1
|
Boughanem H, de Larriva APA, Camargo A, Torres-Peña JD, Ojeda-Rodriguez A, Alcala-Diaz JF, Romero-Cabrera JL, Rangel-Zuñiga OA, Rodríguez-Cantalejo F, Soehnlein O, Macias-Gonzalez M, Tinahones FJ, Perez-Martinez P, Delgado-Lista J, López-Miranda J. Decreased Neutrophils Are Associated With Reduced Risk of Type 2 Diabetes Incidence: Results From the CORDIOPREV Study. J Clin Endocrinol Metab 2025; 110:1550-1558. [PMID: 39470387 DOI: 10.1210/clinem/dgae736] [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: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/25/2024] [Indexed: 10/30/2024]
Abstract
CONTEXT Numerous studies have reported an association between neutrophils and type 2 diabetes mellitus (T2DM), although this relationship remains unclear. OBJECTIVE To investigate the interaction of neutrophils and a dietary intervention on T2DM incidence after 60 months of follow-up. METHODS A comprehensive analysis was conducted on the framework of the CORDIOPREV study, which included 462 patients without T2DM at the beginning of the study, randomly assigned to either a Mediterranean or a low-fat diet; 107 developed T2DM. Absolute neutrophil counts and neutrophil-related ratios were measured. RESULTS Kaplan-Meier curves showed that the lowest tertile of basal neutrophils was associated with a reduced likelihood of T2DM incidence when compared to the middle (hazard ratio [HR] = 0.499 [95% CI, 0.287-0.866]) and the highest tertiles (HR = 0.442 [95% CI, 0.255-0.768]) in the overall population, after adjusting for clinical variables. This association only remained significant in patients who followed a Mediterranean diet when comparing the lowest to the middle (HR = 0.423 [95% CI, 0.213-0.842]) and the highest tertiles (HR = 0.371 [95% CI, 0.182-0.762]). The predictive capacity yielded an AUC of 0.711 (95% CI, 0.652-0.769), with neutrophils being the most important variable in the in the model. Decrease in neutrophils over the 60 months was associated with increased insulin sensitivity index (R = -0.31; P = .019), particularly in patients who followed the Mediterranean diet. CONCLUSION These findings suggest that monitoring neutrophils can help prevent the development of T2DM, as a reduction in neutrophil counts could be associated with improved insulin sensitivity. Following a Mediterranean diet might be a potential strategy to reduce the incidence of T2DM by lowering neutrophil levels. Further research is necessary to gain a deeper understanding regarding this mechanism.
Collapse
Affiliation(s)
- Hatim Boughanem
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Antonio Pablo Arenas de Larriva
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José D Torres-Peña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ana Ojeda-Rodriguez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan L Romero-Cabrera
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Oriol Alberto Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Oliver Soehnlein
- Institute of Experimental Pathology (ExPat), Center of Molecular Biology of Inflammation (ZMBE), University of Münster, 48149 Münster, Germany
| | - Manuel Macias-Gonzalez
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Malaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, 29590 Malaga, Spain
| | - Francisco J Tinahones
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Malaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, 29590 Malaga, Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José López-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, 14004 Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), 14004 Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
2
|
Loft A, Emont MP, Weinstock A, Divoux A, Ghosh A, Wagner A, Hertzel AV, Maniyadath B, Deplancke B, Liu B, Scheele C, Lumeng C, Ding C, Ma C, Wolfrum C, Strieder-Barboza C, Li C, Truong DD, Bernlohr DA, Stener-Victorin E, Kershaw EE, Yeger-Lotem E, Shamsi F, Hui HX, Camara H, Zhong J, Kalucka J, Ludwig JA, Semon JA, Jalkanen J, Whytock KL, Dumont KD, Sparks LM, Muir LA, Fang L, Massier L, Saraiva LR, Beyer MD, Jeschke MG, Mori MA, Boroni M, Walsh MJ, Patti ME, Lynes MD, Blüher M, Rydén M, Hamda N, Solimini NL, Mejhert N, Gao P, Gupta RK, Murphy R, Pirouzpanah S, Corvera S, Tang S, Das SK, Schmidt SF, Zhang T, Nelson TM, O'Sullivan TE, Efthymiou V, Wang W, Tong Y, Tseng YH, Mandrup S, Rosen ED. Towards a consensus atlas of human and mouse adipose tissue at single-cell resolution. Nat Metab 2025; 7:875-894. [PMID: 40360756 DOI: 10.1038/s42255-025-01296-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025]
Abstract
Adipose tissue (AT) is a complex connective tissue with a high relative proportion of adipocytes, which are specialized cells with the ability to store lipids in large droplets. AT is found in multiple discrete depots throughout the body, where it serves as the primary repository for excess calories. In addition, AT has an important role in functions as diverse as insulation, immunity and regulation of metabolic homeostasis. The Human Cell Atlas Adipose Bionetwork was established to support the generation of single-cell atlases of human AT as well as the development of unified approaches and consensus for cell annotation. Here, we provide a first roadmap from this bionetwork, including our suggested cell annotations for humans and mice, with the aim of describing the state of the field and providing guidelines for the production, analysis, interpretation and presentation of AT single-cell data.
Collapse
Affiliation(s)
- Anne Loft
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark.
| | - Margo P Emont
- Section of Endocrinology, Diabetes and Metabolism, University of Chicago, Chicago, IL, USA.
| | - Ada Weinstock
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Adhideb Ghosh
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Allon Wagner
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, The University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Babukrishna Maniyadath
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Boxiang Liu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular-Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Carey Lumeng
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Changhai Ding
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Chenkai Ma
- Human Health, Health and Biosecurity, CSIRO, Canberra, Australian Capital Territory, Australia
| | - Christian Wolfrum
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Clarissa Strieder-Barboza
- Department of Veterinary Sciences, Texas Tech University, Lubbock, TX, USA
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, USA
| | - Congru Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Danh D Truong
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, The University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | | | - Erin E Kershaw
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Farnaz Shamsi
- Department of Molecular Pathobiology, New York University, New York, NY, USA
- Departments of Cell Biology and Medicine, Grossman School of Medicine, New York University, New York, NY, USA
| | - Hannah X Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Henrique Camara
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Jiawei Zhong
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Joseph A Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Jutta Jalkanen
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Kyle D Dumont
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lindsey A Muir
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Lucas Massier
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Luis R Saraiva
- Sidra Medicine, Doha, Qatar
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Marc D Beyer
- Immunogenomics and Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Platform for Single Cell Genomics and Epigenomics (PRECISE), German Center for Neurodegenerative Diseases (DZNE) and University of Bonn and West German Genome Center (WGGC), Bonn, Germany
| | - Marc G Jeschke
- Centre for Burn Research, Hamilton Health Sciences Centre, Department of Surgery and Department of Biochemistry, McMaster University, Hamilton, Ontario, Canada
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Mariana Boroni
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Martin J Walsh
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary-Elizabeth Patti
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- Department of Medicine - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, Stockholm, Sweden
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | | | - Nicole L Solimini
- Department of Medical Oncology, Sarcoma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, Stockholm, Sweden
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Peng Gao
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rana K Gupta
- Department of Medicine, Division of Endocrinology, and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Saeed Pirouzpanah
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Silvia Corvera
- University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Su'an Tang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Swapan K Das
- Department of Internal Medicine, Section on Endocrinology and Metabolism, Medical Center Boulevard, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Søren F Schmidt
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark
| | - Tao Zhang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore M Nelson
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Vissarion Efthymiou
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Wenjing Wang
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yihan Tong
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark.
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Chang YH, Lee YC, Chen SH, Fang SY, Cheng TP, Chi CH, Tsai KC, Chen PJ, Hung HY. Discovery of a novel C2-functionalized chromen-4-one scaffold for the development of p38α MAPK signaling inhibitors to mitigate neutrophilic inflammatory responses. Biochem Pharmacol 2025; 235:116806. [PMID: 39956209 DOI: 10.1016/j.bcp.2025.116806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Neutrophil dysregulation is implicated in a spectrum of inflammatory pathologies, suggesting the potential for targeting neutrophilic hyperactivation as a pharmacological strategy to manage inflammatory disorders. Building upon prior research where 2-thiolphenoxychromone derivatives were found to inhibit neutrophilic generation of superoxide anions, this study focused on exploring the structure-activity relationship (SAR) of different C2 bridging moieties and anti-inflammatory effects using bioisosteric replacements and scaffold-hopping approaches. Among various chemotypes, the N-(4-oxo-4H-chromen-2-yl)benzenesulfonamide derivatives emerged as robust inhibitors of both superoxide anion generation and elastase release from fMLF-activated human neutrophils, with IC50 values in the single-digit micromolar range. Leveraging a forward pharmacology approach through computational prediction, compound 15b, a representative within this active molecular class, was discovered to exert these anti-inflammatory functions by blocking the p38α mitogen-activated protein kinase (MAPK) signaling cascade. This responded to a significant reduction in p38α MAPK and its downstream MK2 phosphorylation in activated neutrophils treated with 15b, with no apparent impact on extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and protein kinase B (AKT) phosphorylation levels. Additionally, this molecule exhibited inhibitory potential on intracellular reactive oxygen species (ROS) production, granule exocytosis, and chemotactic responses. Collectively, this study provides a novel skeleton for the development of inhibitors targeting the p38α MAPK pathway to mitigate neutrophilic inflammation.
Collapse
Affiliation(s)
- Yi-Han Chang
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Yi-Chen Lee
- Department of Nutrition Therapy, E-Da Cancer Hospital, I-Shou University, Kaohsiung 824, Taiwan; Department of Nutrition Therapy, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan; Department of Nutrition Therapy, E-Da Dachang Hospital, I-Shou University, Kaohsiung 824, Taiwan; Department of Nutrition, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan
| | - Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung 831, Taiwan
| | - Shu-Yen Fang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Tzu-Peng Cheng
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ching-Ho Chi
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan
| | - Keng-Chang Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan
| | - Po-Jen Chen
- Department of Medical Research, E-DA Hospital, I-Shou University, Kaohsiung 824, Taiwan; Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Hsin-Yi Hung
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
4
|
Wu Q, Yang Y, Lin S, Geller DA, Yan Y. The microenvironment in the development of MASLD-MASH-HCC and associated therapeutic in MASH-HCC. Front Immunol 2025; 16:1569915. [PMID: 40370443 PMCID: PMC12074932 DOI: 10.3389/fimmu.2025.1569915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a series of obesity-related metabolic liver diseases, ranging from relatively benign hepatic steatosis to metabolic-associated steatohepatitis (MASH). With the changes in lifestyle, its incidence and prevalence have risen to epidemic proportions globally. In recent years, an increasing amount of evidence has indicated that the hepatic microenvironment is involved in the pathophysiological processes of MASH-induced liver fibrosis and the formation of hepatocellular carcinoma (HCC). The hepatic microenvironment is composed of various parenchymal and non-parenchymal cells, which communicate with each other through various factors. In this review, we focus on the changes in hepatocytes, cholangiocytes, liver sinusoidal endothelial cells (LSECs), hepatic stellate cells (HSCs), Kupffer cells (KC), dendritic cells (DC), neutrophils, monocytes, T and B lymphocytes, natural killer cells (NK), natural killer T cells (NKT), mucosal-associated invariant T cells (MAIT), γδT cells, and gut microbiota during the progression of MASLD. Furthermore, we discuss promising therapeutic strategies targeting the microenvironment of MASLD-MASH-HCC.
Collapse
Affiliation(s)
- Qiulin Wu
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yan Yang
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shixun Lin
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - David A. Geller
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Yihe Yan
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
5
|
Lang F, Li Y, Yao R, Jiang M. Osteopontin in Chronic Inflammatory Diseases: Mechanisms, Biomarker Potential, and Therapeutic Strategies. BIOLOGY 2025; 14:428. [PMID: 40282293 PMCID: PMC12024743 DOI: 10.3390/biology14040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), atherosclerosis, and inflammatory bowel disease (IBD), pose major global health concerns. These disorders are marked by persistent inflammation, immune system dysfunction, tissue injury, and fibrosis, ultimately leading to severe organ dysfunction and diminished quality of life. Osteopontin (OPN), a multifunctional extracellular matrix protein, plays a crucial role in immune regulation, inflammation, and tissue remodeling. It promotes immune cell recruitment, stimulates pro-inflammatory cytokine production, and contributes to fibrosis through interactions with integrins and CD44 receptors. Additionally, OPN activates key inflammatory pathways, including NF-κB, MAPK, and PI3K/Akt, further aggravating tissue damage in chronic inflammatory conditions. Our review highlights the role of OPN in chronic inflammation, its potential as a biomarker, and its therapeutic implications. We explore promising preclinical approaches, such as monoclonal antibodies, small molecule inhibitors, and natural compounds like curcumin, which have demonstrated potential in mitigating OPN-driven inflammation. However, challenges persist in selectively targeting OPN while maintaining its essential physiological roles, including bone remodeling and wound healing. Our review offers insights into therapeutic strategies and future research directions.
