1
|
Yin S, Wang J, Jia Y, Wang X, Zhao Y, Liu T, Lv W, Duan Y, Zhao S, Wang S, Liu L. Sleep deprivation-induced sympathetic activation promotes pro-tumoral macrophage phenotype via the ADRB2/KLF4 pathway to facilitate NSCLC metastasis. iScience 2025; 28:112321. [PMID: 40276761 PMCID: PMC12018092 DOI: 10.1016/j.isci.2025.112321] [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: 09/10/2024] [Revised: 01/01/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Sleep deprivation is one of concomitant symptoms of cancer patients, particularly those with non-small cell lung cancer (NSCLC). The potential effect of sleep deprivation on tumor progression and underlying mechanisms remain to be fully investigated. Using a sleep-deprived tumor-bearing mouse model, we found that sleep deprivation altered immune cell composition and regulated pro-tumoral M2 macrophage polarization by the sympathetic nervous system. Furthermore, we identified a role of catecholaminergic neurons in the rostral ventrolateral medulla (RVLM) in influencing NSCLC metastasis. Clinical analyses revealed a correlation between sympathetic-related indicators and poor prognosis. Mechanistically, our findings indicate that sleep deprivation facilitates the polarization of pro-tumoral macrophages by upregulating β2-adrenergic receptor (ADRB2), which subsequently enhances the expression of Kruppel-like transcription factor 4 (KLF4) through the JAK1/STAT6 phosphorylation pathway. These findings highlight a neuro-immune mechanism linking sleep deprivation to NSCLC metastasis, suggesting that targeting the ADRB2/KLF4 axis could improve outcomes for sleep-deprived NSCLC patients.
Collapse
Affiliation(s)
- Shuxian Yin
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
- Hebei Key Laboratory of Stomatology, Hebei Technology Innovation Center of Oral Health, Hebei Medical University, Shijiazhuang, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Yunlong Jia
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Xiaoyi Wang
- Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, China
| | - Yan Zhao
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Tianxu Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Wei Lv
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Yuqing Duan
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
| | - Song Zhao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Sheng Wang
- Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University and Hebei Cancer Research Institute, Shijiazhuang, China
- International Cooperation Laboratory of Stem Cell Research, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2
|
Rahman SMN, Yung JHM, Volchuk A, Goldenberg NM, Giacca A. Metabolic phenotypes in a Lyz2Cre recombinase mouse model. Front Immunol 2025; 16:1499858. [PMID: 40170862 PMCID: PMC11958952 DOI: 10.3389/fimmu.2025.1499858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
The Cre-Lox system is essential in biomedical research for precise gene deletion in specific cell types, crucial for understanding genetic roles in disease. Although generally considered non-detrimental, Cre recombinase expression has been associated with potential adverse effects, including Cre toxicity, ectopic expression, and disruption of endogenous genes. We investigated the role of macrophage nucleotide-binding oligomerization domain (Nod1) in obesity-associated diabetes using myeloid-specific Nod1-knockout mice (Nod1 floxed crossed with Lyz2Cre). Our study examined Lyz2Cre as well as floxed control mice separately, unlike most research. Results indicated that Lyz2Cre expression alone impacts glucose metabolism, challenging the notion that Cre expression is harmless. This finding highlights the critical importance of including Cre-only controls in studies using floxed alleles to generate conditional knockout mouse models in order to ensure robust and accurate conclusions in molecular research.
Collapse
Affiliation(s)
- S. M. Niazur Rahman
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Justin Hou Ming Yung
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Allen Volchuk
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Neil M. Goldenberg
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Anesthesiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Adria Giacca
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Díaz-Castro F, Morselli E, Claret M. Interplay between the brain and adipose tissue: a metabolic conversation. EMBO Rep 2024; 25:5277-5293. [PMID: 39558137 PMCID: PMC11624209 DOI: 10.1038/s44319-024-00321-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
The central nervous system and adipose tissue interact through complex communication. This bidirectional signaling regulates metabolic functions. The hypothalamus, a key homeostatic brain region, integrates exteroceptive and interoceptive signals to control appetite, energy expenditure, glucose, and lipid metabolism. This regulation is partly achieved via the nervous modulation of white (WAT) and brown (BAT) adipose tissue. In this review, we highlight the roles of sympathetic and parasympathetic innervation in regulating WAT and BAT activities, such as lipolysis and thermogenesis. Adipose tissue, in turn, plays a dual role as an energy reservoir and an endocrine organ, secreting hormones that influence brain function and metabolic health. In addition, this review focuses on recently uncovered communication pathways, including extracellular vesicles and neuro-mesenchymal units, which add new layers of regulation and complexity to the brain-adipose tissue interaction. Finally, we also examine the consequences of disrupted communication between the brain and adipose tissue in metabolic disorders like obesity and type-2 diabetes, emphasizing the potential for new therapeutic strategies targeting these pathways to improve metabolic health.
