and Th1 response suggesting a key modulatory role for intramacrophage lipid accumulation in anti-leishmanial host defence. We, therefore, examined this causal relationship between lipids and immunomodulation using an in vivo high-fat diet (HFD) mouse model. HFD increased the susceptibility to L. donovani infection accompanied by a defective CD4+ Th1 and CD8+ T cell response. The white adipose tissue of HFD mice displays increased susceptibility to L. donovani infection with the preferential infection of F4/80+ CD11b+ CD11c+ macrophages with higher levels of neutral lipids reserve. The HFD increased resistance to a first-line anti-leishmanial drug associated with a defective adaptive immune response. These data demonstrate that the accumulation of neutral lipids contributes to susceptibility to visceral leishmaniasis hindering host-protective immune response and reducing the efficacy of antiparasitic drug therapies.
Collapse
Affiliation(s)
- Marta Araújo
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Diana Moreira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Inês Mesquita
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Carolina Ferreira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Mendes-Frias
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sónia Barros-Carvalho
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
- MTG Research and Development Lab, Porto, Portugal
| | - Cláudio Duarte-Oliveira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Anabela Cordeiro-da-Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Jérôme Estaquier
- INSERM U1124, Université Paris Cité, Paris, France
- Pathophysiology of Cell Death in Host-Pathogen Interactions, CHU de Québec - Université Laval Research Center, Québec City, Québec, Canada
| | - Ricardo Silvestre
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
39
|
Zur Tulod J, Witman ND, Grond K, Duddleston KN, Kurtz CC. Treatment with gut-specific nonsteroidal anti-inflammatory drug attenuates metabolic inflammation but not body mass in fattening ground squirrels. Am J Physiol Regul Integr Comp Physiol 2023; 325:R456-R464. [PMID: 37602382 PMCID: PMC11178295 DOI: 10.1152/ajpregu.00078.2023] [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: 04/06/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
The active season of hibernators corresponds to rapid adiposity in preparation for the next hibernation season. We have previously shown that this dramatic increase in adipose mass is associated with metabolic inflammation similar to what is seen in obesity and metabolic disease. We next sought to determine whether curbing this inflammation at its source (i.e., the gut) would attenuate weight gain in fattening 13-lined ground squirrels (Ictidomys tridecemlineatus). We fed active yearling ground squirrels a diet containing the gut-specific nonsteroidal anti-inflammatory drug mesalazine (5-aminosalicylic acid) for 10 wk. Mesalazine treatment had slight effects on microbial community diversity in the cecum and colon. Not surprisingly, mesalazine treatment decreased inflammatory cytokine levels in the ileum and colon. Mesalazine also decreased proinflammatory and increased anti-inflammatory cytokines in omental white adipose tissue (oWAT). Despite this, body mass was unaffected, and caloric intake increased in mesalazine-treated squirrels, mainly in males. Mass of the primary WAT depot, intra-abdominal WAT (iaWAT), or the highly metabolic oWAT were unaltered by treatment, as was adiposity index. Together, these results suggest that mesalazine treatment has some effects on adiposity in fattening ground squirrels, but this treatment needs to be modified to overcome the strong drive to fatten in this species.NEW & NOTEWORTHY Adiposity and obesity are caused, at least in part, by inflammation of metabolic tissues. Hibernators, like ground squirrels, undergo this same metabolic inflammation during their summer fattening period. We attempted to curb this inflammation, and thus fattening, using mesalazine. We found that mesalazine did curb the inflammation but did not affect fattening, likely due to the strong drive to fatten in hibernators.