Collapse
Affiliation(s)
- Fuyuan Lang
- Queen Mary College, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330001, China; (F.L.); (Y.L.); (R.Y.)
| | - Yuanheng Li
- Queen Mary College, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330001, China; (F.L.); (Y.L.); (R.Y.)
| | - Ruizhe Yao
- Queen Mary College, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330001, China; (F.L.); (Y.L.); (R.Y.)
| | - Meixiu Jiang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330031, China
| |
Collapse
|
6
|
Lin S, Li Y, Liu W, Du Y, Tao T. Metabolic Implications of Elevated Neutrophil Extracellular Traps in Polycystic Ovary Syndrome: A Focus on Hepatic Glycolysis. Biomolecules 2025; 15:572. [PMID: 40305335 PMCID: PMC12025135 DOI: 10.3390/biom15040572] [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: 01/23/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Polycystic ovary syndrome (PCOS) is a leading cause of infertility but also a metabolic disorder, frequently associated with obesity, insulin resistance, and diabetes. However, its etiology remains inadequately understood. Recent studies have increasingly implicated neutrophil extracellular traps (NETs) in the pathogenesis of metabolic diseases. Methods: Serum and follicular fluid samples were collected from patients with PCOS and control populations to assess NETs levels. The effects of NETs were investigated using DNase I to reduce NETs in dehydroepiandrosterone sulfate (DHEAS)-induced PCOS rats. Metabolic differences were further analyzed by untargeted metabolomics, and in vitro studies were conducted using primary bone marrow-derived neutrophils and normal mouse liver cell lines. Results: Markers of NETs in both the serum and follicular fluid of patients with PCOS were significantly higher than those in the control group. PCOS rats treated with DNase I exhibited significant improvements in glucose metabolism. Untargeted metabolomics analysis of liver tissue from these rats revealed alterations in the glycolysis pathway. Subsequent in vitro experiments demonstrated that treatment with NETs-conditioned medium (NETs CM) led to reduced insulin sensitivity, glucose uptake, and glucose utilization in liver cells, accompanied by varying degrees of decline in the transcription, translation, and function of glycolysis pathway proteins. Conclusions: NETs may be involved in the regulation of insulin resistance pathogenesis in PCOS by downregulating glycolytic pathways in the liver. Our study offers a novel strategy for insulin resistance intervention in PCOS.
Collapse
Affiliation(s)
- Siyu Lin
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (S.L.)
| | - Yushan Li
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (S.L.)
| | - Wei Liu
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (S.L.)
| | - Yanzhi Du
- Center for Reproductive Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Tao Tao
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (S.L.)
| |
Collapse
|
7
|
Zhao Y, Yue R. White adipose tissue in type 2 diabetes and the effect of antidiabetic drugs. Diabetol Metab Syndr 2025; 17:116. [PMID: 40186308 PMCID: PMC11969724 DOI: 10.1186/s13098-025-01678-9] [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: 08/20/2024] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
White adipose tissue (WAT) is highly flexible and was previously considered a passive location for energy storage. Its endocrine function has been established for several years, earning it the title of an "endocrine organ" due to its ability to secrete many adipokines that regulate metabolism. WAT is one of the core tissues that influence insulin sensitivity. Its dysfunction enhances insulin resistance and type 2 diabetes (T2D) progression. However, T2D may cause WAT dysfunction, including changes in distribution, metabolism, adipocyte hypertrophy, inflammation, aging, and adipokines and free fatty acid levels, which may exacerbate insulin resistance. This review used PubMed to search WAT dysfunction in T2D and the effects of these changes on insulin resistance. Additionally, we described and discussed the effects of antidiabetic drugs, including insulin therapy, sulfonylureas, metformin, glucose-like peptide-1 receptor agonists, thiazolidinediones, and sodium-dependent glucose transporters-2 inhibitors, on WAT parameters under T2D conditions.
Collapse
Affiliation(s)
- Yixuan Zhao
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu, University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan Province, 610072, P. R. China
| | - Rensong Yue
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu, University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan Province, 610072, P. R. China.
| |
Collapse
|
8
|
Tsilingiris D, Natsi A, Gavriilidis E, Antoniadou C, Eleftheriadou I, Anastasiou IA, Tentolouris A, Papadimitriou E, Eftalitsidis E, Kolovos P, Tsironidou V, Giatromanolaki A, Koffa M, Tentolouris N, Skendros P, Ritis K. Interleukin-8/Matrix Metalloproteinase-9 Axis Impairs Wound Healing in Type 2 Diabetes through Neutrophil Extracellular Traps-Fibroblast Crosstalk. Eur J Immunol 2025; 55:e202451664. [PMID: 40170410 PMCID: PMC11962236 DOI: 10.1002/eji.202451664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 04/03/2025]
Abstract
Neutrophils interact with and activate fibroblasts through the release of neutrophil extracellular traps (NETs). We investigated the role of NETs-fibroblast crosstalk in the cutaneous wound healing of type 2 diabetes (T2D). Neutrophils/NETs, serum, and primary human skin fibroblasts (HSFs) were obtained from individuals with T2D and age/sex-matched controls. NET-stimulation studies were performed on neutrophils/HSFs, with and without specific inhibitors, while HSF healing capacity was assessed using a scratch wound healing assay. T2D HSFs display a profibrotic phenotype, showing increased CCN2/CTGF, α-smooth muscle actin, and collagen release, albeit with impaired healing capacity, elevated type I collagen C-terminal telopeptide, and collagen degradation associated with increased (∼3.5-fold) matrix metalloproteinase-9 (MMP-9) in T2D neutrophils/NETs. IL-8 induced the expression of MMP-9 in neutrophils/NETs. Moreover, T2D neutrophils/NETs exhibited increased IL-8 content, which acted in an autocrine/paracrine fashion to further augment its production by neutrophils/HSFs. The findings were validated in normoglycemic individuals during a hyperglycemic clamp with concomitant lipid infusion and further corroborated immunohistochemically in diabetic plantar ulcer biopsies. This novel, vicious circle of NETs/interleukin-8/MMP-9/HSFs was hindered by IL-8 or MMP-9 blockade via specific inhibitors or by dismantling the NET-scaffold with DNase I, suggesting candidate therapeutic targets in wound healing impairment of T2D.
Collapse
Affiliation(s)
- Dimitrios Tsilingiris
- First Department of Internal MedicineUniversity Hospital of AlexandroupolisDemocritus University of ThraceAlexandroupolisGreece
- Laboratory of Molecular HematologyDepartment of MedicineDemocritus University of ThraceAlexandroupolisGreece
| | - Anastasia‐Maria Natsi
- Laboratory of Molecular HematologyDepartment of MedicineDemocritus University of ThraceAlexandroupolisGreece
| | - Efstratios Gavriilidis
- First Department of Internal MedicineUniversity Hospital of AlexandroupolisDemocritus University of ThraceAlexandroupolisGreece
- Laboratory of Molecular HematologyDepartment of MedicineDemocritus University of ThraceAlexandroupolisGreece
| | - Christina Antoniadou
- First Department of Internal MedicineUniversity Hospital of AlexandroupolisDemocritus University of ThraceAlexandroupolisGreece
- Laboratory of Molecular HematologyDepartment of MedicineDemocritus University of ThraceAlexandroupolisGreece
| | - Ioanna Eleftheriadou
- First Department of Propaedeutic Internal MedicineMedical SchoolNational and Kapodistrian University of AthensLaiko General HospitalAthensGreece
| | - Ioanna A. Anastasiou
- First Department of Propaedeutic Internal MedicineMedical SchoolNational and Kapodistrian University of AthensLaiko General HospitalAthensGreece
| | - Anastasios Tentolouris
- First Department of Propaedeutic Internal MedicineMedical SchoolNational and Kapodistrian University of AthensLaiko General HospitalAthensGreece
| | - Evangelos Papadimitriou
- First Department of Internal MedicineUniversity Hospital of AlexandroupolisDemocritus University of ThraceAlexandroupolisGreece
- Laboratory of Molecular HematologyDepartment of MedicineDemocritus University of ThraceAlexandroupolisGreece
| | - Evgenios Eftalitsidis
- Laboratory of Cell BiologyProteomics and Cell CycleDepartment of Molecular Biology and GeneticsDemocritus University of ThraceAlexandroupolisGreece
| | - Panagiotis Kolovos
- First Department of Internal MedicineUniversity Hospital of AlexandroupolisDemocritus University of ThraceAlexandroupolisGreece
| | - Victoria Tsironidou
- Laboratory of Molecular HematologyDepartment of MedicineDemocritus University of ThraceAlexandroupolisGreece
| | - Alexandra Giatromanolaki
- Department of PathologyUniversity Hospital of AlexandroupolisDemocritus University of ThraceAlexandroupolisGreece
| | - Maria Koffa
- Laboratory of Cell BiologyProteomics and Cell CycleDepartment of Molecular Biology and GeneticsDemocritus University of ThraceAlexandroupolisGreece
| | - Nikolaos Tentolouris
- First Department of Propaedeutic Internal MedicineMedical SchoolNational and Kapodistrian University of AthensLaiko General HospitalAthensGreece
| | - Panagiotis Skendros
- First Department of Internal MedicineUniversity Hospital of AlexandroupolisDemocritus University of ThraceAlexandroupolisGreece
- Laboratory of Molecular HematologyDepartment of MedicineDemocritus University of ThraceAlexandroupolisGreece
| | - Konstantinos Ritis
- First Department of Internal MedicineUniversity Hospital of AlexandroupolisDemocritus University of ThraceAlexandroupolisGreece
- Laboratory of Molecular HematologyDepartment of MedicineDemocritus University of ThraceAlexandroupolisGreece
| |
Collapse
|
9
|
Bowers E, Entrup GP, Islam M, Mohan R, Lerner A, Mancuso P, Moore BB, Singer K. High fat diet feeding impairs neutrophil phagocytosis, bacterial killing, and neutrophil-induced hematopoietic regeneration. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:680-693. [PMID: 40094316 PMCID: PMC12041776 DOI: 10.1093/jimmun/vkaf024] [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: 06/03/2024] [Revised: 01/08/2025] [Accepted: 02/01/2025] [Indexed: 03/19/2025]
Abstract
The prevalence of obesity and metabolic diseases have risen significantly over the past decades. Chronic inflammation in obesity is a link between obesity and secondary disease. While macrophages and monocytes are known to contribute to metabolic disease risk during diet exposure, little is known about the contribution of neutrophils. We assessed the impact of obesity on neutrophils using a 16-week model of diet-induced obesity. Bone marrow (BM) neutrophils significantly expanded with chronic high-fat diet (HFD), significantly decreased TNFɑ protein release, and impaired neutrophil regenerative function compared to normal diet (ND) neutrophils. scRNAseq and flow cytometry demonstrated HFD neutrophil heterogeneity and validated that these cells do not have elevated expression of proinflammatory genes without secondary stimulation. HFD neutrophils showed elevated expression of genes associated with lipid metabolism-acyl-CoA thioesterase 1 (Acot1), carnitine palmitoyltransferase 1a (Cpt1a), and perilipin 2 (Plin2). Consistent with the importance of lipid metabolism in driving dysfunction, neutrophils from HFD-fed animals and neutrophils treated with palmitate had impaired bacterial phagocytosis and killing responses. These data shed light on the complex regulation of intracellular lipids and the role of metabolism on neutrophil function during homeostasis and disease.
Collapse
Affiliation(s)
- Emily Bowers
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Gabrielle P Entrup
- Immunology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Mohammed Islam
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ramkumar Mohan
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Arianna Lerner
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Peter Mancuso
- Immunology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kanakadurga Singer
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
- Department Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Qu H, Yang Y, Xie Q, Ye L, Shao Y. Linear association of the dietary index for gut microbiota with insulin resistance and type 2 diabetes mellitus in U.S. adults: the mediating role of body mass index and inflammatory markers. Front Nutr 2025; 12:1557280. [PMID: 40191795 PMCID: PMC11968382 DOI: 10.3389/fnut.2025.1557280] [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: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Background Gut microbiota is reported to be related to the onset of insulin resistance (IR) and type 2 diabetes mellitus (T2DM). The dietary index for gut microbiota (DI-GM) is a novel index for reflecting gut microbiota diversity. We aimed to evaluate the association of DI-GM with T2DM and IR. Methods This cross-sectional research comprised 10,600 participants aged ≥20 from the National Health and Nutrition Examination Survey (NHANES) 2007-2018. We employed weighted multivariable linear and logistic regression models to examine the correlation of DI-GM with T2DM and IR. Linear or nonlinear relationships were examined by restricted cubic spline (RCS) regression. Additionally, subgroup and sensitivity analyses were performed to ensure the reliability of the results. Mediation analysis explored the roles of body mass index (BMI) and inflammatory factors in these associations. Results Higher DI-GM were inversely associated with T2DM (OR = 0.93, 95%CI: 0.89-0.98) and IR (OR = 0.95, 95%CI: 0.91-0.99) after adjusting for confounders. DI-GM ≥ 6 group showed significantly lower risks of T2DM (OR = 0.74, 95%CI: 0.60-0.91) and IR (OR = 0.77, 95%CI: 0.62-0.95). RCS demonstrated a linear relationship between DI-GM and T2DM, as well as IR. DI-GM was also inversely correlated with the risk markers of T2DM. Mediation analysis showed that BMI and the systemic inflammation response index partly mediated the association of DI-GM with T2DM and IR, while the systemic immune-inflammation index mediated only the association with T2DM. Conclusion DI-GM is inversely associated with T2DM and IR, with BMI and inflammatory markers partly mediating this association.
