1
|
Hsu JCN, Chiu KT, Chen CH, Wang CH, Shyue SK, Lee TS. HMGB1 Regulates Adipocyte Lipolysis via Caveolin-1 Signaling: Implications for Metabolic and Cardiovascular Diseases. Int J Mol Sci 2025; 26:4222. [PMID: 40362460 DOI: 10.3390/ijms26094222] [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: 04/01/2025] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
High-mobility group box 1 (HMGB1) is a nuclear protein that can be secreted or released into the extracellular environment during cellular stress, functioning as a damage-associated molecular pattern molecule. This study investigates the role of HMGB1 in adipocyte development and metabolism, explicitly examining its interaction with β3-adrenergic receptor-mediated lipolysis and caveolin-1 (CAV1) regulation, which may influence cardiovascular risk factors. Using 3T3-L1 preadipocytes and mouse embryonic fibroblasts, we demonstrated that HMGB1 expression increases progressively during adipogenesis, reaching peak levels in mature adipocytes. While exogenous HMGB1 treatment did not affect preadipocyte proliferation or differentiation, it inhibited lipolysis in mature adipocytes. Mechanistically, HMGB1 suppressed β3-adrenergic receptor agonist CL-316,243-induced hormone-sensitive lipase activation by reducing protein kinase A-mediated phosphorylation and attenuating extracellular signal-regulated kinase signaling without affecting upstream cyclic AMP levels. We discovered a novel regulatory mechanism wherein CAV1 physically interacts with HMGB1 in mature adipocytes, with c-Src-dependent CAV1 phosphorylation functioning as a negative regulator of HMGB1 secretion. This finding was confirmed in CAV1-deficient models, which displayed increased HMGB1 secretion and diminished lipolytic activity both in vitro and in vivo. Furthermore, administering HMGB1-neutralizing antibodies to wild-type mice enhanced fasting-induced lipolysis, establishing circulating HMGB1 as a crucial antilipolytic factor. These findings reveal HMGB1's previously uncharacterized role in adipose tissue metabolism as a negative regulator of lipolysis through CAV1-dependent mechanisms. This work provides new insights into adipose tissue metabolism regulation and identifies potential therapeutic targets for obesity-related metabolic disorders and cardiovascular diseases.
Collapse
Affiliation(s)
- Julia Chu-Ning Hsu
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, 145, Xingda Road, South District, Taichung 402202, Taiwan
| | - Kuan-Ting Chiu
- Department of Physiology, School of Medicine, National Yang-Ming University, 155, Sec. 2, Linong Street, Beitou District, Taipei 112304, Taiwan
| | - Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, 1, Sec. 1, Jenai Road, Zhongzheng District, Taipei 100233, Taiwan
| | - Chih-Hsien Wang
- Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, 7, Chungshan South Road, Zhongzheng District, Taipei 100225, Taiwan
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, 128, Sec. 2, Academia Road, Nankang District, Taipei 115201, Taiwan
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, 1, Sec. 1, Jenai Road, Zhongzheng District, Taipei 100233, Taiwan
| |
Collapse
|
2
|
Muszka Z, Jenei V, Mácsik R, Mezhonova E, Diyab S, Csősz R, Bácsi A, Mázló A, Koncz G. Life-threatening risk factors contribute to the development of diseases with the highest mortality through the induction of regulated necrotic cell death. Cell Death Dis 2025; 16:273. [PMID: 40216765 PMCID: PMC11992264 DOI: 10.1038/s41419-025-07563-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025]
Abstract
Chronic diseases affecting the cardiovascular system, diabetes mellitus, neurodegenerative diseases, and various other organ-specific conditions, involve different underlying pathological processes. However, they share common risk factors that contribute to the development and progression of these diseases, including air pollution, hypertension, obesity, high cholesterol levels, smoking and alcoholism. In this review, we aim to explore the connection between four types of diseases with different etiologies and various risk factors. We highlight that the presence of risk factors induces regulated necrotic cell death, leading to the release of damage-associated molecular patterns (DAMPs), ultimately resulting in sterile inflammation. Therefore, DAMP-mediated inflammation may be the link explaining how risk factors can lead to the development and maintenance of chronic diseases. To explore these processes, we summarize the main cell death pathways activated by the most common life-threatening risk factors, the types of released DAMPs and how these events are associated with the pathophysiology of diseases with the highest mortality. Various risk factors, such as smoking, air pollution, alcoholism, hypertension, obesity, and high cholesterol levels induce regulated necrosis. Subsequently, the release of DAMPs leads to chronic inflammation, which increases the risk of many diseases, including those with the highest mortality rates.