Collapse
Affiliation(s)
- Francisco Díaz-Castro
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile
- Physiology Department, Biological Science Faculty, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- IBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Song L, Huang S, Yan H, Ma Q, Luo Q, Qiu J, Chen M, Li Z, Jiang H, Chen Y, Chen F, Du Y, Fu H, Zhao L, Zhao K, Qiu P. ADRB2 serves as a novel biomarker and attenuates alcoholic hepatitis via the SIRT1/PGC-1α/PPARα pathway: integration of WGCNA, machine learning and experimental validation. Front Pharmacol 2024; 15:1423031. [PMID: 39640486 PMCID: PMC11617210 DOI: 10.3389/fphar.2024.1423031] [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: 04/25/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Background Alcoholic hepatitis is a severe inflammatory liver disease. In recent years, the incidence of AH has been on the rise, leading to an increasingly severe disease burden. Currently, there is a lack of specific biomarkers for the diagnosis and prognosis of AH in clinical practice. Therefore, the main objective of this study is to identify biomarkers closely associated with the progression of AH, to address the shortcomings in pathological diagnosis, and to identify potential therapeutic targets. Methods Bioinformatics and machine learning methods were used to comparatively study the differentially expressed genes (DEGs) between AH patients and healthy individuals by analyzing four mRNA microarray data sets obtained from the GEO database. Subsequently, the role of potential biomarkers in AH and their mechanism of action were further confirmed by AH patients and in vitro and in vivo experiments. Results Using differential analysis and WGCNA of the data set, a total of 167 key genes that may be related to AH were obtained. Among 167 genes, the LASSO logistic regression algorithm identified four potential biomarkers (KCNJ10, RPL21P23, ADRB2, and AC025279.1). Notably, ADRB2 showed biomarker potential in GSE28619, GSE94397, and E-MTAB-2664 datasets, and clinical liver samples. Furthermore, AH patients and in vivo experiments demonstrated ADRB2 inhibition and suppression of SIRT1/PPARα/PGC-1α signaling pathways, accompanied by elevated inflammatory factors and lipid deposition. In vitro experiments showed that ADRB2 overexpression mitigated the inhibition of the SIRT1/PPARα/PGC-1α signaling pathway, reversing the decrease in mitochondrial membrane potential, cell apoptosis, oxidative stress, and lipid deposition induced by alcohol exposure. Besides, the results also showed that ADRB2 expression in AH was negatively correlated with the levels of inflammatory factors (e.g., CCL2, CXCL8, and CXCL10). Conclusion This study points to ADRB2 as a promising biomarker with potential diagnostic and prognostic value in clinical cohort data. In addition, in AH patients, in vivo and in vitro experiments confirmed the key role of ADRB2 in the progression of AH. These findings suggest that ADRB2 may alleviate AH by activating the SIRT1/PPARα/PGC-1α pathway. This finding provides a new perspective for the diagnosis and treatment of AH.