Collapse
Affiliation(s)
- Haoran Qu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyun Yang
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qihang Xie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liu Ye
- Department of Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Shao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Teo JMN, Chen W, Ling GS. Neutrophil plasticity in liver diseases. J Leukoc Biol 2025; 117:qiae222. [PMID: 39383213 DOI: 10.1093/jleuko/qiae222] [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: 09/08/2024] [Accepted: 10/09/2024] [Indexed: 10/11/2024] Open
Abstract
The liver has critical digestive, metabolic, and immunosurveillance roles, which get disrupted during liver diseases such as viral hepatitis, fatty liver disease, and hepatocellular carcinoma. While previous research on the pathological development of these diseases has focused on liver-resident immune populations, such as Kupffer cells, infiltrating immune cells responding to pathogens and disease also play crucial roles. Neutrophils are one such key population contributing to hepatic inflammation and disease progression. Belonging to the initial waves of immune response to threats, neutrophils suppress bacterial and viral spread during acute infections and have homeostasis-restoring functions, whereas during chronic insults, they display their plastic nature by responding to the inflammatory environment and develop new phenotypes alongside longer life spans. This review summarizes the diversity in neutrophil function and subpopulations present at steady state, during liver disease, and during liver cancer.
Collapse
Affiliation(s)
- Jia Ming Nickolas Teo
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Weixin Chen
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Guang Sheng Ling
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong, China
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Faculty Administration Wing, 21 Sassoon Road, Pokfulam, Hong Kong, China
- State Key Laboratory of Liver Research, The University of Hong Kong, HK Jockey Club Building for Interdisciplinary Research, 5 Sassoon Road, Pokfulam, Hong Kong, China
| |
Collapse
|
12
|
Gomez‐Casado G, Jimenez‐Gonzalez A, Rodriguez‐Muñoz A, Tinahones FJ, González‐Mesa E, Murri M, Ortega‐Gomez A. Neutrophils as indicators of obesity-associated inflammation: A systematic review and meta-analysis. Obes Rev 2025; 26:e13868. [PMID: 39610288 PMCID: PMC11791391 DOI: 10.1111/obr.13868] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/21/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024]
Abstract
INTRODUCTION The aim of this study is to evaluate and compare the suitability of routine blood neutrophil values as indicators of obesity-associated inflammation. METHODS In this systematic review and meta-analysis, we assess absolute neutrophil counts (ANCs) and neutrophil-to-lymphocyte ratio (NLR) values in subjects with and without obesity and analyze the weight of both parameters on the disease. Additionally, correlation studies between ANC and NLR with BMI, a parameter internationally accepted to define obesity are performed. RESULTS Quantitative data from 12 (ANC) and 11 (NLR) studies were included, with a total of 4475 participants. The meta-analysis shows that while both parameters are increased in the obesity group, ANC values present higher differences with the control and less heterogeneity among studies. Additionally, unlike NLR, ANC demonstrates a positive and significant correlation with BMI. CONCLUSION Overall, this meta-analysis demonstrates that ANC is a more reliable and stable parameter than NLR for the assessment of obesity-related inflammation, which offers clinicians a novel tool to assist in preventing complications related to obesity.
Collapse
Affiliation(s)
- Gema Gomez‐Casado
- Endocrinology and Nutrition UGCVirgen de la Victoria University HospitalMálagaSpain
- Biomedical Research Institute of Malaga ‐IBIMA Plataforma BIONANDMálagaSpain
- Department of Surgical Specialties, Biochemistry and Immunology Department, Faculty of MedicineUniversity of MalagaMálagaSpain
| | | | - Alba Rodriguez‐Muñoz
- Endocrinology and Nutrition UGCVirgen de la Victoria University HospitalMálagaSpain
- Biomedical Research Institute of Malaga ‐IBIMA Plataforma BIONANDMálagaSpain
| | - Francisco J. Tinahones
- Endocrinology and Nutrition UGCVirgen de la Victoria University HospitalMálagaSpain
- Biomedical Research Institute of Malaga ‐IBIMA Plataforma BIONANDMálagaSpain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos IIIMálagaSpain
- Department of Dermatology and Medicine, Faculty of MedicineUniversity of MalagaMálagaSpain
| | - Ernesto González‐Mesa
- Biomedical Research Institute of Malaga ‐IBIMA Plataforma BIONANDMálagaSpain
- Department of Surgical Specialties, Biochemistry and Immunology Department, Faculty of MedicineUniversity of MalagaMálagaSpain
- Obstetrics and Gynecology ServiceRegional University Hospital of MalagaMálagaSpain
| | - Mora Murri
- Endocrinology and Nutrition UGCVirgen de la Victoria University HospitalMálagaSpain
- Biomedical Research Institute of Malaga ‐IBIMA Plataforma BIONANDMálagaSpain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos IIIMálagaSpain
- Heart AreaVirgen de la Victoria University HospitalMálagaSpain
| | - Almudena Ortega‐Gomez
- Endocrinology and Nutrition UGCVirgen de la Victoria University HospitalMálagaSpain
- Biomedical Research Institute of Malaga ‐IBIMA Plataforma BIONANDMálagaSpain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos IIIMálagaSpain
| |
Collapse
|
13
|
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
|
14
|
Zinna L, Verde L, Tolla MFD, Barrea L, Parascandolo A, D'Alterio F, Colao A, Formisano P, D'Esposito V, Muscogiuri G. Chronodisruption enhances inflammatory cytokine release from visceral adipose tissue in obesity. J Transl Med 2025; 23:231. [PMID: 40011933 PMCID: PMC11863425 DOI: 10.1186/s12967-025-06250-7] [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: 01/24/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Chronodisruption, marked by circadian rhythm misalignment, is linked to inflammatory diseases like obesity. Chronotypes, reflecting individual circadian behavior, include morning, intermediate, and evening types, with evening chronotypes showing worse body composition and higher metabolic risk. This study evaluated the inflammatory profile of visceral adipose tissue (VAT) across chronotypes in individuals with obesity and examined clock gene expression. METHODS Twenty-five participants with obesity (11/14 F/M, BMI 41.59 ± 7.69 kg/m², age 41.13 ± 11.08 years) candidates for bariatric surgery were classified using the Morningness-Eveningness Questionnaire (MEQ): morning (36%), intermediate (28%), or evening (36%) chronotypes. VAT biopsies were analyzed for cytokines, chemokines, and growth factors via multiplex ELISA, and clock genes (PER1, CLOCK, BMAL1) were assessed using qPCR. RESULTS Body composition and biochemical parameters were similar across groups, but evening chronotypes had higher triglyceride levels (p = 0.012) and lower phase angle (p = 0.035). VAT inflammatory markers, including IL-1β (p = 0.04), IL-8 (p = 0.03), bFGF (p = 0.01), MCP-1 (p = 0.01), and MIP-1β (p = 0.05), were highest in evening and lowest in morning chronotypes. Evening chronotypes had significantly elevated bFGF levels compared to other groups (p = 0.04). PER1 mRNA expression was also higher in evening chronotypes (p = 0.02) and correlated with VAT-released bFGF (p = 0.03) and IL-1β (p = 0.03). MEQ scores negatively correlated with VAT bFGF (p = 0.02), MCP-1 (p = 0.02), and PER1 expressions. CONCLUSION Despite similar metabolic profiles, evening chronotypes exhibit heightened VAT inflammation and altered clock gene expression, potentially worsening their metabolic risk.
Collapse
Affiliation(s)
- Lorenza Zinna
- Department of Translational Medicine, University Federico II, Naples, Italy
| | - Ludovica Verde
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
| | | | - Luigi Barrea
- Dipartimento di Psicologia e Scienze della Salute, Centro Direzionale, Università Telematica Pegaso, Via Porzio, Isola F2, 80143, Naples, Italy
| | | | - Federica D'Alterio
- Institute for Experimental Endocrinology and Oncology "G. Salvatore"- National Research Council (IEOS-CNR), Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Unità di Endocrinologia, Diabetologia ed Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", Università degli Studi di Napoli Federico II, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, University Federico II, Naples, Italy
| | - Vittoria D'Esposito
- Institute for Experimental Endocrinology and Oncology "G. Salvatore"- National Research Council (IEOS-CNR), Naples, Italy
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA.
- Unità di Endocrinologia, Diabetologia ed Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", Università degli Studi di Napoli Federico II, Naples, Italy.
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia ed Andrologia, Università Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
15
|
He W, Yan L, Hu D, Hao J, Liou Y, Luo G. Neutrophil heterogeneity and plasticity: unveiling the multifaceted roles in health and disease. MedComm (Beijing) 2025; 6:e70063. [PMID: 39845896 PMCID: PMC11751288 DOI: 10.1002/mco2.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Neutrophils, the most abundant circulating leukocytes, have long been recognized as key players in innate immunity and inflammation. However, recent discoveries unveil their remarkable heterogeneity and plasticity, challenging the traditional view of neutrophils as a homogeneous population with a limited functional repertoire. Advances in single-cell technologies and functional assays have revealed distinct neutrophil subsets with diverse phenotypes and functions and their ability to adapt to microenvironmental cues. This review provides a comprehensive overview of the multidimensional landscape of neutrophil heterogeneity, discussing the various axes along which diversity manifests, including maturation state, density, surface marker expression, and functional polarization. We highlight the molecular mechanisms underpinning neutrophil plasticity, focusing on the complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications that shape neutrophil responses. Furthermore, we explore the implications of neutrophil heterogeneity and plasticity in physiological processes and pathological conditions, including host defense, inflammation, tissue repair, and cancer. By integrating insights from cutting-edge research, this review aims to provide a framework for understanding the multifaceted roles of neutrophils and their potential as therapeutic targets in a wide range of diseases.
Collapse
Affiliation(s)
- Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Dongxue Hu
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| |
Collapse
|
16
|
Albiero M, Baragetti A. Exploring neutrophils as therapeutic targets in cardiometabolic diseases. Trends Pharmacol Sci 2025; 46:102-116. [PMID: 39855946 DOI: 10.1016/j.tips.2024.12.003] [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: 05/24/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/27/2025]
Abstract
Current therapies for diabetes and atherosclerotic cardiovascular diseases (ACVDs) mainly target metabolic risk factors, but often fall short in addressing systemic inflammation, a key driver of disease onset and progression. Advances in our understanding of the biology of neutrophils, the cells that are principally involved in inflammatory situations, have highlighted their pivotal role in cardiometabolic diseases. Yet, neutrophils can reprogram their immune-metabolic functions based on the energetic substrates available, thus influencing both tissue homeostasis and the resolution of inflammation. In this review, we examine the effects of canonical therapies for cardiometabolic diseases on the key molecular pathways through which neutrophils respond to inflammatory stimuli. In addition, we explore potential synergies between these established therapeutic approaches and the anti-inflammatory therapies being evaluated for repurposing in the treatment of cardiometabolic diseases.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padua, Italy; Regional Center for the Cellular Therapy of Diabetes, University Hospital of Padova, Padua, Italy; Veneto Institute of Molecular Medicine, Laboratory of Experimental Diabetology, Padua, Italy.
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
17
|
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
|
18
|
Zhu Y, Shrestha A. Metabolic syndrome and its effect on immune cells in apical periodontitis- a narrative review. Clin Oral Investig 2025; 29:67. [PMID: 39825203 DOI: 10.1007/s00784-025-06161-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
OBJECTIVES Apical periodontitis (AP) is an inflammatory immune response in periapical tissues caused by microbial infections. Failure of root canal treatment or delayed healing is often due to intracanal or extra-radicular bacteria. However, beyond microbial factors, the patient's systemic health can significantly influence the progression and healing of AP. Metabolic syndrome is a risk factor and it is characterized by a cluster of interconnected metabolic risk factors, including abdominal obesity, hyperlipidemia, hypertension, and hyperglycemia. MATERIALS AND METHODS A comprehensive literature review was conducted on apical periodontitis and metabolic syndrome, and their impact on the roles of different immune cell populations. RESULTS Both AP and metabolic syndrome are inflammatory diseases that involve complex and interwoven immune responses. The affected immune cells are categorized into the innate (neutrophils, macrophages, and dendritic cells) and adaptive immune systems (T cells and B cells). CONCLUSIONS Metabolic diseases and AP are closely correlated, possibly intertwined in a two-way relationship driven by a shared dysregulated immune response. CLINICAL RELEVANCE Understanding the pathophysiology and immune mechanisms underlying the two-way relationship between metabolic syndrome and AP can help improve treatment outcomes and enhance the overall well-being of patients with endodontic disease complicated by metabolic syndrome.