Collapse
Affiliation(s)
- Jewel Zur Tulod
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, United States
| | - Nathan D Witman
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, United States
| | - Kirsten Grond
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, Alaska, United States
| | - Khrystyne N Duddleston
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, Alaska, United States
| | - Courtney C Kurtz
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, United States
| |
Collapse
|
40
|
Stansbury CM, Dotson GA, Pugh H, Rehemtulla A, Rajapakse I, Muir LA. A lipid-associated macrophage lineage rewires the spatial landscape of adipose tissue in early obesity. JCI Insight 2023; 8:e171701. [PMID: 37651193 PMCID: PMC10619435 DOI: 10.1172/jci.insight.171701] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/29/2023] [Indexed: 09/02/2023] Open
Abstract
Adipose tissue macrophage (ATM) infiltration is associated with adipose tissue dysfunction and insulin resistance in mice and humans. Recent single-cell data highlight increased ATM heterogeneity in obesity but do not provide a spatial context for ATM phenotype dynamics. We integrated single-cell RNA-Seq, spatial transcriptomics, and imaging of murine adipose tissue in a time course study of diet-induced obesity. Overall, proinflammatory immune cells were predominant in early obesity, whereas nonresident antiinflammatory ATMs predominated in chronic obesity. A subset of these antiinflammatory ATMs were transcriptomically intermediate between monocytes and mature lipid-associated macrophages (LAMs) and were consistent with a LAM precursor (pre-LAM). Pre-LAMs were spatially associated with early obesity crown-like structures (CLSs), which indicate adipose tissue dysfunction. Spatial data showed colocalization of ligand-receptor transcripts related to lipid signaling among monocytes, pre-LAMs, and LAMs, including Apoe, Lrp1, Lpl, and App. Pre-LAM expression of these ligands in early obesity suggested signaling to LAMs in the CLS microenvironment. Our results refine understanding of ATM diversity and provide insight into the dynamics of the LAM lineage during development of metabolic disease.
Collapse
Affiliation(s)
- Cooper M. Stansbury
- Department of Computational Medicine and Bioinformatics
- The Michigan Institute for Computational Discovery and Engineering
| | | | - Harrison Pugh
- Department of Computational Medicine and Bioinformatics
| | | | - Indika Rajapakse
- Department of Computational Medicine and Bioinformatics
- Department of Mathematics, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
41
|
Knights AJ, Farrell EC, Ellis OM, Song MJ, Appleton CT, Maerz T. Synovial macrophage diversity and activation of M-CSF signaling in post-traumatic osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.559514. [PMID: 37873464 PMCID: PMC10592932 DOI: 10.1101/2023.10.03.559514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Objective Synovium is home to immune and stromal cell types that orchestrate inflammation following a joint injury; in particular, macrophages are central protagonists in this process. We sought to define the cellular and temporal dynamics of the synovial immune niche in a mouse model of post-traumatic osteoarthritis (PTOA), and to identify stromal-immune crosstalk mechanisms that coordinate macrophage function and phenotype. Design We induced PTOA in mice using a non-invasive tibial compression model of anterior cruciate ligament rupture (ACLR). Single cell RNA-seq and flow cytometry were used to assess immune cell populations in healthy (Sham) and injured (7d and 28d post-ACLR) synovium. Characterization of synovial macrophage polarization states was performed, alongside computational modeling of macrophage differentiation, as well as implicated transcriptional regulators and stromal-immune communication axes. Results Immune cell types are broadly represented in healthy synovium, but experience drastic expansion and speciation in PTOA, most notably in the macrophage portion. We identified several polarization states of macrophages in synovium following joint injury, underpinned by distinct transcriptomic signatures, and regulated in part by stromal-derived macrophage colony-stimulating factor signaling. The transcription factors Pu.1, Cebpα, Cebpβ, and Jun were predicted to control differentiation of systemically derived monocytes into pro-inflammatory synovial macrophages. Conclusions We defined different synovial macrophage subpopulations present in healthy and injured mouse synovium. Nuanced characterization of the distinct functions, origins, and disease kinetics of macrophage subtypes in PTOA will be critical for targeting these highly versatile cells for therapeutic purposes.