Collapse
Affiliation(s)
- Li Song
- Tongde Hospital of Zhejiang Province affiliated to Zhejiang Chinese Medical University, Analysis and Testing Center, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Honghao Yan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qihan Luo
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiang Qiu
- Department of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Minxia Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zongyuan Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - He Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufan Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangming Chen
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haozhe Fu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lisha Zhao
- Tongde Hospital of Zhejiang Province affiliated to Zhejiang Chinese Medical University, Analysis and Testing Center, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Kanglu Zhao
- Zhejiang Rehabilitation Medical Center, Rehabilitation Hospital Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- School of Medicine, The Fourth Affiliated Hospital Zhejiang University, Yiwu, Zhejiang, China
| | - Ping Qiu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
5
|
Gu L, Chen H, Geng R, Liang T, Chen Y, Wang Z, Ye L, Sun M, Shi Q, Wan G, Chang J, Wei J, Ma W, Xiao J, Bao X, Wang R. Endothelial pyroptosis-driven microglial activation in choroid plexus mediates neuronal apoptosis in hemorrhagic stroke rats. Neurobiol Dis 2024; 201:106695. [PMID: 39370051 DOI: 10.1016/j.nbd.2024.106695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Spontaneous intracerebral hemorrhage (ICH) is associated with alarmingly high rates of disability and mortality, and current therapeutic options are suboptimal. A critical component of ICH pathology is the initiation of a robust inflammatory response, often termed "cytokine storm," which amplifies the secondary brain injury following the initial hemorrhagic insult. The precise sources and consequences of this cytokine-driven inflammation are not fully elucidated, necessitating further investigation. METHODS To address this knowledge gap, our study conducted a comprehensive cytokine profiling using Luminex® assays, assessing 23 key cytokines. We then employed single-cell RNA sequencing and spatial transcriptomics at three critical time points post-ICH: the hyperacute, acute, and subacute phases. Integrating these multimodal analyses allowed us to identify the cellular origins of cytokines and elucidate their mechanisms of action. RESULTS Luminex® cytokine assays revealed a significant upregulation of IL-6 and IL-1β levels at the 24-h post-ICH time point. Through the integration of scRNA-seq and spatial transcriptomics in the hemorrhagic hemisphere of rats, we observed a pronounced activation of cytokine-related signaling pathways within the choroid plexus. Initially, immune cell presence was sparse, but it surged 24 h post-ICH, particularly in the choroid plexus, indicating a substantial shift in the immune microenvironment. We traced the source of IL-1β and IL-6 to endothelial cells, establishing a link to pyroptosis. Endothelial pyroptosis post-ICH induced the production of IL-1β and IL-6, which activated microglial polarization characterized by elevated expression of Msr1, Lcn2, and Spp1 via the NF-κB pathway in the choroid plexus. Furthermore, we identified neuronal populations undergoing apoptosis, mediated by the Lcn2-SLC22A17 pathway in response to IL-1β and IL-6 signaling. Notably, the inhibition of pyroptosis using VX-765 significantly mitigated neurological impairments. CONCLUSIONS Our study provides evidence that endothelial pyroptosis, characterized by the release of IL-1β and IL-6, triggers microglial polarization through NF-κB pathway activation, ultimately leading to microglia-mediated neuronal apoptosis in the choroid plexus post-ICH. These findings suggest that targeted therapeutic strategies aimed at mitigating endothelial cell pyroptosis and neutralizing inflammatory cytokines may offer neuroprotection for both microglia and neurons, presenting a promising avenue for ICH treatment.
Collapse
Affiliation(s)
- Lingui Gu
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Hualin Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Ruxu Geng
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Tingyu Liang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yihao Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhuo Wang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China; Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, PR China.
| | - Liguo Ye
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Mingjiang Sun
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Qinglei Shi
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China; Shenzhen Research Institute of Big Data, Shenzhen 518172, China
| | - Gui Wan
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianbo Chang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Junji Wei
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Wenbin Ma
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Jiashun Xiao
- Shenzhen Research Institute of Big Data, Shenzhen 518172, China.
| | - Xinjie Bao
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China.
| | - Renzhi Wang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, PR China.
| |
Collapse
|
6
|
Cieślik M, Strobel SD, Bryniarski P, Twardowska H, Chmielowski A, Rudek M, Felkle D, Zięba K, Kaleta K, Jarczyński M, Nowak B, Bryniarski K, Nazimek K. Hypotensive drugs mitigate the high-sodium diet-induced pro-inflammatory activation of mouse macrophages in vivo. Biomed Pharmacother 2024; 175:116648. [PMID: 38677242 DOI: 10.1016/j.biopha.2024.116648] [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: 02/01/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024] Open
Abstract
Nowadays, there is an increasing emphasis on the need to alleviate the chronic inflammatory response to effectively treat hypertension. However, there are still gaps in our understanding on how to achieve this. Therefore, research on interaction of antihypertensive drugs with the immune system is extremely interesting, since their therapeutic effect could partly result from amelioration of hypertension-related inflammation, in which macrophages seem to play a pivotal role. Thus, current comprehensive studies have investigated the impact of repeatedly administered hypotensive drugs (captopril, olmesartan, propranolol, carvedilol, amlodipine, verapamil) on macrophage functions in the innate and adaptive immunity, as well as if drug-induced effects are affected by a high-sodium diet (HSD), one of the key environmental risk factors of hypertension. Although the assayed medications increased the generation of reactive oxygen and nitrogen intermediates by macrophages from standard fed donors, they reversed HSD-induced enhancing effects on macrophage oxidative burst and secretion of pro-inflammatory cytokines. On the other hand, some drugs increased macrophage phagocytic activity and the expression of surface markers involved in antigen presentation, which translated into enhanced macrophage ability to activate B cells for antibody production. Moreover, the assayed medications augmented macrophage function and the effector phase of contact hypersensitivity reaction, but suppressed the sensitization phase of cell-mediated hypersensitivity under HSD conditions. Our current findings contribute to the recognition of mechanisms, by which excessive sodium intake affects macrophage immune activity in hypertensive individuals, and provide evidence that the assayed medications mitigate most of the HSD-induced adverse effects, suggesting their additional protective therapeutic activity.