Collapse
Affiliation(s)
- Yi Zhu
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada
| | - Annie Shrestha
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Department of Dentistry, Mt. Sinai Hospital, Toronto 412-600 University Avenue, Toronto, ON, M5G 1X5, Canada.
| |
Collapse
|
19
|
Zhu N, Li Y, Lin Y, Cui X, Li X. Association between neutrophil-to-high-density lipoprotein cholesterol ratio and non-alcoholic fatty liver disease or metabolic dysfunction-associated steatotic liver disease: evidence from NHANES 2017-2020. Front Med (Lausanne) 2025; 11:1491858. [PMID: 39882525 PMCID: PMC11774988 DOI: 10.3389/fmed.2024.1491858] [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: 09/05/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) or metabolic dysfunction-associated steatotic liver disease (MASLD) is closely associated with chronic inflammation and lipid metabolism disorders. The neutrophil-to-high-density lipoprotein cholesterol ratio (NHR) is an integrative marker reflecting inflammatory responses and lipid metabolism disorders and is associated with various diseases. This cross-sectional study aimed to determine the association between NHR and NAFLD, MASLD, and liver fibrosis. Methods Data for this study were obtained from the 2017-2020 National Health and Nutrition Examination Survey (NHANES), we employed weighted multiple regression and restricted cubic spline (RCS) analysis to assess the relationship between NHR and NAFLD, MASLD, and liver fibrosis. Additionally, we performed stratified analyses based on gender, age, body mass index, diabetes, hypertension, smoking status, and history of cardiovascular disease to evaluate the consistency of these associations across different subgroups. Results A total of 6,526 participants were included in the study. 2,839 (weighted 44.1%) participants were diagnosed with NAFLD and 2,813 (weighted 43.7%) participants were diagnosed with MASLD. After adjusting for confounders, NHR was positively associated with the risk of NAFLD/MASLD, and the correlation was particularly significant in the subgroups of females, those without hypertension, and those without diabetes (p < 0.05). By the NHR quartile, the risk of NAFLD/MASLD increased progressively with higher NHR levels (P for trend <0.001). In addition, RCS analysis showed a nonlinear association between NHR and NAFLD/MASLD and liver fibrosis (P-non-linear <0.05). Conclusion NHR may serve as a potential marker for NAFLD/MASLD and liver fibrosis, and lowering NHR levels could help reduce the incidence of these conditions.
Collapse
Affiliation(s)
- Na Zhu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yanyan Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yingying Lin
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing, China
| | - XinYu Cui
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xin Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing, China
| |
Collapse
|
20
|
Feješ A, Šebeková K, Borbélyová V. Pathophysiological Role of Neutrophil Extracellular Traps in Diet-Induced Obesity and Metabolic Syndrome in Animal Models. Nutrients 2025; 17:241. [PMID: 39861371 PMCID: PMC11768048 DOI: 10.3390/nu17020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The global pandemic of obesity poses a serious health, social, and economic burden. Patients living with obesity are at an increased risk of developing noncommunicable diseases or to die prematurely. Obesity is a state of chronic low-grade inflammation. Neutrophils are first to be recruited to sites of inflammation, where they contribute to host defense via phagocytosis, degranulation, and extrusion of neutrophil extracellular traps (NETs). NETs are web-like DNA structures of nuclear or mitochondrial DNA associated with cytosolic antimicrobial proteins. The primary function of NETosis is preventing the dissemination of pathogens. However, neutrophils may occasionally misidentify host molecules as danger-associated molecular patterns, triggering NET formation. This can lead to further recruitment of neutrophils, resulting in propagation and a vicious cycle of persistent systemic inflammation. This scenario may occur when neutrophils infiltrate expanded obese adipose tissue. Thus, NETosis is implicated in the pathophysiology of autoimmune and metabolic disorders, including obesity. This review explores the role of NETosis in obesity and two obesity-associated conditions-hypertension and liver steatosis. With the rising prevalence of obesity driving research into its pathophysiology, particularly through diet-induced obesity models in rodents, we discuss insights gained from both human and animal studies. Additionally, we highlight the potential offered by rodent models and the opportunities presented by genetically modified mouse strains for advancing our understanding of obesity-related inflammation.
Collapse
Affiliation(s)
| | - Katarína Šebeková
- Institute of Molecular Biomedicine, Medical Faculty, Comenius University, 83303 Bratislava, Slovakia; (A.F.); (V.B.)
| | | |
Collapse
|
21
|
Boyle LD, Miguelez-Crespo A, Paul M, Villalobos E, Toews JNC, Ivatt L, Nagy B, Magennis M, Homer NZM, Andrew R, Viau V, Hammond GL, Stimson RH, Walker BR, Nixon M. The NE/AAT/CBG axis regulates adipose tissue glucocorticoid exposure. Nat Commun 2025; 16:545. [PMID: 39788946 PMCID: PMC11718191 DOI: 10.1038/s41467-024-55693-x] [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: 09/12/2023] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
Corticosteroid binding globulin (CBG; SERPINA6) binds >85% of circulating glucocorticoids but its influence on their metabolic actions is unproven. Targeted proteolytic cleavage of CBG by neutrophil elastase (NE; ELANE) significantly reduces CBG binding affinity, potentially increasing 'free' glucocorticoid levels at sites of inflammation. NE is inhibited by alpha-1-antitrypsin (AAT; SERPINA1). Using complementary approaches in mice and humans to manipulate NE or AAT, we show high-fat diet (HFD) increases the NE:AAT ratio specifically in murine visceral adipose tissue, an effect only observed in males. Notably, HFD-fed male mice lacking NE have reduced glucocorticoid levels and action specifically in visceral adipose tissue, with improved glucose tolerance and insulin sensitivity, independent of systemic changes in free glucocorticoids. The protective effect of NE deficiency is lost when the adrenals are removed. Moreover, human asymptomatic heterozygous carriers of deleterious mutations in SERPINA1 resulting in lower AAT levels have increased adipose tissue glucocorticoid levels and action. However, in contrast to mice, humans present with systemic increases in free circulating glucocorticoid levels, an effect independent of HPA axis activation. These findings show that NE and AAT regulate local tissue glucocorticoid bioavailability in vivo, providing crucial evidence of a mechanism linking inflammation and metabolism.
Collapse
Affiliation(s)
- Luke D Boyle
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Mhairi Paul
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Elisa Villalobos
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Julia N C Toews
- Life Sciences Centre, University of British Columbia, Vancouver, Canada
| | - Lisa Ivatt
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Boglarka Nagy
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Marisa Magennis
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Natalie Z M Homer
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ruth Andrew
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Victor Viau
- Life Sciences Centre, University of British Columbia, Vancouver, Canada
| | | | - Roland H Stimson
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Brian R Walker
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Nixon
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
22
|
Zhang P, Watari K, Karin M. Innate immune cells link dietary cues to normal and abnormal metabolic regulation. Nat Immunol 2025; 26:29-41. [PMID: 39747429 PMCID: PMC12040443 DOI: 10.1038/s41590-024-02037-y] [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: 01/12/2024] [Accepted: 10/24/2024] [Indexed: 01/04/2025]
Abstract
A slew of common metabolic disorders, including type 2 diabetes, metabolic dysfunction-associated steatotic liver disease and steatohepatitis, are exponentially increasing in our sedentary and overfed society. While macronutrients directly impact metabolism and bioenergetics, new evidence implicates immune cells as critical sensors of nutritional cues and important regulators of metabolic homeostasis. A deeper interrogation of the intricate and multipartite interactions between dietary components, immune cells and metabolically active tissues is needed for a better understanding of metabolic regulation and development of new treatments for common metabolic diseases. Responding to macronutrients and micronutrients, immune cells play pivotal roles in interorgan communication between the microbiota, small intestine, metabolically active cells including hepatocytes and adipocytes, and the brain, which controls feeding behavior and energy expenditure. This Review focuses on the response of myeloid cells and innate lymphocytes to dietary cues, their cross-regulatory interactions and roles in normal and aberrant metabolic control.
Collapse
Affiliation(s)
- Peng Zhang
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kosuke Watari
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
23
|
Zhu B, Cao C, Liu W, Liu Y, Luo Y, Peng D. The predictive value of estimated glucose disposal rate for all-cause and cardiovascular mortality in the US non-diabetic population aged ≥60 years: A population-based cohort study. Diabetes Metab Syndr 2025; 19:103182. [PMID: 39721490 DOI: 10.1016/j.dsx.2024.103182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
AIMS This study seeks to evaluate the prognostic significance of eGDR in predicting mortality outcomes within non-diabetic older adults. METHODS 8131 non-diabetic participants aged ≥60 years from the National Health and Nutrition Examination Survey (2001-2018) was included in this study. eGDR was calculated as: eGDR (mg/kg/min) = 21.158 - [0.09 × waist circumference (cm)] - [3.407 × Hypertension (Yes = 1/No = 0)] - [0.551 × HbA1c (%)]. Weighted Cox proportional hazards models, cumulative hazard curves, restricted cubic spline (RCS), and threshold effects analyses were performed to explore the relationship between eGDR and mortality outcomes. Subgroup analyses and mediation effects analyses were conducted. RESULTS 2566 all-cause deaths and 689 cardiovascular deaths were recorded. Lower eGDR was associated with higher all-cause (HR = 0.76, 95 % CI: 0.63-0.91) and cardiovascular mortality (HR = 0.56, 95 % CI: 0.40-0.80). Inflection points were identified through RCS curve analyses, and the threshold effect was significant. The eGDR-mortality association remained consistent across subgroups. Mediation analyses showed that neutrophil to high-density lipoprotein cholesterol ratio mediated the association. CONCLUSIONS Lower eGDR levels are linked to higher risk of both all-cause and cardiovascular mortality in non-diabetic older adults, suggesting its potential utility for risk assessment among this population.
Collapse
Affiliation(s)
- Botao Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chenghui Cao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wenwu Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yuxuan Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yonghong Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
24
|
Altamura S, Lombardi F, Palumbo P, Cinque B, Ferri C, Del Pinto R, Pietropaoli D. The Evolving Role of Neutrophils and Neutrophil Extracellular Traps (NETs) in Obesity and Related Diseases: Recent Insights and Advances. Int J Mol Sci 2024; 25:13633. [PMID: 39769394 PMCID: PMC11727698 DOI: 10.3390/ijms252413633] [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: 11/15/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
Obesity is a chronic, multifactorial disease characterized by persistent low-grade tissue and systemic inflammation. Fat accumulation in adipose tissue (AT) leads to stress and dysfunctional adipocytes, along with the infiltration of immune cells, which initiates and sustains inflammation. Neutrophils are the first immune cells to infiltrate AT during high-fat diet (HFD)-induced obesity. Emerging evidence suggests that the formation and release of neutrophil extracellular traps (NETs) play a significant role in the progression of obesity and related diseases. Additionally, obesity is associated with an imbalance in gut microbiota and increased intestinal barrier permeability, resulting in the translocation of live bacteria, bacterial deoxyribonucleic acid (DNA), lipopolysaccharides (LPS), and pro-inflammatory cytokines into the bloodstream and AT, thereby contributing to metabolic inflammation. Recent research has also shown that short-chain fatty acids (SCFAs), produced by gut microbiota, can influence various functions of neutrophils, including their activation, migration, and the generation of inflammatory mediators. This review comprehensively summarizes recent advancements in understanding the role of neutrophils and NET formation in the pathophysiology of obesity and related disorders while also focusing on updated potential therapeutic approaches targeting NETs based on studies conducted in humans and animal models.
Collapse
Affiliation(s)
- Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Prevention and Translational Research—Dental Clinic, Center of Oral Diseases, 67100 L’Aquila, Italy
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
| | - Claudio Ferri
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Unit of Internal Medicine and Nephrology, San Salvatore Hospital, Center for Hypertension and Cardiovascular Prevention, 67100 L’Aquila, Italy
| | - Rita Del Pinto
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Unit of Internal Medicine and Nephrology, San Salvatore Hospital, Center for Hypertension and Cardiovascular Prevention, 67100 L’Aquila, Italy
| | - Davide Pietropaoli
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (F.L.); (P.P.); (B.C.); (C.F.); (R.D.P.)