Collapse
Affiliation(s)
| | - Easton C. Farrell
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Olivia M. Ellis
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Michelle J. Song
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - C. Thomas Appleton
- Department of Physiology and Pharmacology, Western University, London ON, Canada
- Bone and Joint Institute, Western University, London, ON, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine – Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
42
|
Zhang XJ, Han XW, Jiang YH, Wang YL, He XL, Liu DH, Huang J, Liu HH, Ye TC, Li SJ, Li ZR, Dong XM, Wu HY, Long WJ, Ni SH, Lu L, Yang ZQ. Impact of inflammation and anti-inflammatory modalities on diabetic cardiomyopathy healing: From fundamental research to therapy. Int Immunopharmacol 2023; 123:110747. [PMID: 37586299 DOI: 10.1016/j.intimp.2023.110747] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/18/2023] [Accepted: 07/29/2023] [Indexed: 08/18/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a prevalent cardiovascular complication of diabetes mellitus, characterized by high morbidity and mortality rates worldwide. However, treatment options for DCM remain limited. For decades, a substantial body of evidence has suggested that the inflammatory response plays a pivotal role in the development and progression of DCM. Notably, DCM is closely associated with alterations in inflammatory cells, exerting direct effects on major resident cells such as cardiomyocytes, vascular endothelial cells, and fibroblasts. These cellular changes subsequently contribute to the development of DCM. This article comprehensively analyzes cellular, animal, and human studies to summarize the latest insights into the impact of inflammation on DCM. Furthermore, the potential therapeutic effects of current anti-inflammatory drugs in the management of DCM are also taken into consideration. The ultimate goal of this work is to consolidate the existing literature on the inflammatory processes underlying DCM, providing clinicians with the necessary knowledge and tools to adopt a more efficient and evidence-based approach to managing this condition.
Collapse
Affiliation(s)
- Xiao-Jiao Zhang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Xiao-Wei Han
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Yan-Hui Jiang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Ya-Le Wang
- Shanghai University of Traditional Chinese Medicine, 1200 Cai lun Road, Pudong New District, Shanghai 201203, China; Shenzhen Hospital, Shanghai University of Traditional Chinese Medicine, 16 Xian tong Road, Luo hu District, Shenzhen, Guangdong 518004, China
| | - Xing-Ling He
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Dong-Hua Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Jie Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Hao-Hui Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Tao-Chun Ye
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Si-Jing Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Zi-Ru Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Xiao-Ming Dong
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Hong-Yan Wu
- Shanghai University of Traditional Chinese Medicine, 1200 Cai lun Road, Pudong New District, Shanghai 201203, China; Shenzhen Hospital, Shanghai University of Traditional Chinese Medicine, 16 Xian tong Road, Luo hu District, Shenzhen, Guangdong 518004, China.
| | - Wen-Jie Long
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China.
| | - Shi-Hao Ni
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China.
| | - Lu Lu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China.
| | - Zhong-Qi Yang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; University Key Laboratory of Traditional Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangdong Province 510407, China; Guangzhou Key Laboratory for Chinese Medicine Prevention and Treatment of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China.
| |
Collapse
|
43
|
Vick LV, Canter RJ, Monjazeb AM, Murphy WJ. Multifaceted effects of obesity on cancer immunotherapies: Bridging preclinical models and clinical data. Semin Cancer Biol 2023; 95:88-102. [PMID: 37499846 PMCID: PMC10836337 DOI: 10.1016/j.semcancer.2023.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/04/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Obesity, defined by excessive body fat, is a highly complex condition affecting numerous physiological processes, such as metabolism, proliferation, and cellular homeostasis. These multifaceted effects impact cells and tissues throughout the host, including immune cells as well as cancer biology. Because of the multifaceted nature of obesity, common parameters used to define it (such as body mass index in humans) can be problematic, and more nuanced methods are needed to characterize the pleiotropic metabolic effects of obesity. Obesity is well-accepted as an overall negative prognostic factor for cancer incidence, progression, and outcome. This is in part due to the meta-inflammatory and immunosuppressive effects of obesity. Immunotherapy is increasingly used in cancer therapy, and there are many different types of immunotherapy approaches. The effects of obesity on immunotherapy have only recently been studied with the demonstration of an "obesity paradox", in which some immune therapies have been demonstrated to result in greater efficacy in obese subjects despite the direct adverse effects of obesity and excess body fat acting on the cancer itself. The multifactorial characteristics that influence the effects of obesity (age, sex, lean muscle mass, underlying metabolic conditions and drugs) further confound interpretation of clinical data and necessitate the use of more relevant preclinical models mirroring these variables in the human scenario. Such models will allow for more nuanced mechanistic assessment of how obesity can impact, both positively and negatively, cancer biology, host metabolism, immune regulation, and how these intersecting processes impact the delivery and outcome of cancer immunotherapy.