Collapse
Affiliation(s)
- Martyna Cieślik
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Spencer D Strobel
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Paweł Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Hanna Twardowska
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Adam Chmielowski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Michał Rudek
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Dominik Felkle
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Katarzyna Zięba
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Konrad Kaleta
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Mateusz Jarczyński
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Bernadeta Nowak
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Katarzyna Nazimek
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland.
| |
Collapse
|
7
|
Chavakis T, Alexaki VI, Ferrante AW. Macrophage function in adipose tissue homeostasis and metabolic inflammation. Nat Immunol 2023; 24:757-766. [PMID: 37012544 DOI: 10.1038/s41590-023-01479-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 01/23/2023] [Indexed: 04/05/2023]
Abstract
Obesity-related metabolic organ inflammation contributes to cardiometabolic disorders. In obese individuals, changes in lipid fluxes and storage elicit immune responses in the adipose tissue (AT), including expansion of immune cell populations and qualitative changes in the function of these cells. Although traditional models of metabolic inflammation posit that these immune responses disturb metabolic organ function, studies now suggest that immune cells, especially AT macrophages (ATMs), also have important adaptive functions in lipid homeostasis in states in which the metabolic function of adipocytes is taxed. Adverse consequences of AT metabolic inflammation might result from failure to maintain local lipid homeostasis and long-term effects on immune cells beyond the AT. Here we review the complex function of ATMs in AT homeostasis and metabolic inflammation. Additionally, we hypothesize that trained immunity, which involves long-term functional adaptations of myeloid cells and their bone marrow progenitors, can provide a model by which metabolic perturbations trigger chronic systemic inflammation.
Collapse
Affiliation(s)
- Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anthony W Ferrante
- Department of Medicine, Institute of Human Nutrition, Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA
| |
Collapse
|
8
|
Qian X, Meng X, Zhang S, Zeng W. Neuroimmune regulation of white adipose tissues. FEBS J 2022; 289:7830-7853. [PMID: 34564950 DOI: 10.1111/febs.16213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/21/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
The white adipose tissues (WAT) are located in distinct depots throughout the body. They serve as an energy reserve, providing fatty acids for other tissues via lipolysis when needed, and function as an endocrine organ to regulate systemic metabolism. Their activities are coordinated through intercellular communications among adipocytes and other cell types such as residential and infiltrating immune cells, which are collectively under neuronal control. The adipocytes and immune subtypes including macrophages/monocytes, eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2s), T and B cells, dendritic cells (DCs), and natural killer (NK) cells display cellular and functional diversity in response to the energy states and contribute to metabolic homeostasis and pathological conditions. Accumulating evidence reveals that neuronal innervations control lipid deposition and mobilization via regulating lipolysis, adipocyte size, and cellularity. Vice versa, the neuronal innervations and activity are influenced by cellular factors in the WAT. Though the literature describing adipose tissue cells is too extensive to cover in detail, we strive to highlight a selected list of neuronal and immune components in this review. The cell-to-cell communications and the perspective of neuroimmune regulation are emphasized to enlighten the potential therapeutic opportunities for treating metabolic disorders.