- Prevention and Translational Research—Dental Clinic, Center of Oral Diseases, 67100 L’Aquila, Italy
| |
Collapse
|
25
|
Zhang F, Xia Y, Su J, Quan F, Zhou H, Li Q, Feng Q, Lin C, Wang D, Jiang Z. Neutrophil diversity and function in health and disease. Signal Transduct Target Ther 2024; 9:343. [PMID: 39638788 PMCID: PMC11627463 DOI: 10.1038/s41392-024-02049-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/21/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Neutrophils, the most abundant type of granulocyte, are widely recognized as one of the pivotal contributors to the acute inflammatory response. Initially, neutrophils were considered the mobile infantry of the innate immune system, tasked with the immediate response to invading pathogens. However, recent studies have demonstrated that neutrophils are versatile cells, capable of regulating various biological processes and impacting both human health and disease. Cytokines and other active mediators regulate the functional activity of neutrophils by activating multiple receptors on these cells, thereby initiating downstream signal transduction pathways. Dysfunctions in neutrophils and disruptions in neutrophil homeostasis have been implicated in the pathogenesis of numerous diseases, including cancer and inflammatory disorders, often due to aberrant intracellular signaling. This review provides a comprehensive synthesis of neutrophil biological functions, integrating recent advancements in this field. Moreover, it examines the biological roles of receptors on neutrophils and downstream signaling pathways involved in the regulation of neutrophil activity. The pathophysiology of neutrophils in numerous human diseases and emerging therapeutic approaches targeting them are also elaborated. This review also addresses the current limitations within the field of neutrophil research, highlighting critical gaps in knowledge that warrant further investigation. In summary, this review seeks to establish a comprehensive and multidimensional model of neutrophil regulation, providing new perspectives for potential clinical applications and further research.
Collapse
Affiliation(s)
- Fengyuan Zhang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yidan Xia
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiayang Su
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fushi Quan
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qiang Feng
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China.
| | - Ziping Jiang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China.
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
26
|
Li Q, Li Z, Guo S, Li S, Yao M, Li Y, Luo X. Increased circulating serpinB1 levels in children with overweight/obesity are associated with obesity-related parameters: a cross‑sectional study. BMC Pediatr 2024; 24:762. [PMID: 39578813 PMCID: PMC11585230 DOI: 10.1186/s12887-024-05251-7] [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: 06/17/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Circulating serpinB1 levels are increased in obese mice and have been shown to promote β-cell proliferation in several species. However, the data on serum serpinB1 levels in children with obesity are scarce. This study aimed to determine serum serpinB1 levels in children with overweight/obesity, and to study its association with obesity-related parameters. METHODS A total of 54 children with overweight/obesity and 36 normal-weight healthy controls aged 6-14 were recruited in this study. Anthropometric parameters, glucolipid metabolic biochemical parameters, sex hormones, and serum serpinB1 levels were measured in all subjects. The association of serum serpinB1 levels with obesity-related parameters and the risk of overweight/obesity were analyzed using correlation analysis and binary regression analysis, respectively. RESULTS The serum serpinB1 level in overweight/obese children was notably greater than in normal-weight controls (2.03 ± 0.70 vs. 1.41 ± 0.58 ng/mL, p < 0.001). SerpinB1 levels were positively correlated with body mass index (BMI), BMI Z-score, triglyceride (TG), uric acid, fasting insulin, C-peptide, and homeostasis model assessment of insulin resistance (HOMA-IR) levels. Additionally, we found that elevated circulating serpinB1 levels were associated with the increased risk of childhood overweight/obesity even after adjustment for age, gender, and HOMA-IR (odds ratio, 4.132; 95% confidence interval, 1.315-12.983; p = 0.015). CONCLUSIONS Circulating serpinB1 level was significantly increased in children with overweight/obesity and positively associated with obesity-related glucolipid metabolic parameters. These results indicate a close association between serum serpinB1 concentrations and childhood overweight/obesity.
Collapse
Affiliation(s)
- Qing Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuxi Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shusen Guo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sujuan Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minglan Yao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan, China.
| |
Collapse
|
27
|
Wang M, Min M, Duan H, Mai J, Liu X. The role of macrophage and adipocyte mitochondrial dysfunction in the pathogenesis of obesity. Front Immunol 2024; 15:1481312. [PMID: 39582861 PMCID: PMC11581950 DOI: 10.3389/fimmu.2024.1481312] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Obesity has emerged as a prominent global public health concern, leading to the development of numerous metabolic disorders such as cardiovascular diseases, type-2 diabetes mellitus (T2DM), sleep apnea and several system diseases. It is widely recognized that obesity is characterized by a state of inflammation, with immune cells-particularly macrophages-playing a significant role in its pathogenesis through the production of inflammatory cytokines and activation of corresponding pathways. In addition to their immune functions, macrophages have also been implicated in lipogenesis. Additionally, the mitochondrial disorders existed in macrophages commonly, leading to decreased heat production. Meantime, adipocytes have mitochondrial dysfunction and damage which affect thermogenesis and insulin resistance. Therefore, enhancing our comprehension of the role of macrophages and mitochondrial dysfunction in both macrophages and adipose tissue will facilitate the identification of potential therapeutic targets for addressing this condition.
Collapse
Affiliation(s)
- Min Wang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Min Min
- Outpatient Department, The Air Force Hospital of Western Theater, PLA, Chengdu, Sichuan, China
| | - Haojie Duan
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Jia Mai
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| | - Xiaojuan Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
28
|
Ji W, Li H, Qi Y, Zhou W, Chang Y, Xu D, Wei Y. Association between neutrophil-percentage-to-albumin ratio (NPAR) and metabolic syndrome risk: insights from a large US population-based study. Sci Rep 2024; 14:26646. [PMID: 39496695 PMCID: PMC11535182 DOI: 10.1038/s41598-024-77802-y] [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: 09/02/2024] [Accepted: 10/25/2024] [Indexed: 11/06/2024] Open
Abstract
Metabolic syndrome (MetS) is a cluster of conditions that increase the risk of cardiovascular disease and diabetes. This study aimed to investigate the association between Neutrophil-Percentage-to-Albumin Ratio (NPAR) and MetS in a large, nationally representative US population. We analyzed data from 28,178 participants in the National Health and Nutrition Examination Survey (NHANES) 2005-2018. Logistic regression models were used to evaluate the association between NPAR and MetS. Restricted cubic spline (RCS) models were employed to assess the dose-response relationship. Mediation analyses were conducted to explore potential mediating effects of serum uric acid and triglyceride-glucose (TyG) index. After adjusting for confounders, participants in the highest NPAR quartile had a 14% higher risk of MetS compared to those in the lowest quartile (OR 1.14, 95%CI 1.03-1.27, P = 0.010). RCS models revealed a monotonic increasing trend between NPAR and MetS risk (P for overall association = 0.002). Mediation analyses showed that serum uric acid and TyG index mediated 14.93% and 29.45% of the total effect of NPAR on MetS, respectively. Subgroup analyses indicated that the positive association between NPAR and MetS was more pronounced in Mexican Americans, individuals aged 20-65 years, those with lower income, males, current smokers, and moderate drinkers. Higher NPAR is associated with increased risk of MetS in the US adult population. This association is partially mediated by serum uric acid and TyG index. These findings suggest that NPAR may serve as a novel biomarker for MetS risk assessment and provide insights into potential mechanisms linking inflammation and metabolic disorders.
Collapse
Affiliation(s)
- Wei Ji
- The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130012, China
| | - Hongwei Li
- The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130012, China
| | - Yue Qi
- The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130012, China
| | - Wenshuo Zhou
- The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130012, China
| | - Yu Chang
- The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130012, China
| | - Dongsheng Xu
- The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130012, China.
| | - Yuxi Wei
- Pharmacy Department of Medical Security Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
29
|
Connolly BJ, Saxton SN. Recent updates on the influence of iron and magnesium on vascular, renal, and adipose inflammation and possible consequences for hypertension. J Hypertens 2024; 42:1848-1861. [PMID: 39258532 PMCID: PMC11451934 DOI: 10.1097/hjh.0000000000003829] [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: 02/06/2024] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
The inflammatory status of the kidneys, vasculature, and perivascular adipose tissue (PVAT) has a significant influence on blood pressure and hypertension. Numerous micronutrients play an influential role in hypertension-driving inflammatory processes, and recent reports have provided bases for potential targeted modulation of these micronutrients to reduce hypertension. Iron overload in adipose tissue macrophages and adipocytes engenders an inflammatory environment and may contribute to impaired anticontractile signalling, and thus a treatment such as chelation therapy may hold a key to reducing blood pressure. Similarly, magnesium intake has proven to greatly influence inflammatory signalling and concurrent hypertension in both healthy animals and in a model for chronic kidney disease, demonstrating its potential clinical utility. These findings highlight the importance of further research to determine the efficacy of micronutrient-targeted treatments for the amelioration of hypertension and their potential translation into clinical application.
Collapse
Affiliation(s)
- Benjamin J Connolly
- Divison of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | | |
Collapse
|
30
|
Jeon J, Lee SW, Park HJ, Park YH, Kim TC, Lee S, Lee S, Van Kaer L, Hong S. Overexpression of Chromatin Remodeling Factor SRG3 Down-Regulates IL1β-Expressing M1 Macrophages and IL17-Producing T Cells in Adipose Tissues. Int J Mol Sci 2024; 25:11681. [PMID: 39519233 PMCID: PMC11546064 DOI: 10.3390/ijms252111681] [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: 10/08/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The SWItch3-related gene (SRG3) is a core component of ATP-dependent SWI/SNF complexes, which are crucial for regulating immune cell development and function (e.g., macrophages and CD4+ T cells), embryonic development, and non-immune cell differentiation. Notably, SRG3 overexpression has been shown to polarize macrophages in the central nervous system toward an anti-inflammatory M2 phenotype, thereby protecting against the development of experimental autoimmune encephalomyelitis in mice. However, the effect of SRG3 on immune responses in adipose tissues remains unclear. To address this issue, we examined the cellularity and inflammatory status of adipose tissue in B10.PL mice overexpressing the SRG3 gene under the ubiquitous β-actin promoter (SRG3β-actin). Interestingly, SRG3 overexpression significantly reduced adipocyte size in both white and brown adipose tissues, without affecting the overall adipose tissue weight. Such phenotypic effects might be associated with the improved glucose tolerance observed in SRG3β-actin B10.PL mice. Moreover, we found that SRG3 overexpression down-regulates IL1β-expressing M1 macrophages, leading to a significant decrease in the M1/M2 macrophage ratio. Additionally, SRG3β-actin B10.PL mice showed a dramatic reduction in neutrophils as well as IL1β- and IL17-producing T cells in adipose tissues. Taken together, our results indicate that SRG3 plays a vital role in maintaining immune homeostasis within adipose tissues.
Collapse
Affiliation(s)
- Jungmin Jeon
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Sung Won Lee
- Department of Biomedical Laboratory Science, College of Health and Biomedical Services, Sangji University, Wonju 26339, Republic of Korea;
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Yun Hoo Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Tae-Cheol Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Sujin Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Seyeong Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Republic of Korea; (J.J.); (H.J.P.); (Y.H.P.); (T.-C.K.); (S.L.); (S.L.)
| |
Collapse
|
31
|
Ettel P, Weichhart T. Not just sugar: metabolic control of neutrophil development and effector functions. J Leukoc Biol 2024; 116:487-510. [PMID: 38450755 PMCID: PMC7617515 DOI: 10.1093/jleuko/qiae057] [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: 11/30/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
The mammalian immune system is constantly surveying our tissues to clear pathogens and maintain tissue homeostasis. In order to fulfill these tasks, immune cells take up nutrients to supply energy for survival and for directly regulating effector functions via their cellular metabolism, a process now known as immunometabolism. Neutrophilic granulocytes, the most abundant leukocytes in the human body, have a short half-life and are permanently needed in the defense against pathogens. According to a long-standing view, neutrophils were thought to primarily fuel their metabolic demands via glycolysis. Yet, this view has been challenged, as other metabolic pathways recently emerged to contribute to neutrophil homeostasis and effector functions. In particular during neutrophilic development, the pentose phosphate pathway, glycogen synthesis, oxidative phosphorylation, and fatty acid oxidation crucially promote neutrophil maturation. At steady state, both glucose and lipid metabolism sustain neutrophil survival and maintain the intracellular redox balance. This review aims to comprehensively discuss how neutrophilic metabolism adapts during development, which metabolic pathways fuel their functionality, and how these processes are reconfigured in case of various diseases. We provide several examples of hereditary diseases, in which mutations in metabolic enzymes validate their critical role for neutrophil function.
Collapse
Affiliation(s)
- Paul Ettel
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090Vienna, Austria
| | - Thomas Weichhart
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090Vienna, Austria
| |
Collapse
|
32
|
Esteves JV, Stanford KI. Exercise as a tool to mitigate metabolic disease. Am J Physiol Cell Physiol 2024; 327:C587-C598. [PMID: 38981607 PMCID: PMC11427015 DOI: 10.1152/ajpcell.00144.2024] [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: 03/12/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Metabolic diseases, notably obesity and type 2 diabetes (T2D), have reached alarming proportions and constitute a significant global health challenge, emphasizing the urgent need for effective preventive and therapeutic strategies. In contrast, exercise training emerges as a potent intervention, exerting numerous positive effects on metabolic health through adaptations to the metabolic tissues. Here, we reviewed the major features of our current understanding with respect to the intricate interplay between metabolic diseases and key metabolic tissues, including adipose tissue, skeletal muscle, and liver, describing some of the main underlying mechanisms driving pathogenesis, as well as the role of exercise to combat and treat obesity and metabolic disease.