Collapse
Affiliation(s)
- Zsuzsa Muszka
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Viktória Jenei
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Rebeka Mácsik
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Evgeniya Mezhonova
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Silina Diyab
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Réka Csősz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| |
Collapse
|
3
|
Listyoko AS, Okazaki R, Harada T, Inui G, Yamasaki A. Impact of obesity on airway remodeling in asthma: pathophysiological insights and clinical implications. FRONTIERS IN ALLERGY 2024; 5:1365801. [PMID: 38562155 PMCID: PMC10982419 DOI: 10.3389/falgy.2024.1365801] [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: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of obesity among asthma patients has surged in recent years, posing a significant risk factor for uncontrolled asthma. Beyond its impact on asthma severity and patients' quality of life, obesity is associated with reduced lung function, increased asthma exacerbations, hospitalizations, heightened airway hyperresponsiveness, and elevated asthma-related mortality. Obesity may lead to metabolic dysfunction and immune dysregulation, fostering chronic inflammation characterized by increased pro-inflammatory mediators and adipocytokines, elevated reactive oxygen species, and reduced antioxidant activity. This chronic inflammation holds the potential to induce airway remodeling in individuals with asthma and obesity. Airway remodeling encompasses structural and pathological changes, involving alterations in the airway's epithelial and subepithelial layers, hyperplasia and hypertrophy of airway smooth muscle, and changes in airway vascularity. In individuals with asthma and obesity, airway remodeling may underlie heightened airway hyperresponsiveness and increased asthma severity, ultimately contributing to the development of persistent airflow limitation, declining lung function, and a potential increase in asthma-related mortality. Despite efforts to address the impact of obesity on asthma outcomes, the intricate mechanisms linking obesity to asthma pathophysiology, particularly concerning airway remodeling, remain incompletely understood. This comprehensive review discusses current research investigating the influence of obesity on airway remodeling, to enhance our understanding of obesity's role in the context of asthma airway remodeling.
Collapse
Affiliation(s)
- Aditya Sri Listyoko
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
- Pulmonology and Respiratory Medicine Department, Faculty of Medicine, Brawijaya University-Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tomoya Harada
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Genki Inui
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
4
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
5
|
Kamiya H, Komatsu S, Nishibeppu K, Ohashi T, Konishi H, Shiozaki A, Kubota T, Fujiwara H, Otsuji E. Obesity paradox as a new insight from postoperative complications in gastric cancer. Sci Rep 2023; 13:10116. [PMID: 37344511 DOI: 10.1038/s41598-023-36968-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023] Open
Abstract
The obesity paradox is reported to exist in various diseases. However, obesity is a pivotal issue in gastric cancer (GC) patients because of the surgical difficulty related to postoperative abdominal infectious complications (PAIC). This study clarified the existence of the obesity paradox in GC. Between 1997 and 2015, 1536 consecutive patients underwent curative gastrectomy. Of all patients, 18.6% (285/1536) were obese and tended to have a better prognosis (P = 0.073). In patients without PAIC, obesity was a significant prognostic factor for 5-year overall survival (P = 0.017). PAIC was an independent poor prognostic factor in both obese and non-obese patients (P < 0.001; hazard ratio [HR] 4.22 and 1.82). In pStage II-III patients, there was a large and significant prognostic difference between non-PAIC and PAIC obese patients (P = 0.006; 5-year overall survival: 69.7% vs. 43.8%) related to the higher incidence of peritoneal recurrence in PAIC obese patients (P = 0.035; 31% vs. 10%). Whereas, there was a small prognostic difference between non-PAIC and PAIC non-obese patients (P = 0.102; 5-year overall survival: 56.5% vs. 51.9%). Although the obesity paradox is present in GC, PAIC had a more negative prognostic impact through peritoneal recurrence in obese GC patients.