Collapse
Affiliation(s)
- Xinmin Qian
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xia Meng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Shan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
9
|
Puente-Ruiz SC, Jais A. Reciprocal signaling between adipose tissue depots and the central nervous system. Front Cell Dev Biol 2022; 10:979251. [PMID: 36200038 PMCID: PMC9529070 DOI: 10.3389/fcell.2022.979251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
In humans, various dietary and social factors led to the development of increased brain sizes alongside large adipose tissue stores. Complex reciprocal signaling mechanisms allow for a fine-tuned interaction between the two organs to regulate energy homeostasis of the organism. As an endocrine organ, adipose tissue secretes various hormones, cytokines, and metabolites that signal energy availability to the central nervous system (CNS). Vice versa, the CNS is a critical regulator of adipose tissue function through neural networks that integrate information from the periphery and regulate sympathetic nerve outflow. This review discusses the various reciprocal signaling mechanisms in the CNS and adipose tissue to maintain organismal energy homeostasis. We are focusing on the integration of afferent signals from the periphery in neuronal populations of the mediobasal hypothalamus as well as the efferent signals from the CNS to adipose tissue and its implications for adipose tissue function. Furthermore, we are discussing central mechanisms that fine-tune the immune system in adipose tissue depots and contribute to organ homeostasis. Elucidating this complex signaling network that integrates peripheral signals to generate physiological outputs to maintain the optimal energy balance of the organism is crucial for understanding the pathophysiology of obesity and metabolic diseases such as type 2 diabetes.
Collapse
|
10
|
Solernó LM, Sobol NT, Gottardo MF, Capobianco CS, Ferrero MR, Vásquez L, Alonso DF, Garona J. Propranolol blocks osteosarcoma cell cycle progression, inhibits angiogenesis and slows xenograft growth in combination with cisplatin-based chemotherapy. Sci Rep 2022; 12:15058. [PMID: 36075937 PMCID: PMC9458647 DOI: 10.1038/s41598-022-18324-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
Osteosarcoma is still associated with limited response to standard-of-care therapy and alarmingly elevated mortality rates, especially in low- and middle-income countries. Despite multiple efforts to repurpose β-blocker propranolol in oncology, its potential application in osteosarcoma management remains largely unexplored. Considering the unsatisfied clinical needs of this aggressive disease, we evaluated the antitumoral activity of propranolol using different in vitro and in vivo osteosarcoma preclinical models, alone or in addition to chemotherapy. Propranolol significantly impaired cellular growth in β2-adrenergic receptor-expressing MG-63 and U-2OS cells, and was capable of blocking growth-stimulating effects triggered by catecholamines. siRNA-mediated ADRB2 knockdown in MG-63 cells was associated with decreased cell survival and a significant attenuation of PPN anti-osteosarcoma activity. Direct cytostatic effects of propranolol were independent of apoptosis induction and were associated with reduced mitosis, G0/G1 cell cycle arrest and a significant down-regulation of cell cycle regulator Cyclin D1. Moreover, colony formation, 3D spheroid growth, cell chemotaxis and capillary-like tube formation were drastically impaired after propranolol treatment. Interestingly, anti-migratory activity of β-blocker was associated with altered actin cytoskeleton dynamics. In vivo, propranolol treatment (10 mg/kg/day i.p.) reduced the early angiogenic response triggered by MG-63 cells in nude mice. Synergistic effects were observed in vitro after combining propranolol with chemotherapeutic agent cisplatin. Sustained administration of propranolol (10 mg/kg/day i.p., five days a week), alone and especially in addition to low-dose metronomic cisplatin (2 mg/kg/day i.p., three times a week), markedly reduced xenograft progression. After histological analysis, propranolol and cisplatin combination resulted in low tumor mitotic index and increased tumor necrosis. β-blockade using propranolol seems to be an achievable and cost-effective therapeutic approach to modulate osteosarcoma aggressiveness. Further translational studies of propranolol repurposing in osteosarcoma are warranted.