Collapse
Affiliation(s)
- Joao Victor Esteves
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
33
|
Yu Y, Tan T, Yang W, Xu Z, Liu Y. Association between the systemic immune-inflammation index and obesity among adults: Insights from the NHANES 2017-2018. PLoS One 2024; 19:e0308288. [PMID: 39116149 PMCID: PMC11309425 DOI: 10.1371/journal.pone.0308288] [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: 03/20/2024] [Accepted: 07/21/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Inflammation is an important causative factor of obesity. This study aimed to explore the possible association between the systemic immune-inflammatory index, a novel indicator of inflammation, and obesity. METHODS Data were collected from 4395 participants of the National Health and Nutrition Examination Survey 2017-2018 aged ≥ 20 years. The systemic immune-inflammatory index was calculated by multiplying the platelet count by the neutrophil-to-lymphocyte ratio. Obesity was defined as a body mass index ≥ 30 kg/m2. RESULTS A significant positive correlation was observed between the systemic immune-inflammatory index and body mass index following multivariate linear regression analysis (β = 1.75; 95% confidence interval = 1.16-2.33), which was greatest in adults aged < 60 years without hypertension and diabetes. Smoothed curve fitting and threshold effect analysis were used to characterize the nonlinear association between the systemic immune-inflammatory index and body mass index, and the inflection point was found to be 729.3. CONCLUSIONS The systemic immune-inflammatory index is positively associated with body mass index among adults in the United States and has the potential to enhance efforts to prevent adult obesity.
Collapse
Affiliation(s)
- Yanmei Yu
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Tongcai Tan
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wei Yang
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhitao Xu
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yong Liu
- Department of Rehabilitation Medicine, Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Han XX, Zhang HY, Kong JW, Liu YX, Zhang KR, Ren WY. Systemic immune inflammation index is a valuable marker for predicting hemodialysis patients with depression: a cross-sectional study. Front Psychiatry 2024; 15:1423200. [PMID: 39161547 PMCID: PMC11331312 DOI: 10.3389/fpsyt.2024.1423200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Objective Maintenance hemodialysis (MHD) patients suffer from enormous physical, mental stress and poor quality of life, so an increasing number of patients are in a long-term state of depression. A prominent feature of MHD patients is chronic persistent inflammation, which is also an important mechanism for the onset of depression. Therefore, finding economically convenient inflammatory markers to predict and diagnose the onset of depression in MHD patients is of great value. As a novel inflammatory marker, systemic immune inflammation index (SII) can more comprehensively reflect the inflammation and immunity level of patients. This study aims to explore the relationship between SII and depressive symptoms in MHD patients. Methods A cross-sectional study was conducted on 206 MHD patients from three dialysis centers. Based on the Hospital Anxiety and Depression Scale (HADS) scores, patients were divided into non-depression and depression groups. Inter group comparison and multivariate logistic regression analysis were performed to determine whether SII is an independent risk factor for depression in MHD patients. Receiver operating characteristic (ROC) curve was used to evaluate the predictive value of SII on depression symptoms in MHD patients. Results According to the HADS scale score, 38.83% of the included patients were in a state of depression. After adjusting for all confounding factors, MHD patients with SII>963.93 had a 4.709 times higher risk of depression than those with SII ≤ 478.32 (OR=4.709, 95% CI 1.821-12.178, P<0.01). ROC analysis showed that SII>685.11 was the best cutoff value for MHD depression patients, and the area under the curve (AUC) was 0.681. Conclusions High SII is an independent risk factor for depressed MHD patients and an ideal inflammatory marker for predicting and identifying depression in MHD patients as assessed by the HADS scale.
Collapse
Affiliation(s)
- Xi-xi Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Nephrology Department, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, China
| | - Hui-ying Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Nephrology Department, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, China
| | - Jing-wen Kong
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu-xin Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ke-ren Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wen-ying Ren
- Nephrology Department, Beijing Integrated Traditional Chinese and Western Medicine Hospital, Beijing, China
| |
Collapse
|
35
|
Choi YJ, Kim Y, Hwang S. Role of Neutrophils in the Development of Steatotic Liver Disease. Semin Liver Dis 2024; 44:300-318. [PMID: 39117322 DOI: 10.1055/s-0044-1789207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
This review explores the biological aspects of neutrophils, their contributions to the development of steatotic liver disease, and their potential as therapeutic targets for the disease. Although alcohol-associated and metabolic dysfunction-associated liver diseases originate from distinct etiological factors, the two diseases frequently share excessive lipid accumulation as a common contributor to their pathogenesis, thereby classifying them as types of steatotic liver disease. Dysregulated lipid deposition in the liver induces hepatic injury, triggering the activation of the innate immunity, partially through neutrophil recruitment. Traditionally recognized for their role in microbial clearance, neutrophils have recently garnered attention for their involvement in sterile inflammation, a pivotal component of steatotic liver disease pathogenesis. In conclusion, technological innovations, including single-cell RNA sequencing, have gradually disclosed the existence of various neutrophil subsets; however, how the distinct subsets of neutrophil population contribute differentially to the development of steatotic liver disease remains unclear.
Collapse
Affiliation(s)
- You-Jin Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Republic of Korea
| | - Yeonsoo Kim
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
36
|
Yan K. Recent advances in the effect of adipose tissue inflammation on insulin resistance. Cell Signal 2024; 120:111229. [PMID: 38763181 DOI: 10.1016/j.cellsig.2024.111229] [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: 04/25/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
Obesity is one of the major risk factors for diabetes. Excessive accumulation of fat leads to inflammation of adipose tissue, which can increase the risk of developing diabetes. Obesity-related chronic inflammation can result in anomalies in glucose-lipid metabolism and insulin resistance, and it is a major cause of β-cell dysfunction in diabetes mellitus. Thus, a long-term tissue inflammatory response is crucial for metabolic diseases, particularly type 2 diabetes. Chronic inflammation associated with obesity increases oxidative stress, secretes inflammatory factors, modifies endocrine variables, and interferes with insulin signalling pathways, all of which contribute to insulin resistance and glucose tolerance. Insulin resistance and diabetes are ultimately caused by chronic inflammation in the stomach, pancreas, liver, muscle, and fat tissues. In this article, we systematically summarize the latest research progress on the mechanisms of adipose tissue inflammation and insulin resistance, as well as the mechanisms of cross-talk between adipose tissue inflammation and insulin resistance, with a view to providing some meaningful therapeutic strategies for the treatment of insulin resistance by controlling adipose tissue inflammation.
Collapse
Affiliation(s)
- Kaiyi Yan
- The Second Clinical College of China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
37
|
Shafiei-Jahani P, Yan S, Kazemi MH, Li X, Akbari A, Sakano K, Sakano Y, Hurrell BP, Akbari O. CB2 stimulation of adipose resident ILC2s orchestrates immune balance and ameliorates type 2 diabetes mellitus. Cell Rep 2024; 43:114434. [PMID: 38963763 PMCID: PMC11317174 DOI: 10.1016/j.celrep.2024.114434] [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: 01/30/2024] [Revised: 05/22/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
Development of type 2 diabetes mellitus (T2DM) is associated with low-grade chronic type 2 inflammation and disturbance of glucose homeostasis. Group 2 innate lymphoid cells (ILC2s) play a critical role in maintaining adipose homeostasis via the production of type 2 cytokines. Here, we demonstrate that CB2, a G-protein-coupled receptor (GPCR) and member of the endocannabinoid system, is expressed on both visceral adipose tissue (VAT)-derived murine and human ILC2s. Moreover, we utilize a combination of ex vivo and in vivo approaches to explore the functional and therapeutic impacts of CB2 engagement on VAT ILC2s in a T2DM model. Our results show that CB2 stimulation of ILC2s protects against insulin-resistance onset, ameliorates glucose tolerance, and reverses established insulin resistance. Our mechanistic studies reveal that the therapeutic effects of CB2 are mediated through activation of the AKT, ERK1/2, and CREB pathways on ILC2s. The results reveal that the CB2 agonist can serve as a candidate for the prevention and treatment of T2DM.
Collapse
Affiliation(s)
- Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shi Yan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Mohammad H Kazemi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amitis Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kei Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yoshihiro Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
38
|
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
|
39
|
Singal AK, Shah VH, Malhi H. Emerging targets for therapy in ALD: Lessons from NASH. Hepatology 2024; 80:223-237. [PMID: 36938877 PMCID: PMC10511666 DOI: 10.1097/hep.0000000000000381] [Citation(s) in RCA: 2] [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: 12/26/2022] [Accepted: 03/09/2023] [Indexed: 03/21/2023]
Abstract
Alcohol-associated liver disease due to harmful alcohol use and NAFLD associated with metabolic syndrome are the 2 most common liver diseases worldwide. Control of respective risk factors is the cornerstone in the long-term management of these diseases. Furthermore, there are no effective therapies. Both diseases are characterized by metabolic derangements; thus, the focus of this review was to broaden our understanding of metabolic targets investigated in NAFLD, and how these can be applied to alcohol-associated liver disease. Conserved pathogenic pathways such as dysregulated lipid metabolism, cell death pathways including apoptosis and activation of innate immune cells, and stellate cells mediate both alcohol and NAFLDs, resulting in histological abnormalities of steatosis, inflammation, fibrosis, and cirrhosis. However, pathways such as gut microbiome changes, glucose metabolism and insulin resistance, inflammatory signaling, and microRNA abnormalities are distinct in these 2 diseases. In this review article, we describe conserved and distinct pathogenic pathways highlighting therapeutic targets that may be of potential in both diseases and those that are unique to each disease.
Collapse
Affiliation(s)
- Ashwani K. Singal
- Department of Internal Medicine, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
- Division of Gastroenterology and Hepatology, Avera Transplant Institute, Sioux Falls, South Dakota, USA
- VA Medical Center, Sioux Falls, South Dakota, USA
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
40
|
Aydın MS, Eren MA, Uyar N, Kankılıç N, Karaaslan H, Sabuncu T, Çelik H. Relationship between systemic immune inflammation index and amputation in patients with diabetic foot ulcer. J Orthop Sci 2024; 29:1060-1063. [PMID: 37532650 DOI: 10.1016/j.jos.2023.07.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/04/2023]
Abstract
AIM The systemic immune inflammation index (SII) is a cost-effective biomarker calculated by lymphocyte, neutrophil and platelet counts and is currently being studied in various diseases. Since there is no study examining the relationship between SII and diabetic foot ulcers (DFU) in the literature, our aim was to investigate the relationship between SII and amputation rate in DFU. METHODS Type 2 DM 511 patients with DFU were screened from 2017 to 2021. Laboratory data obtained on the first day of hospitalization were considered. Platelet-to-lymphocyte ratio (PLR), neutrophil-to-lymphocyte ratio (NLR) and SII were calculated from routine blood count. Participants were divided into two groups as amputation (Group 1) and non-amputation (Group 2). RESULTS Amputation rate was 18.8%. The A1c (8.80 (3.26) % vs. 9.52 (3.10) %, p = 0.007) and HGB (10.17 ± 2.16 g/dL vs. 12.05 ± 2.20 g/dL, p < 0.001) levels, and lymphocyte count (1.81 (1.16) vs. 2.05 (1.11), p = 0.015) were significantly lower in Group 1 than Group 2. The counts of WBC (14.01 (9.16) × 109/L vs. 10.41 (5.82) × 109/L), PLT (393.35 (196.98) × 109/L vs. 312.05 (141.33) × 109/L), neutrophil (11.52 (8.75) × 109/L vs. 6.93 (5.96) × 109/L), PLR (226.04 (159.24) × 109/L vs. 153.12 (101.91) × 109/L), NLR (6.64 (6.93) vs. 3.34 (3.99)) and SII (2505.86 (3957.47) × 109/L vs. 1092.50 (1476.08) × 109/L), and the levels of CRP (14.12 (12.66) mg/dL vs. 3.86 (12.63) mg/dL) and ESR (87.50 (50.50) mm/h vs. 63.00 (57.25) mm/h) were significantly higher in Group 1 than Group 2 (all p < 0.001). AUC of ROC analysis of PLR was 0.666 (95% CI, 0.604-0.728), NLR was 0.695 (95% CI, 0.638-0.752) and SII was 0.716 (95% CI, 0.661-0.772) for the predicting of amputation and the SII had the best AUC with 67.4% sensitivity and 63.3%specificty. CONCLUSION SII is a cost-effective and readily available marker, but alone may not be sufficient to predict the risk of amputation in DFU. In our results, the predictive role of SII alone or with other markers for future DFU and its role in predicting other chronic diabetic complications will be evaluated in extensive studies.