Collapse
Affiliation(s)
- Logan V Vick
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Robert J Canter
- Department of Surgery, Division of Surgical Oncology, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, University of California School of Medicine, Sacramento, CA, USA
| | - William J Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA; Department of Internal Medicine, Division of Malignant Hematology, Cellular Therapy and Transplantation, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
44
|
Townsend LK, Steinberg GR. AMPK and the Endocrine Control of Metabolism. Endocr Rev 2023; 44:910-933. [PMID: 37115289 DOI: 10.1210/endrev/bnad012] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/10/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Complex multicellular organisms require a coordinated response from multiple tissues to maintain whole-body homeostasis in the face of energetic stressors such as fasting, cold, and exercise. It is also essential that energy is stored efficiently with feeding and the chronic nutrient surplus that occurs with obesity. Mammals have adapted several endocrine signals that regulate metabolism in response to changes in nutrient availability and energy demand. These include hormones altered by fasting and refeeding including insulin, glucagon, glucagon-like peptide-1, catecholamines, ghrelin, and fibroblast growth factor 21; adipokines such as leptin and adiponectin; cell stress-induced cytokines like tumor necrosis factor alpha and growth differentiating factor 15, and lastly exerkines such as interleukin-6 and irisin. Over the last 2 decades, it has become apparent that many of these endocrine factors control metabolism by regulating the activity of the AMPK (adenosine monophosphate-activated protein kinase). AMPK is a master regulator of nutrient homeostasis, phosphorylating over 100 distinct substrates that are critical for controlling autophagy, carbohydrate, fatty acid, cholesterol, and protein metabolism. In this review, we discuss how AMPK integrates endocrine signals to maintain energy balance in response to diverse homeostatic challenges. We also present some considerations with respect to experimental design which should enhance reproducibility and the fidelity of the conclusions.
Collapse
Affiliation(s)
- Logan K Townsend
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Gregory R Steinberg
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
45
|
Pimenta R, Camargo JA, Gonçalves GL, Ghazarian V, Candido P, Guimarães VR, Romão P, Chiovatto C, da Silva KS, Dos Santos GA, Silva IA, Nahas WC, Leite KR, Pessoa AFM, Viana NI, Reis ST. Overexpression of miR-17-5p may negatively impact p300/CBP factor-associated inflammation in a hypercholesterolemic advanced prostate cancer model. Mol Biol Rep 2023; 50:7333-7345. [PMID: 37439896 DOI: 10.1007/s11033-023-08638-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/26/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND Previously, we demonstrated that cholesterol triggers the increase in p300/CBP-associated factor (PCAF), targeted by miR-17-5p. The p300, IL-6, PCAF, and miR-17-5p genes have important and contradictory roles in inflammation and prostate cancer (PCa). This study aimed to demonstrate the potential anti-inflammatory effect of miR-17-5 in an advanced PCa model with diet-induced hypercholesterolemia. METHODS AND RESULTS In vitro, using the PC-3 cell line, we show that induction of miR-17-5p reduces p300 and PCAF expression, increases apoptosis, and decreases cell migration. Furthermore, we demonstrate that supplementing this same cell with cholesterol (2 µg/mL) triggers increased p300, IL-6, and PCAF. In vivo, after establishing the hypercholesterolemic (HCOL) model, xenografts were treated with miR-17-5p. Increased expression of this miR after intratumoral injections attenuated tumor growth in the control and HCOL animals and reduced cell proliferation. CONCLUSION Our results demonstrate that inducing miR-17-5p expression suppresses tumor growth and inflammatory mediator expression. Further studies should be conducted to fully explore the role of miR-17-5p and the involvement of inflammatory mediators p300, PCAF, and IL-6.