Collapse
Affiliation(s)
- Hajime Kamiya
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Keiji Nishibeppu
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery (Gastric Surgery Division), Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
6
|
Psoriatic arthritis: review of potential biomarkers predicting response to TNF inhibitors. Inflammopharmacology 2023; 31:77-87. [PMID: 36508130 PMCID: PMC9957889 DOI: 10.1007/s10787-022-01092-x] [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: 07/20/2022] [Accepted: 10/18/2022] [Indexed: 12/14/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic and painful inflammatory immune-mediated disease. It affects up to 40% of people with psoriasis and it is associated with several comorbidities such as obesity, diabetes, metabolic syndrome, and hypertension. PsA is difficult to diagnose because of its diverse symptoms, namely axial and peripheral arthritis, enthesitis, dactylitis, skin changes, and nail dystrophy. Different drugs exist to treat the inflammation and pain. When patients do not respond to conventional drugs, they are treated with biologic drugs. Tumour necrosis factor inhibitors (TNFi's) are commonly given as the first biologic drug; beside being expensive, they also lack efficacy in 50% of patients. A biomarker predicting individual patient's response to TNFi would help treating them earlier with an appropriate biologic drug. This study aimed to review the literature to identify potential biomarkers that should be investigated for their predictive ability. Several such biomarkers were identified, namely transmembrane TNFα (tmTNF), human serum albumin (HSA) and its half-life receptor, the neonatal Fc receptor (FcRn) which is also involved in IgG lifespan; calprotectin, high mobility group protein B1 (HMGB1) and advanced glycation end products (AGEs) whose overexpression lead to excessive production of pro-inflammatory cytokines; lymphotoxin α (LTα) which induces inflammation by binding to TNF receptor (TNFR); and T helper 17 (Th17) cells which induce inflammation by IL-17A secretion.
Collapse
|
7
|
Leisengang S, Gluding D, Hörster J, Peek V, Ott D, Rummel C, Schmidt MJ. Expression of adipokines and adipocytokines by epidural adipose tissue in cauda equina syndrome in dogs. J Vet Intern Med 2022; 36:1373-1381. [PMID: 35838307 PMCID: PMC9308421 DOI: 10.1111/jvim.16483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background Compression of epidural adipose tissue (EAT) within the scope of cauda equina syndrome (CES) could lead to an enhanced expression of inflammatory mediators, possibly contributing to pain amplification in dogs. Objectives To analyze expression of inflammatory adipo(‐cyto)kines within the EAT of dogs with CES. Animals Client‐owned dogs: 15 dogs with CES and 9 dogs euthanized for unrelated medical reasons (controls). Methods Prospective, experimental study. Epidural adipose tissue and subcutaneous adipose tissue were collected during dorsal laminectomy and used for real‐time quantitative polymerase chain reaction. Tissue explants were cultured for measurements of inflammation‐induced release of cytokines. Results Results show a CES‐associated upregulation of the cytokines tumor necrosis factor alpha (TNFα: mean ± SD: 18.88 ± 11.87, 95% CI: 10.90‐26.86 vs 9.66 ± 5.22, 95% CI: 5.29‐14.02, *: P = .04) and interleukin‐ (IL‐) 10 (20.1 ± 9.15, 95% CI: 14.82‐25.39 vs 11.52 ± 6.82, 95% CI: 5.82‐17.22, *: P = .03), whereas the expression of the adipokine leptin was attenuated in EAT of dogs with CES (3.07 ± 2.29, 95% CI: 1.80‐3.34 vs 9.83 ± 8.42, 95% CI: 3.36‐16.30, **: P = .007). Inflammatory stimulation of EAT explant cultures resulted in an enhanced release of IL‐6 (LPS: 5491.55 ± 4438, 95% CI: 833.7‐10 149; HMGB1: 1001.78 ± 522.2, 95% CI: 518.8‐1485; PBS: 310.9 ± 98.57, 95% CI: 228.5‐393.3, ***: P < .001). Conclusion and Clinical Importance Expression profile of inflammatory adipo(‐cyto)kines by EAT is influenced from compressive forces acting in dogs with CES and might contribute to amplification of pain.