Collapse
Affiliation(s)
- Mehmet Salih Aydın
- Cardioavascular Surgery Department, Harran University Faculty of Medicine, Sanliurfa, Turkey.
| | - Mehmet Ali Eren
- Endokrinology Department, Harran University Faculty of Medicine, Sanliurfa, Turkey
| | - Nida Uyar
- Endokrinology Department, Harran University Faculty of Medicine, Sanliurfa, Turkey
| | - Nazım Kankılıç
- Cardioavascular Surgery Department, Harran University Faculty of Medicine, Sanliurfa, Turkey
| | - Hüseyin Karaaslan
- Endokrinology Department, Harran University Faculty of Medicine, Sanliurfa, Turkey
| | - Tevfik Sabuncu
- Endokrinology Department, Harran University Faculty of Medicine, Sanliurfa, Turkey
| | - Hakim Çelik
- Biochemistry Department, Harran University Faculty of Medicine, Sanliurfa, Turkey
| |
Collapse
|
41
|
Shantaram D, Hoyd R, Blaszczak AM, Antwi L, Jalilvand A, Wright VP, Liu J, Smith AJ, Bradley D, Lafuse W, Liu Y, Williams NF, Snyder O, Wheeler C, Needleman B, Brethauer S, Noria S, Renton D, Perry KA, Nagareddy P, Wozniak D, Mahajan S, Rana PSJB, Pietrzak M, Schlesinger LS, Spakowicz DJ, Hsueh WA. Obesity-associated microbiomes instigate visceral adipose tissue inflammation by recruitment of distinct neutrophils. Nat Commun 2024; 15:5434. [PMID: 38937454 PMCID: PMC11211470 DOI: 10.1038/s41467-024-48935-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/17/2024] [Indexed: 06/29/2024] Open
Abstract
Neutrophils are increasingly implicated in chronic inflammation and metabolic disorders. Here, we show that visceral adipose tissue (VAT) from individuals with obesity contains more neutrophils than in those without obesity and is associated with a distinct bacterial community. Exploring the mechanism, we gavaged microbiome-depleted mice with stool from patients with and without obesity during high-fat or normal diet administration. Only mice receiving high-fat diet and stool from subjects with obesity show enrichment of VAT neutrophils, suggesting donor microbiome and recipient diet determine VAT neutrophilia. A rise in pro-inflammatory CD4+ Th1 cells and a drop in immunoregulatory T cells in VAT only follows if there is a transient spike in neutrophils. Human VAT neutrophils exhibit a distinct gene expression pattern that is found in different human tissues, including tumors. VAT neutrophils and bacteria may be a novel therapeutic target for treating inflammatory-driven complications of obesity, including insulin resistance and colon cancer.
Collapse
Affiliation(s)
- Dharti Shantaram
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - Rebecca Hoyd
- Pelotonia Institute for Immuno-Oncology at The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, 43210, USA
| | - Alecia M Blaszczak
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - Linda Antwi
- Pelotonia Institute for Immuno-Oncology at The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, 43210, USA
| | - Anahita Jalilvand
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - Valerie P Wright
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - Joey Liu
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - Alan J Smith
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA
| | - William Lafuse
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - YunZhou Liu
- Pelotonia Institute for Immuno-Oncology at The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, 43210, USA
| | - Nyelia F Williams
- Pelotonia Institute for Immuno-Oncology at The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, 43210, USA
| | - Owen Snyder
- Pelotonia Institute for Immuno-Oncology at The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, 43210, USA
| | - Caroline Wheeler
- Pelotonia Institute for Immuno-Oncology at The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, 43210, USA
| | - Bradley Needleman
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University, Columbus, OH, 43210, USA
| | - Stacy Brethauer
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University, Columbus, OH, 43210, USA
| | - Sabrena Noria
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University, Columbus, OH, 43210, USA
| | - David Renton
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University, Columbus, OH, 43210, USA
| | - Kyle A Perry
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University, Columbus, OH, 43210, USA
| | - Prabha Nagareddy
- Department of Internal Medicine, Cardiovascular Section University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, 73117, USA
| | - Daniel Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Sahil Mahajan
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Pranav S J B Rana
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Larry S Schlesinger
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Daniel J Spakowicz
- Pelotonia Institute for Immuno-Oncology at The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, 43210, USA.
| | - Willa A Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center at The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
42
|
Gavin KM, Conrad B, Sullivan T, Vianzon R, Acosta AS, Kohrt WM, Klemm DJ. Adipogenic differentiation of hematopoietic lineage cells isolated from adipose tissue of humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.22.600230. [PMID: 38979365 PMCID: PMC11230218 DOI: 10.1101/2024.06.22.600230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
We previously discovered some adipocytes in the major white fat depots of mice and humans arise from bone marrow-derived cells of hematopoietic lineage rather than conventional mesenchymal precursors, termed bone marrow-derived adipocytes (BMDA). Here we aimed to determine if hematopoietic lineage cells isolated from adipose tissue and circulation of humans could undergo adipogenic differentiation in vitro, thereby establishing an in vitro model for studies of BMDA. We hypothesized that hematopoietic lineage cells isolated from adipose tissue, but not circulation, of humans would demonstrate adipogenic potential. Participants included younger (20-50 years) and older (>50-75 years) men and women, BMI 20-37 kg/m2. Subcutaneous abdominal adipose tissue biopsies were obtained and stromal cell populations identified by flow cytometry. Sorted cells underwent in vitro cultivation via traditional mesenchymal culture methodology (mesenchymal lineage) or a novel 3D-fibrin clot followed by traditional adherent culture (hematopoietic lineage) for assessment of proliferation and differentiation capacity. We found hematopoietic lineage cells isolated from the adipose tissue stroma, but not the circulation, were capable of proliferation and multilineage (adipogenic and osteogenic) differentiation in vitro. We provide a new investigative tool that can be used to perform translational studies of BMDAs and provide initial evidence that hematopoietic lineage cells isolated from the adipose tissue of humans can undergo hematopoietic-to-mesenchymal transition with multilineage differentiation potential in an in vitro environment.
Collapse
Affiliation(s)
- Kathleen M Gavin
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado Geriatric Research, Education and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045 USA
| | - Bogdan Conrad
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy Sullivan
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado Geriatric Research, Education and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045 USA
| | - Ruby Vianzon
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Alistaire S Acosta
- University of Colorado Cancer Center Flow Cytometry Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wendy M Kohrt
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado Geriatric Research, Education and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045 USA
| | - Dwight J Klemm
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Eastern Colorado Geriatric Research, Education and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045 USA
| |
Collapse
|
43
|
Ogino N, Leite MF, Guerra MT, Kruglov E, Asashima H, Hafler DA, Ito T, Pereira JP, Peiffer BJ, Sun Z, Ehrlich BE, Nathanson MH. Neutrophils insert elastase into hepatocytes to regulate calcium signaling in alcohol-associated hepatitis. J Clin Invest 2024; 134:e171691. [PMID: 38916955 PMCID: PMC11324315 DOI: 10.1172/jci171691] [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: 04/24/2023] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
Neutrophil infiltration occurs in a variety of liver diseases, but it is unclear how neutrophils and hepatocytes interact. Neutrophils generally use granule proteases to digest phagocytosed bacteria and foreign substances or neutralize them in neutrophil extracellular traps. In certain pathological states, granule proteases play a destructive role against the host as well. More recently, nondestructive actions of neutrophil granule proteins have been reported, such as modulation of tissue remodeling and metabolism. Here, we report a completely different mechanism by which neutrophils act nondestructively, by inserting granules directly into hepatocytes. Specifically, elastase-containing granules were transferred to hepatocytes where elastase selectively degraded intracellular calcium channels to reduce cell proliferation without cytotoxicity. In response, hepatocytes increased expression of Serpin E2 and A3, which inhibited elastase activity. Elastase insertion was seen in patient specimens of alcohol-associated hepatitis, and the relationship between elastase-mediated ITPR2 degradation and reduced cell proliferation was confirmed in mouse models. Moreover, neutrophils from patients with alcohol-associated hepatitis were more prone to degranulation and more potent in reducing calcium channel expression than neutrophils from healthy individuals. This nondestructive and reversible action on hepatocytes defines a previously unrecognized role for neutrophils in the transient regulation of epithelial calcium signaling mechanisms.
Collapse
Affiliation(s)
- Noriyoshi Ogino
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - M. Fatima Leite
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
- INCT - NanoBiofar – Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mateus T. Guerra
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emma Kruglov
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | - Takeshi Ito
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - João P. Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Brandon J. Peiffer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barbara E. Ehrlich
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Michael H. Nathanson
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
44
|
Liu X, Peng G, Liu R, Zang X, Zou C, Sun H, Zhu Q, Geng H, Liang J. Follow-up study to explore the relationship between Neutrophil to lymphocyte ratio and impaired fasting glucose-using the group-based trajectory modeling. Sci Rep 2024; 14:14064. [PMID: 38890369 PMCID: PMC11189411 DOI: 10.1038/s41598-024-64701-5] [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: 03/03/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Previous studies have indicated a link between neutrophil to lymphocyte ratio (NLR) and impaired fasting glucose (IFG), but the findings have been disputed. By conducting a real-world follow-up study, we can monitor the development of diseases and confirm the connection between NLR and IFG. A total of 1168 patients without IFG or T2DM were followed up for six years. At baseline, participants' NLR levels, fasting plasma glucose and other clinical characteristics were recorded. During the follow-up period, NLR levels and the prevalence of IFG were recorded. Ultimately, 45 individuals were lost to follow-up, leaving 1,123 participants for analysis. Using Group-Based Trajectory Modeling (GBTM), the sample was divided into three groups. The prevalence of IFG in the three groups was 12.1%, 19.4%, and 20.85%, respectively. Compared with the low-level NLR group, the hazard ratio of IFG in the moderate-level NLR group and high-level NLR group were 1.628 (1.109-2.390) and 1.575 (1.001-2.497), respectively. There was a significant interaction effect of BMI and NLR on the risk of IFG (P < 0.001). In this real-world follow-up study, we observed a positive association between NLR and the risk of IFG, with this relationship being exacerbated by obesity status.
Collapse
Affiliation(s)
- Xuekui Liu
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Medical University Affiliated Xuzhou Clinical Collage, Xuzhou, Jiangsu, China
| | | | - Ran Liu
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiu Zang
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Medical University Affiliated Xuzhou Clinical Collage, Xuzhou, Jiangsu, China
| | - Caiyan Zou
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Medical University Affiliated Xuzhou Clinical Collage, Xuzhou, Jiangsu, China
| | - Haojie Sun
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Medical University Affiliated Xuzhou Clinical Collage, Xuzhou, Jiangsu, China
| | - Qian Zhu
- Quanshan Taishan Community Hospital, Xuzhou, Jiangsu, China.
| | - Houfa Geng
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Medical University Affiliated Xuzhou Clinical Collage, Xuzhou, Jiangsu, China.
| | - Jun Liang
- Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
45
|
Liu F, Wang T, Wang S, Zhao X, Hua Y. The association of platelet to white blood cell ratio with diabetes: a nationwide survey in China. Front Endocrinol (Lausanne) 2024; 15:1418583. [PMID: 38957446 PMCID: PMC11217324 DOI: 10.3389/fendo.2024.1418583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/27/2024] [Indexed: 07/04/2024] Open
Abstract
Background Inflammation is integral to diabetes pathogenesis. The novel hematological inflammatory biomarker, platelet to white blood cell ratio (PWR), is linked with various conditions such as chronic kidney disease and stroke. However, the association of this novel clinical indicator with diabetes still remains unclear, which is investigated in this study. Materials and Methods A total of 10,973 Chinese participants were included and grouped according to the tertiles of PWR (T1, T2, and T3 groups). Diagnosis of prediabetes and diabetes adhered to American Diabetes Association criteria. Binary logistic regression was adopted to assess the relationship between PWR and both diabetes and prediabetes. The dose-response relationship of PWR and diabetes was examined using restricted cubic spline regression. Subgroup and interaction analyses were conducted to investigate potential covariate interactions. Results Individuals with higher PWR had better lifestyles and lipid profiles (all P < 0.05). After adjusting for all the covariates, the T2 group had a 0.83-fold (95% CI: 0.73-0.93, P < 0.01) risk of diabetes and that for the T3 group was 0.68-fold (95% CI: 0.60-0.78. P < 0.001). Dose-response analysis identified non-linear PWR-diabetes associations in the general population and females (both P < 0.05), but absent in males. Participants with prediabetes in the T2 and T3 groups had lower risks of diabetes (OR = 0.80 for the T2 group, P < 0.001 and 0.68 for the T3 group, P < 0.001) in the full models. All the sensitivity analysis support consistent conclusions. Conclusions An increase in PWR significantly correlates with reduced diabetes risks. A non-linear PWR-diabetes relationship exists in the general population and females, but not in males. The correlation between PWR and diabetes indicates that PWR holds potentials in early identification and prevention of diabetes.