Collapse
Affiliation(s)
- Ruan Pimenta
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil.
- D'Or Institute for Research and Education (IDOR), Sao Paulo, Brazil.
| | - Juliana A Camargo
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
| | - Guilherme L Gonçalves
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Vitória Ghazarian
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
| | - Patrícia Candido
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
| | - Vanessa R Guimarães
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
| | - Poliana Romão
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
| | - Caroline Chiovatto
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
- Centro Universitário São Camilo, São Paulo, 04263-200, Brazil
| | - Karina Serafim da Silva
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
- Centro Universitário São Camilo, São Paulo, 04263-200, Brazil
| | - Gabriel A Dos Santos
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
- D'Or Institute for Research and Education (IDOR), Sao Paulo, Brazil
| | - Iran A Silva
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
| | - William C Nahas
- Uro-Oncology Group, Urology Department, University of Sao Paulo Medical School and Institute of Cancer Estate of Sao Paulo (ICESP), Sao Paulo, SP, 01246-000, Brazil
| | - Kátia R Leite
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
| | - Ana Flávia Marçal Pessoa
- Natural Products and Derivatives Laboratory, Department of Surgery, University of São Paulo Medical School, São Paulo, SP, 01246-903, Brazil
| | - Nayara I Viana
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
- Universidade do Estado de Minas Gerais - UEMG, Avenida Juca Stockler, Passos, MG, 1130, Brasil
| | - Sabrina T Reis
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo 455, 2° floor, room 2145, Sao Paulo, Sao Paulo, SP, BR, SP, 01246- 903, Brazil
| |
Collapse
|
46
|
Zapata RC, Nasamran CA, Chilin-Fuentes DR, Dulawa SC, Osborn O. Identification of adipose tissue transcriptomic memory of anorexia nervosa. Mol Med 2023; 29:109. [PMID: 37582711 PMCID: PMC10428576 DOI: 10.1186/s10020-023-00705-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND Anorexia nervosa (AN) is a complex debilitating disease characterized by intense fear of weight gain and excessive exercise. It is the deadliest of any psychiatric disorder with a high rate of recidivism, yet its pathophysiology is unclear. The Activity-Based Anorexia (ABA) paradigm is a widely accepted mouse model of AN that recapitulates hypophagia and hyperactivity despite reduced body weight, however, not the chronicity. METHODS Here, we modified the prototypical ABA paradigm to increase the time to lose 25% of baseline body weight from less than 7 days to more than 2 weeks. We used this paradigm to identify persistently altered genes after weight restoration that represent a transcriptomic memory of under-nutrition and may contribute to AN relapse using RNA sequencing. We focused on adipose tissue as it was identified as a major location of transcriptomic memory of over-nutririon. RESULTS We identified 300 dysregulated genes that were refractory to weight restroration after ABA, including Calm2 and Vps13d, which could be potential global regulators of transcriptomic memory in both chronic over- and under-nutrition. CONCLUSION We demonstrated the presence of peristent changes in the adipose tissue transcriptome in the ABA mice after weight restoration. Despite being on the opposite spectrum of weight perturbations, majority of the transcriptomic memory genes of under- and over-nutrition did not overlap, suggestive of the different mechanisms involved in these extreme nutritional statuses.