Collapse
Affiliation(s)
- Luisina M Solernó
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - Natasha T Sobol
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - María F Gottardo
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - Carla S Capobianco
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina
| | - Maximiliano R Ferrero
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Biomedicine Research Institute of Buenos Aires (IBioBA), Buenos Aires, Argentina
| | - Liliana Vásquez
- Precision Medicine Research Center, School of Medicine, University of San Martín de Porres, Lima, Perú
| | - Daniel F Alonso
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina.,National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Juan Garona
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina. .,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina. .,National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
11
|
You D, Yu H, Wang Z, Wei X, Wu X, Pan C. The correlation of pericoronary adipose tissue with coronary artery disease and left ventricular function. BMC Cardiovasc Disord 2022; 22:398. [PMID: 36068548 PMCID: PMC9446702 DOI: 10.1186/s12872-022-02843-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/31/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE We sought to investigate the correlation of pericoronary adipose tissue with coronary artery disease and left ventricular (LV) function. METHODS Participants with clinically suspected coronary artery disease were enrolled. All participants underwent coronary computed tomography angiography (CCTA) and echocardiography followed by invasive coronary angiography (ICA) within 6 months. Pericoronary adipose tissue (PCAT) was extracted to analyze the correlation with the Gensini score and LV function parameters, including IVS, LVPW, LVEDD, LVESD, LVEDV, LVESV, FS, LVEF, LVM, and LVMI. The correlation between PCAT and the Gensini score was assessed using Spearman's correlation analysis, and that between the PCAT volume or FAI and LV function parameters was determined using partial correlation analysis. RESULTS One hundred and fifty-nine participants (mean age, 64.55 ± 10.64 years; men, 65.4% [104/159]) were included in the final analysis. Risk factors for coronary artery disease, such as hypertension, diabetes, dyslipidemia, and a history of smoking or drinking, had no significant association with PCAT (P > 0.05), and there was also no correlation between PCAT and the Gensini score. However, the LAD-FAI was positively correlated with the IVS (r = 0.203, P = 0.013), LVPW (r = 0.218, P = 0.008), LVEDD (r = 0.317, P < 0.001), LVESD (r = 0.298, P < 0.001), LVEDV (r = 0.317, P < 0.001), LVESV (r = 0.301, P < 0.001), LVM (r = 0.371, P < 0.001), and LVMI (r = 0.304, P < 0.001). Also, the LCX-FAI was positively correlated with the LVEDD (r = 0.199, P = 0.015), LVESD (r = 0.190, P = 0.021), LVEDV (r = 0.203, P = 0.013), LVESV (r = 0.197, P = 0.016), LVM (r = 0.220, P = 0.007), and LVMI (r = 0.172, P = 0.036), and the RCA-FAI was positively correlated with the LVEDD (r = 0.258, P = 0.002), LVESD (r = 0.238, P = 0.004), LVEDV (r = 0.266, P = 0.001), LVESV (r = 0.249, P = 0.002), LVM (r = 0.237, P = 0.004), and LVMI (r = 0.218, P = 0.008), respectively. Finally, the total volume was positively correlated with FS (r = 0.167, P = 0.042). CONCLUSION The FAI was positively correlated with the LV function but was not associated with the severity of coronary artery disease.
Collapse
Affiliation(s)
- Deshu You
- Department of Radiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China
| | - Haiyang Yu
- Department of Interventional and Vascular Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Zhiwei Wang
- Department of Radiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China
| | - Xiaoyu Wei
- Department of Radiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China
| | - Xiangxiang Wu
- Department of Radiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China
| | - Changjie Pan
- Department of Radiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
12
|
Liu SQ, Li B, Li JJ, Sun S, Sun SR, Wu Q. Neuroendocrine regulations in tissue-specific immunity: From mechanism to applications in tumor. Front Cell Dev Biol 2022; 10:896147. [PMID: 36072337 PMCID: PMC9442449 DOI: 10.3389/fcell.2022.896147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/27/2022] [Indexed: 11/26/2022] Open
Abstract
Immune responses in nonlymphoid tissues play a vital role in the maintenance of homeostasis. Lots of evidence supports that tissue-specific immune cells provide defense against tumor through the localization in different tissue throughout the body, and can be regulated by diverse factors. Accordingly, the distribution of nervous tissue is also tissue-specific which is essential in the growth of corresponding organs, and the occurrence and development of tumor. Although there have been many mature perspectives on the neuroendocrine regulation in tumor microenvironment, the neuroendocrine regulation of tissue-specific immune cells has not yet been summarized. In this review, we focus on how tissue immune responses are influenced by autonomic nervous system, sensory nerves, and various neuroendocrine factors and reversely how tissue-specific immune cells communicate with neuroendocrine system through releasing different factors. Furthermore, we pay attention to the potential mechanisms of neuroendocrine-tissue specific immunity axis involved in tumors. This may provide new insights for the immunotherapy of tumors in the future.