Collapse
Affiliation(s)
- Stephan Leisengang
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany.,Center for Mind, Brain and Behavior - CMBB, Philipps University Marburg & Justus Liebig University Giessen, Giessen, Germany.,Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dennis Gluding
- Department of Veterinary Clinical Sciences, Clinic for Small Animals (Surgery), Justus Liebig University Giessen, Giessen, Germany
| | - Julia Hörster
- Department of Veterinary Clinical Sciences, Small Animal Clinic - Neurosurgery, Neuroradiology and Clinical Neurology, Justus Liebig University Giessen, Giessen, Germany
| | - Verena Peek
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Daniela Ott
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany.,Center for Mind, Brain and Behavior - CMBB, Philipps University Marburg & Justus Liebig University Giessen, Giessen, Germany
| | - Martin J Schmidt
- Department of Veterinary Clinical Sciences, Small Animal Clinic - Neurosurgery, Neuroradiology and Clinical Neurology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
8
|
NUCB2/Nesfatin-1 Reduces Obesogenic Diet Induced Inflammation in Mice Subcutaneous White Adipose Tissue. Nutrients 2022; 14:nu14071409. [PMID: 35406022 PMCID: PMC9003550 DOI: 10.3390/nu14071409] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Excess adipose tissue accumulation and obesity are characterised by chronic, low-grade, systemic inflammation. Nestfatin-1 is a neuropeptide derived from the precursor protein nucleobindin-2 (NUCB2), which was initially reported to exert anorexigenic effects. The present study aimed to investigate the effects of an obesogenic diet (OD; high-fat, high-sugar) in NUCB2 knockout (KO) mice and of nesfatin-1 treatment in LPS-stimulated 3T3-L1 preadipocytes. Methods: Subcutaneous white adipose tissue (Sc-WAT) samples from wild type (WT) and NUCB2 KO mice that were fed a normal diet (ND), or the OD for 12 weeks were used for RNA and protein extraction, as well as immunohistochemistry. 3T3-L1 cells were treated with 100 nM nesfatin-1 during differentiation and stimulated with 1 µg/mL LPS for measuring the expression and secretion of pro-inflammatory mediators by qPCR, western blotting, immunofluorescence, Bioplex, and ELISA. Results: Following the OD, the mRNA, protein and cellular expression of pro-inflammatory mediators (Tnfα, Il-6, Il-1β, Adgre1, Mcp1, TLR4, Hmbgb1 and NF-kB) significantly increased in the ScWAT of NUCB2 KO mice compared to ND controls. Adiponectin and Nrf2 expression significantly decreased in the ScWAT of OD-fed NUCB2 KO, without changes in the OD-fed WT mice. Furthermore, nesfatin-1 treatment in LPS-stimulated 3T3-L1 cells significantly reduced the expression and secretion of pro-inflammatory cytokines (Tnfα, Il-6, Il-1β, Mcp1) and hmgb1. Conclusion: An obesogenic diet can induce significant inflammation in the ScWAT of NUCB2 KO mice, involving the HMGB1, NRF2 and NF-kB pathways, while nesfatin-1 reduces the pro-inflammatory response in LPS-stimulated 3T3-L1 cells. These findings provide a novel insight into the metabolic regulation of inflammation in WAT.