Collapse
Affiliation(s)
- Rizaldy C Zapata
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, San Diego, USA.
| | - Chanond A Nasamran
- Center for Computational Biology & Bioinformatics, School of Medicine, University of California San Diego, San Diego, USA
| | - Daisy R Chilin-Fuentes
- Center for Computational Biology & Bioinformatics, School of Medicine, University of California San Diego, San Diego, USA
| | - Stephanie C Dulawa
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, 92093, San Diego, CA, USA
| | - Olivia Osborn
- Division of Endocrinology and Metabolism, School of Medicine, University of California San Diego, San Diego, USA
| |
Collapse
|
47
|
Zeng R, Jiang R, Huang W, Wang J, Zhang L, Ma Y, Wu Y, Meng M, Lan H, Lian Q, Leung FW, Sha W, Chen H. Dissecting shared genetic architecture between obesity and multiple sclerosis. EBioMedicine 2023; 93:104647. [PMID: 37300932 PMCID: PMC10363440 DOI: 10.1016/j.ebiom.2023.104647] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Observational studies have associated obesity with an increased risk of multiple sclerosis (MS). However, the role of genetic factors in their comorbidity remains largely unknown. Our study aimed to investigate the shared genetic architecture underlying obesity and MS. METHODS By leveraging data from genome-wide association studies, we investigated the genetic correlation of body mass index (BMI) and MS by linkage disequilibrium score regression and genetic covariance analyser. The casualty was identified by bidirectional Mendelian randomisation. Linkage disequilibrium score regression in specifically expressed genes and multimarker analysis of GenoMic annotation was utilised to explore single-nucleotide polymorphism (SNP) enrichment at the tissue and cell-type levels. Shared risk SNPs were derived using cross-trait meta-analyses and Heritability Estimation from Summary Statistics. We explored the potential functional genes using summary-data-based Mendelian randomization (SMR). The expression profiles of the risk gene in tissues were further examined. FINDINGS We found a significantly positive genetic correlation between BMI and MS, and the causal association of BMI with MS was supported (β = 0.22, P = 8.03E-05). Cross-trait analysis yielded 39 shared risk SNPs, and the risk gene GGNBP2 was consistently identified in SMR. We observed tissue-specific level SNP heritability enrichment for BMI mainly in brain tissues for MS in immune-related tissues, and cell-type-specific level SNP heritability enrichment in 12 different immune cell types in brain, spleen, lung, and whole blood. The expressions of GGNBP2 were significantly altered in the tissues of patients with obesity or MS compared to those of control subjects. INTERPRETATION Our study indicates the genetic correlation and shared risk genes between obesity and MS. These findings provide insights into the potential mechanisms behind their comorbidity and the future development of therapeutics. FUNDING This work was funded by the National Natural Science Foundation of China (82171698, 82170561, 81300279, and 81741067), the Program for High-level Foreign Expert Introduction of China (G2022030047L), the Natural Science Foundation for Distinguished Young Scholars of Guangdong Province (2021B1515020003), Natural Science Foundation of Guangdong Province (2022A1515012081), the Foreign Distinguished Teacher Program of Guangdong Science and Technology Department (KD0120220129), the Climbing Programme of Introduced Talents and High-level Hospital Construction Project of Guangdong Provincial People's Hospital (DFJH201803, KJ012019099, KJ012021143, and KY012021183), and in part by VA Clinical Merit and ASGE clinical research funds (FWL).
Collapse
Affiliation(s)
- Ruijie Zeng
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Rui Jiang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China; School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Wentao Huang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jiaxuan Wang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Lijun Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yuying Ma
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yanjun Wu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Meijun Meng
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Hekui Lan
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Qizhou Lian
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Felix W Leung
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Sepulveda Ambulatory Care Center, Veterans Affairs Greater Los Angeles Healthcare System, North Hills, CA, USA.