Collapse
Affiliation(s)
- Si-Qing Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juan-Juan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Sheng-Rong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- *Correspondence: Sheng-Rong Sun, ; Qi Wu,
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Sheng-Rong Sun, ; Qi Wu,
| |
Collapse
|
13
|
Freire BM, de Melo FM, Basso AS. Adrenergic Signaling Regulation Of Macrophage Function: Do We Understand It Yet? IMMUNOTHERAPY ADVANCES 2022; 2:ltac010. [PMID: 36284839 PMCID: PMC9585663 DOI: 10.1093/immadv/ltac010] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 05/30/2022] [Indexed: 11/30/2022] Open
Abstract
Macrophages are immune cells that are widespread throughout the body and critical for maintaining tissue homeostasis. Their remarkable plasticity allows them to acquire different phenotypes, becoming able either to fight infection (M1-like, classically activated macrophages) or to promote tissue remodeling and repair (M2-like, alternatively activated macrophages). These phenotypes are induced by different cues present in the microenvironment. Among the factors that might regulate macrophage activation are mediators produced by different branches of the nervous system. The regulation exerted by the sympathetic nervous system (SNS) on macrophages (and the immune system in general) is becoming a subject of increasing interest, indeed a great number of articles have been published lately. Catecholamines (noradrenaline and adrenaline) activate α and β adrenergic receptors expressed by macrophages and shape the effector functions of these cells in contexts as diverse as the small intestine, the lung, or the adipose tissue. Activation of different subsets of receptors seems to produce antagonistic effects, with α adrenergic receptors generally associated with pro-inflammatory functions and β adrenergic receptors (particularly β2) related to the resolution of inflammation and tissue remodeling. However, exceptions to this paradigm have been reported, and the factors contributing to these apparently contradictory observations are still far from being completely understood. Additionally, macrophages per se seem to be sources of catecholamines, which is also a subject of some debate. In this review, we discuss how activation of adrenergic receptors modulates macrophage effector functions and its implications for inflammatory responses and tissue homeostasis.
Collapse
Affiliation(s)
- Beatriz Marton Freire
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP) , São Paulo, Brazil
| | - Filipe Menegatti de Melo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP) , São Paulo, Brazil
| | - Alexandre S Basso
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP) , São Paulo, Brazil
| |
Collapse
|
14
|
Abstract
Neuronal innervation in the adipose tissues plays a crucial role in regulating adipose thermogenic capacity and metabolic homeostasis. The tissue-wide nerves display a large extent of structural plasticity under physiological and pathological conditions that alter the neuronal control of metabolic states. We find here that neuronal plasticity is regulated by immune cells, which constitutes an appealing way to reshape neural-controlled energy balance by targeting immune components. Sympathetic innervation regulates energy balance, and the nerve density in the adipose tissues changes under various metabolic states, resulting in altered neuronal control and conferring resilience to metabolic challenges. However, the impact of the immune milieu on neuronal innervation is not known. Here, we examined the regulatory role on nerve plasticity by eosinophils and found they increased cell abundance in response to cold and produced nerve growth factor (NGF) in the white adipose tissues (WAT). Deletion of Ngf from eosinophils or depletion of eosinophils impairs cold-induced axonal outgrowth and beiging process. The spatial proximity between sympathetic nerves, IL-33–expressing stromal cells, and eosinophils was visualized in both human and mouse adipose tissues. At the cellular level, the sympathetic adrenergic signal induced calcium flux in the stromal cells and subsequent release of IL-33, which drove the up-regulation of IL-5 from group 2 innate lymphoid cells (ILC2s), leading to eosinophil accretion. We propose a feed-forward loop between sympathetic activity and type 2 immunity that coordinately enhances sympathetic innervation and promotes energy expenditure.
Collapse
|
15
|
Meng X, Zeng W. Fat mesenchyme closes the neural-ILC2 circuit. Immunity 2021; 54:2191-2193. [PMID: 34644555 DOI: 10.1016/j.immuni.2021.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The immune milieu and neuronal activity each impact adipose metabolic health, yet their interplay remains largely undefined. In a recent issue of Nature, Cardoso et al. uncover a sympathetic neuro-mesenchymal-ILC2s circuit from brain-to-fat controlling obesity and glucose metabolism.
Collapse
Affiliation(s)
- Xia Meng
- Institute for Immunology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Wenwen Zeng
- Institute for Immunology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|