Collapse
|
9
|
Liu S, Wang X, Kai Y, Tian C, Guo S, He L, Zhai D, Song X. Clinical significance of high mobility group box 1/toll-like receptor 4 in obese diabetic patients. Endocr J 2022; 69:235-242. [PMID: 34657898 DOI: 10.1507/endocrj.ej21-0381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
High mobility group box 1 (HMGB1) is an alarmin that may link to obesity and type 2 diabetes mellitus (T2DM). The present study analyzed the correlation between HMGB1/ Toll-like receptor 4 (TLR4) and certain biochemical parameters in obese (OB) diabetic patients. 40 normal glucose tolerant subjects (NGT) and 40 patients with newly diagnosed T2DM were enrolled. All patients were further divided into non-obese NGT (NGT-NOB), obese NGT (NGT-OB), non-obese T2DM (T2DM-NOB) and obese T2DM (T2DM-OB) groups according to body mass index (BMI).The levels of HMGB1 in serum were quantified using ELISA, whereas the mRNA expression levels of TLR4 in peripheral blood mononuclear cells were assessed using reverse transcription-quantitative PCR. The results suggested that the levels of HMGB1 and TLR4 were higher in NGT-OB and T2DM-NOB groups compared with those in NGT-NOB group. Similarly, the levels of these two markers were higher in T2DM-OB group compared with those in NGT-OB group. Correlation analysis indicated that the levels of HMGB1 and TLR4 were positively correlated with triglyceride (TG), fasting plasma glucose (FPG) levels and BMI, whereas a negative correlation between HMGB1 and high density lipoprotein (HDL) was noted. Linear regression analysis suggested that HMGB1 was associated with FPG and TG levels, whereas TLR4 was strongly associated with TG levels and BMI. The results demonstrated that the expression levels of HMGB1 and TLR4 in patients with T2DM or obesity were increased, which were associated with glycolipid metabolism disorders. Therefore, the HMGB1/TLR4 may serve a role in inflammatory process associated with obesity and T2DM.
Collapse
Affiliation(s)
- Shuai Liu
- Department of Blood Transfusion, the Third Affiliated Hospital of Xinxiang Medical University, Henan Xinxiang, 453003, China
| | - Xianchun Wang
- Clinical laboratory, the Third Affiliated Hospital of Xinxiang Medical University, Henan Xinxiang, 453003, China
| | - Yue Kai
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Henan Xinxiang, 453003, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Henan Xinxiang, 453000, China
| | - Chenrui Tian
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Henan Xinxiang, 453003, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Henan Xinxiang, 453000, China
| | - Sheng Guo
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Henan Xinxiang, 453003, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Henan Xinxiang, 453000, China
| | - Ling He
- Department of Ophthalmology, the 371 Affiliated Hospital of Xinxiang Medical University, Henan Xinxiang, 453003, China
| | - Desheng Zhai
- Department of Epidemiology and Biostatistics, School of Public Health, Xinxiang Medical University, Henan Xinxiang, 453003, China
| | - Xiangfeng Song
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Henan Xinxiang, 453003, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Henan Xinxiang, 453000, China
| |
Collapse
|
10
|
Wrba L, Halbgebauer R, Roos J, Huber-Lang M, Fischer-Posovszky P. Adipose tissue: a neglected organ in the response to severe trauma? Cell Mol Life Sci 2022; 79:207. [PMID: 35338424 PMCID: PMC8956559 DOI: 10.1007/s00018-022-04234-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/17/2022] [Accepted: 03/07/2022] [Indexed: 01/01/2023]
Abstract
Despite the manifold recent efforts to improve patient outcomes, trauma still is a clinical and socioeconomical issue of major relevance especially in younger people. The systemic immune reaction after severe injury is characterized by a strong pro- and anti-inflammatory response. Besides its functions as energy storage depot and organ-protective cushion, adipose tissue regulates vital processes via its secretion products. However, there is little awareness of the important role of adipose tissue in regulating the posttraumatic inflammatory response. In this review, we delineate the local and systemic role of adipose tissue in trauma and outline different aspects of adipose tissue as an immunologically active modifier of inflammation and as an immune target of injured remote organs after severe trauma.