Collapse
Affiliation(s)
- Fanglin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Tianhong Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Siman Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiumei Zhao
- Operating Room, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yusi Hua
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Yang Z, Chen F, Zhang Y, Ou M, Tan P, Xu X, Li Q, Zhou S. Therapeutic targeting of white adipose tissue metabolic dysfunction in obesity: mechanisms and opportunities. MedComm (Beijing) 2024; 5:e560. [PMID: 38812572 PMCID: PMC11134193 DOI: 10.1002/mco2.560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 04/09/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024] Open
Abstract
White adipose tissue is not only a highly heterogeneous organ containing various cells, such as adipocytes, adipose stem and progenitor cells, and immune cells, but also an endocrine organ that is highly important for regulating metabolic and immune homeostasis. In individuals with obesity, dynamic cellular changes in adipose tissue result in phenotypic switching and adipose tissue dysfunction, including pathological expansion, WAT fibrosis, immune cell infiltration, endoplasmic reticulum stress, and ectopic lipid accumulation, ultimately leading to chronic low-grade inflammation and insulin resistance. Recently, many distinct subpopulations of adipose tissue have been identified, providing new insights into the potential mechanisms of adipose dysfunction in individuals with obesity. Therefore, targeting white adipose tissue as a therapeutic agent for treating obesity and obesity-related metabolic diseases is of great scientific interest. Here, we provide an overview of white adipose tissue remodeling in individuals with obesity including cellular changes and discuss the underlying regulatory mechanisms of white adipose tissue metabolic dysfunction. Currently, various studies have uncovered promising targets and strategies for obesity treatment. We also outline the potential therapeutic signaling pathways of targeting adipose tissue and summarize existing therapeutic strategies for antiobesity treatment including pharmacological approaches, lifestyle interventions, and novel therapies.
Collapse
Affiliation(s)
- Zi‐Han Yang
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang‐Zhou Chen
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Xiang Zhang
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Min‐Yi Ou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Poh‐Ching Tan
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue‐Wen Xu
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Qing‐Feng Li
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuang‐Bai Zhou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
47
|
Han M, Li J, Wu Y, Tang Z. Potential immune-related therapeutic mechanisms of multiple traditional Chinese medicines on type 2 diabetic nephropathy based on bioinformatics, network pharmacology and molecular docking. Int Immunopharmacol 2024; 133:112044. [PMID: 38648716 DOI: 10.1016/j.intimp.2024.112044] [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: 02/21/2024] [Revised: 03/27/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND The prevalence of type 2 diabetic nephropathy (T2DN) ranges from 20 % to 40 % among individuals with type 2 diabetes. Multiple immune pathways play a pivotal role in the pathogenesis of T2DN. This study aimed to investigate the immunomodulatory effects of active ingredients derived from 14 traditional Chinese medicines (TCMs) on T2DN. METHODS By removing batch effect on the GSE30528 and GSE96804 datasets, we employed a combination of weighted gene co-expression network analysis, least absolute shrinkage and selection operator analysis, protein-protein interaction network analysis, and the CIBERSORT algorithm to identify the active ingredients of TCMs as well as potential hub biomarkers associated with immune cells. Functional analysis was conducted using Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), and gene set variation analysis (GSVA). Additionally, molecular docking was employed to evaluate interactions between active ingredients and potential immunotherapy targets. RESULTS A total of 638 differentially expressed genes (DEGs) were identified in this study, comprising 5 hub genes along with 4 potential biomarkers. Notably, CXCR1, CXCR2, and FOS exhibit significant associations with immune cells while displaying robust or favorable affinities towards the active ingredients kaempferol, quercetin, and luteolin. Furthermore, functional analysis unveiled intricate involvement of DEGs, hub genes and potential biomarkers in pathways closely linked to immunity and diabetes. CONCLUSION The potential hub biomarkers and immunotherapy targets associated with immune cells of T2DN comprise CXCR1, CXCR2, and FOS. Furthermore, kaempferol, quercetin, and luteolin demonstrate potential immunomodulatory effects in modulating T2DN through the regulation of CXCR1, CXCR2, and FOS expression.
Collapse
MESH Headings
- Diabetic Nephropathies/drug therapy
- Diabetic Nephropathies/genetics
- Diabetic Nephropathies/immunology
- Humans
- Molecular Docking Simulation
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/genetics
- Drugs, Chinese Herbal/therapeutic use
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- Medicine, Chinese Traditional
- Computational Biology
- Network Pharmacology
- Protein Interaction Maps
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Receptors, Interleukin-8A/genetics
- Receptors, Interleukin-8A/metabolism
- Gene Regulatory Networks/drug effects
Collapse
Affiliation(s)
- Mingzheng Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jiale Li
- Department of Blood Transfusion, Yuexi Hospital of the Sixth Affiliated Hospital, Sun Yat-sen University (Xinyi People's Hospital), Xinyi, China
| | - Yijin Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
48
|
Fan M, Song E, Zhang Y, Zhang P, Huang B, Yan K, Yang W, Chakrabarti S, Mahajan H, Yan S, Xu Y, Hua S, Liu W, Wang C, Xu A, Ye D. Metabolic Dysfunction-Associated Steatohepatitis Detected by Neutrophilic Crown-Like Structures in Morbidly Obese Patients: A Multicenter and Clinicopathological Study. RESEARCH (WASHINGTON, D.C.) 2024; 7:0382. [PMID: 38812532 PMCID: PMC11134285 DOI: 10.34133/research.0382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is the progressive form of metabolic dysfunction-associated steatotic liver disease (MASLD), and closely associated with a high risk of liver-related morbidity and mortality. Although enhanced neutrophil infiltration of the liver is a histological hallmark of MASH, the morphological pattern of hepatic neutrophils and their relevance to the definition of MASH remain unknown. This clinicopathological study aimed to determine the association of neutrophilic crown-like structures (CLSs) in liver biopsies and evaluate their relevance to the histological diagnosis of MASH. A total of 483 morbidly obese adults who underwent bariatric surgery were recruited. Neutrophilic CLSs in liver biopsies were detected by immunohistochemistry for neutrophil elastase and proteinase 3. All participants were classified into 4 histological subgroups: no MASLD (118, 24.4%), MASLD (76, 15.7%), borderline MASH (185, 38.3%), and definite MASH (104, 21.5%). In the discovery cohort (n = 379), the frequency of neutrophilic CLSs increased in line with the severity of liver disease. The number of neutrophilic CLSs was positively correlated with established histological characteristics of MASH. At a cutoff value of <0.3 per 20× microscopic field, the number of neutrophilic CLSs yielded a robust diagnostic accuracy to discriminate no MASLD and MASLD from borderline MASH and definite MASH; a cutoff at >1.3 per 20× microscopic field exhibited a statistically significant accuracy to distinguish definite MASH from other groups (no MASLD, MASLD, and borderline MASH). The significance of neutrophilic CLSs in identifying borderline MASH and definite MASH was confirmed in an external validation cohort (n = 104). The frequency of neutrophilic CLSs was significantly higher than that of macrophagic CLSs. In conclusion, neutrophilic CLSs in the liver represent a typical histological characteristic of MASH and may serve as a promising indicator to improve the diagnostic accuracy of MASH during histological assessment of liver biopsies.
Collapse
Affiliation(s)
- Mengqi Fan
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Erfei Song
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuying Zhang
- Department of Obstetrics, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Pengfei Zhang
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bing Huang
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Kaixuan Yan
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wah Yang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Hema Mahajan
- Institute of Clinical Pathology and Medical Research, Pathology West, NSW Health Pathology, Sydney, NSW 2145, Australia
| | - Sen Yan
- Dr. Everett Chalmers Hospital, Fredericton, NB, Canada
| | - Ying Xu
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuang Hua
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wei Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cunchuan Wang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dewei Ye
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
49
|
O'Reilly ME, Ho S, Coronel J, Zhu L, Liu W, Xue C, Kim E, Cynn E, Matias CV, Soni RK, Wang C, Ionita-Laza I, Bauer RC, Ross L, Zhang Y, Corvera S, Fried SK, Reilly MP. linc-ADAIN, a human adipose lincRNA, regulates adipogenesis by modulating KLF5 and IL-8 mRNA stability. Cell Rep 2024; 43:114240. [PMID: 38753486 PMCID: PMC11334222 DOI: 10.1016/j.celrep.2024.114240] [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: 07/07/2023] [Revised: 03/01/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Adipose tissue remodeling and dysfunction, characterized by elevated inflammation and insulin resistance, play a central role in obesity-related development of type 2 diabetes (T2D) and cardiovascular diseases. Long intergenic non-coding RNAs (lincRNAs) are important regulators of cellular functions. Here, we describe the functions of linc-ADAIN (adipose anti-inflammatory), an adipose lincRNA that is downregulated in white adipose tissue of obese humans. We demonstrate that linc-ADAIN knockdown (KD) increases KLF5 and interleukin-8 (IL-8) mRNA stability and translation by interacting with IGF2BP2. Upregulation of KLF5 and IL-8, via linc-ADAIN KD, leads to an enhanced adipogenic program and adipose tissue inflammation, mirroring the obese state, in vitro and in vivo. KD of linc-ADAIN in human adipose stromal cell (ASC) hTERT adipocytes implanted into mice increases adipocyte size and macrophage infiltration compared to implanted control adipocytes, mimicking hallmark features of obesity-induced adipose tissue remodeling. linc-ADAIN is an anti-inflammatory lincRNA that limits adipose tissue expansion and lipid storage.
Collapse
Affiliation(s)
- Marcella E O'Reilly
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Sebastian Ho
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Johana Coronel
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Lucie Zhu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Wen Liu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Chenyi Xue
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Eunyoung Kim
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Esther Cynn
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Caio V Matias
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Chen Wang
- Department of Statistics, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Iuliana Ionita-Laza
- Department of Statistics, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | - Robert C Bauer
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Leila Ross
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yiying Zhang
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Muredach P Reilly
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA; Irving Institute for Clinical and Translational Research, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
50
|
Guillot A, Toussaint K, Ebersold L, ElBtaouri H, Thiebault E, Issad T, Peiretti F, Maurice P, Sartelet H, Bennasroune A, Martiny L, Dauchez M, Duca L, Durlach V, Romier B, Baud S, Blaise S. Sialic acids cleavage induced by elastin-derived peptides impairs the interaction between insulin and its receptor in adipocytes 3T3-L1. J Physiol Biochem 2024; 80:363-379. [PMID: 38393636 DOI: 10.1007/s13105-024-01010-5] [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: 06/23/2023] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
The insulin receptor (IR) plays an important role in insulin signal transduction, the defect of which is believed to be the root cause of type 2 diabetes. In 3T3-L1 adipocytes as in other cell types, the mature IR is a heterotetrameric cell surface glycoprotein composed of two α subunits and two β subunits. Our objective in our study, is to understand how the desialylation of N-glycan chains, induced by elastin-derived peptides, plays a major role in the function of the IR. Using the 3T3-L1 adipocyte line, we show that removal of the sialic acid from N-glycan chains (N893 and N908), induced by the elastin receptor complex (ERC) and elastin derived-peptides (EDPs), leads to a decrease in the autophosphorylation activity of the insulin receptor. We demonstrate by molecular dynamics approaches that the absence of sialic acids on one of these two sites is sufficient to generate local and general modifications of the structure of the IR. Biochemical approaches highlight a decrease in the interaction between insulin and its receptor when ERC sialidase activity is induced by EDPs. Therefore, desialylation by EDPs is synonymous with a decrease of IR sensitivity in adipocytes and could thus be a potential source of insulin resistance associated with diabetic conditions.
Collapse
Affiliation(s)
- Alexandre Guillot
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Kevin Toussaint
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Lucrece Ebersold
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Hassan ElBtaouri
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Emilie Thiebault
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Tarik Issad
- Université Paris Cité, Institut Cochin, CNRS, INSERM, 24 Rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Franck Peiretti
- INSERM, INRAE, C2VN, Aix Marseille University, 27 Bd Jean Moulin, 13385, Marseille, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Hervé Sartelet
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Amar Bennasroune
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Laurent Martiny
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Manuel Dauchez
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
- P3M, Multi-Scale Molecular Modeling Platform, Université de Reims Champagne Ardenne, 51100, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Vincent Durlach
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
- Cardiovascular and Thoracic Division, University Hospital of Reims, 51100, Reims, France
| | - Béatrice Romier
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
| | - Stéphanie Baud
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France
- P3M, Multi-Scale Molecular Modeling Platform, Université de Reims Champagne Ardenne, 51100, Reims, France
| | - Sébastien Blaise
- UMR CNRS 7369 MEDyC, University of Reims Champagne-Ardenne, UFR SEN, chemin des Rouliers, 51100, Reims, France.
| |
Collapse
|