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China; School of Medicine, South China University of Technology, Guangzhou 510006, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China; School of Medicine, South China University of Technology, Guangzhou 510006, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| |
Collapse
|
48
|
Leo S, Tremoli E, Ferroni L, Zavan B. Role of Epicardial Adipose Tissue Secretome on Cardiovascular Diseases. Biomedicines 2023; 11:1653. [PMID: 37371748 DOI: 10.3390/biomedicines11061653] [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: 04/15/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Obesity and insulin resistance are associated with the inflamed and defective adipose tissue (AT) phenotype, and are established risk factors for cardiovascular diseases (CVDs). Extracellular vesicles (EVs) are a heterogeneous group of cell-derived lipid membrane vesicles involved in the onset and development of many pathologies, including insulin resistance, diabetes, and CVDs. The inflammation associated with overweight and obesity triggers the transition of the AT secretome from healthy to pathological, with a consequent increased expression of pro-inflammatory mediators. Epicardial adipose tissue (EAT) is a specialized fat depot that surrounds the heart, in direct contact with the myocardium. Recently, the role of EAT in regulating the physiopathology of many heart diseases has been increasingly explored. In particular, the EAT phenotype and derived EVs have been associated with the onset and exacerbation of CVDs. In this review, we will focus on the role of the AT secretome in the case of CVDs, and will discuss the beneficial effects of EVs released by AT as promising therapeutic candidates.
Collapse
Affiliation(s)
- Sara Leo
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy
| | - Letizia Ferroni
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy
| | - Barbara Zavan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
49
|
Kuziel G, Moore BN, Arendt LM. Obesity and Fibrosis: Setting the Stage for Breast Cancer. Cancers (Basel) 2023; 15:cancers15112929. [PMID: 37296891 DOI: 10.3390/cancers15112929] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity is a rising health concern and is linked to a worsened breast cancer prognosis. Tumor desmoplasia, which is characterized by elevated numbers of cancer-associated fibroblasts and the deposition of fibrillar collagens within the stroma, may contribute to the aggressive clinical behavior of breast cancer in obesity. A major component of the breast is adipose tissue, and fibrotic changes in adipose tissue due to obesity may contribute to breast cancer development and the biology of the resulting tumors. Adipose tissue fibrosis is a consequence of obesity that has multiple sources. Adipocytes and adipose-derived stromal cells secrete extracellular matrix composed of collagen family members and matricellular proteins that are altered by obesity. Adipose tissue also becomes a site of chronic, macrophage-driven inflammation. Macrophages exist as a diverse population within obese adipose tissue and mediate the development of fibrosis through the secretion of growth factors and matricellular proteins and interactions with other stromal cells. While weight loss is recommended to resolve obesity, the long-term effects of weight loss on adipose tissue fibrosis and inflammation within breast tissue are less clear. Increased fibrosis within breast tissue may increase the risk for tumor development as well as promote characteristics associated with tumor aggressiveness.
Collapse
Affiliation(s)
- Genevra Kuziel
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| | - Lisa M Arendt
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| |
Collapse
|
50
|
Wang HW, Tang J, Sun L, Li Z, Deng M, Dai Z. Mechanism of immune attack in the progression of obesity-related type 2 diabetes. World J Diabetes 2023; 14:494-511. [PMID: 37273249 PMCID: PMC10236992 DOI: 10.4239/wjd.v14.i5.494] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/06/2023] [Accepted: 03/30/2023] [Indexed: 05/15/2023] Open
Abstract
Obesity and overweight are widespread issues in adults, children, and adolescents globally, and have caused a noticeable rise in obesity-related complications such as type 2 diabetes mellitus (T2DM). Chronic low-grade inflammation is an important promotor of the pathogenesis of obesity-related T2DM. This proinflammatory activation occurs in multiple organs and tissues. Immune cell-mediated systemic attack is considered to contribute strongly to impaired insulin secretion, insulin resistance, and other metabolic disorders. This review focused on highlighting recent advances and underlying mechanisms of immune cell infiltration and inflammatory responses in the gut, islet, and insulin-targeting organs (adipose tissue, liver, skeletal muscle) in obesity-related T2DM. There is current evidence that both the innate and adaptive immune systems contribute to the development of obesity and T2DM.
Collapse
Affiliation(s)
- Hua-Wei Wang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Jun Tang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Li Sun
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Zhen Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Ming Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Zhe Dai
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|