Collapse
Affiliation(s)
- Lisa Wrba
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
- Department of Trauma, Orthopedic, Plastic and Hand Surgery, University Hospital of Augsburg, Augsburg, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Julian Roos
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Eythstr. 24, 89075, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Eythstr. 24, 89075, Ulm, Germany.
| |
Collapse
|
11
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. The Impact of Obesity, Adipose Tissue, and Tumor Microenvironment on Macrophage Polarization and Metastasis. BIOLOGY 2022; 11:339. [PMID: 35205204 PMCID: PMC8869089 DOI: 10.3390/biology11020339] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 02/15/2022] [Indexed: 12/11/2022]
Abstract
Tumor metastasis is a major cause of death in cancer patients. It involves not only the intrinsic alterations within tumor cells, but also crosstalk between these cells and components of the tumor microenvironment (TME). Tumorigenesis is a complex and dynamic process, involving the following three main stages: initiation, progression, and metastasis. The transition between these stages depends on the changes within the extracellular matrix (ECM), in which tumor and stromal cells reside. This matrix, under the effect of growth factors, cytokines, and adipokines, can be morphologically altered, degraded, or reorganized. Many cancers evolve to form an immunosuppressive TME locally and create a pre-metastatic niche in other tissue sites. TME and pre-metastatic niches include myofibroblasts, immuno-inflammatory cells (macrophages), adipocytes, blood, and lymphatic vascular networks. Several studies have highlighted the adipocyte-macrophage interaction as a key driver of cancer progression and dissemination. The following two main classes of macrophages are distinguished: M1 (pro-inflammatory/anti-tumor) and M2 (anti-inflammatory/pro-tumor). These cells exhibit distinct microenvironment-dependent phenotypes that can promote or inhibit metastasis. On the other hand, obesity in cancer patients has been linked to a poor prognosis. In this regard, tumor-associated adipocytes modulate TME through the secretion of inflammatory mediators, which modulate and recruit tumor-associated macrophages (TAM). Hereby, this review describes the cellular and molecular mechanisms that link inflammation, obesity, and cancer. It provides a comprehensive overview of adipocytes and macrophages in the ECM as they control cancer initiation, progression, and invasion. In addition, it addresses the mechanisms of tumor anchoring and recruitment for M1, M2, and TAM macrophages, specifically highlighting their origin, classification, polarization, and regulatory networks, as well as their roles in the regulation of angiogenesis, invasion, metastasis, and immunosuppression, specifically highlighting the role of adipocytes in this process.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, ECREIN, f-63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie moléculaire et anticancéreuse, Faculté des Sciences II, Université libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, ECREIN, f-63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, ECREIN, f-63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
12
|
Rosita R, Yueniwati Y, Endharti AT, Widodo MA. High-Glucose and Free Fatty Acid-Induced Adipocytes Generate Increasing of HMGB1 and Reduced GLUT4 Expression. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background
High-mobility group box 1 protein (HMGB1) is released from necrotic adipocytes into the extracellular milieu as an inflammatory alarmin in obesity. Although the impact of excess nutrient on adipocytes is well known, it is not clear how specific its component drive cell-size and damaged of adipocytes, and how this relates to the risk of insulin resistance.
Objectives
The aim of this study was to determine HMGB1 level in adipocytes cultures after high glucose and/or FFA exposures and to assess GLUT4 expression. We determined cellular features of adipocytes that correlates to HMGB1 released and insulin resistance.
Methods
Differentiated adipocytes were exposed to high glucose and/or FFAs for 7 days. ELISA was performed on supernatant to assess the HMGB1 level. Total GLUT4 expression were quantified by immunofluorescense.
Results
High glucose and FFA-exposed cells have significant increase of HMGB1 level with decreased of cell size and necrotic adipocytes features. The total GLUT4 were reduced in HG-cells (p <0,045), but not in FFA cells. Hypertrophic adipocytes (p <0.05) and slight decrease of GLUT4 expression were showed on HG+FFA exposures with no increase of HMGB1 level. There was a significant correlation between cell size and HMGB1 level (R -0,637, p < 0.026)
Conclusion
The expression level studies between high glucose, FFA, and a combination of both on adipocytes results strongly suggest that high glucose is more damaging to adipocyte compared to FFA. Nevertheless, the combination of the two causes adipocyte dysfunction with general features of adipose tissue in obesity, suggested it can be used as a hypertrophic adipocytes model to study obesity in vitro.
Collapse
|
13
|
Abstract
Obesity is a chronic and progressive process affecting whole-body energy balance and is associated with comorbidities development. In addition to increased fat mass, obesity induces white adipose tissue (WAT) inflammation and fibrosis, leading to local and systemic metabolic dysfunctions, such as insulin resistance (IR). Accordingly, limiting inflammation or fibrosis deposition may improve IR and glucose homeostasis. Although no targeted therapy yet exists to slow or reverse adipose tissue fibrosis, a number of findings have clarified the underlying cellular and molecular mechanisms. In this review, we highlight adipose tissue remodeling events shown to be associated with fibrosis deposition, with a focus on adipose progenitors involved in obesity-induced healthy as well as unhealthy WAT expansion. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Geneviève Marcelin
- INSERM, Nutrition and Obesities: Systemic Approach (NutriOmics) Research Unit, UMRS U1269, Sorbonne Université, Paris, France; ,
| | | | - Karine Clément
- INSERM, Nutrition and Obesities: Systemic Approach (NutriOmics) Research Unit, UMRS U1269, Sorbonne Université, Paris, France; , .,Nutrition Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
14
|
Guzmán-Ruiz R, Tercero-Alcázar C, López-Alcalá J, Sánchez-Ceinos J, Malagón MM, Gordon A. The potential role of the adipokine HMGB1 in obesity and insulin resistance. Novel effects on adipose tissue biology. Mol Cell Endocrinol 2021; 536:111417. [PMID: 34339826 DOI: 10.1016/j.mce.2021.111417] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022]
Abstract
Discovery of the adipose tissue as a major source of signaling molecules almost three decades ago set a novel physiological paradigm that paved the way for the identification of metabolic organs as endocrine organs. Adipocytes, the main adipose tissue cell type, do not only represent the principal site of energy storage in form of triglycerides, but also produce a variety of molecules for short and long distance intercellular communication, named adipokines, which coordinate systemic responses. Although the best known adipokines identified and characterized hitherto are leptin and adiponectin, novel adipokines are continuously being described, what have significantly helped to elucidate the role of adipocyte biology in obesity and associated comorbidities. One of these novel adipokines is high-mobility group box 1 (HMGB1), a ubiquitous nuclear protein that has been recently reported to be dysregulated in obese dysfunctional adipocytes. Although the classical function of HMGB1 is related to inflammation and immunity, acting as an alarmin, novel advances evidence an active implication of HMGB1 in tissue remodeling and fibrosis. This review summarizes the current evidence on the mechanisms controlling HMGB1 release, as well as its role as a regulator of adipocyte function and extracellular matrix remodeling, with special emphasis on the potential of this novel adipokine as a target in the obesity treatment.
Collapse
Affiliation(s)
- R Guzmán-Ruiz
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, 14014, Córdoba, Spain; CIBER Fisiopatología de La Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain.
| | - C Tercero-Alcázar
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, 14014, Córdoba, Spain; CIBER Fisiopatología de La Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain
| | - J López-Alcalá
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, 14014, Córdoba, Spain; CIBER Fisiopatología de La Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain
| | - J Sánchez-Ceinos
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, 14014, Córdoba, Spain; CIBER Fisiopatología de La Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain
| | - M M Malagón
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, 14014, Córdoba, Spain; CIBER Fisiopatología de La Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain
| | - A Gordon
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, 14014, Córdoba, Spain; CIBER Fisiopatología de La Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain
| |
Collapse
|
15
|
Saxena S, Kruys V, De Jongh R, Vamecq J, Maze M. High-Mobility Group Box-1 and Its Potential Role in Perioperative Neurocognitive Disorders. Cells 2021; 10:2582. [PMID: 34685561 PMCID: PMC8533835 DOI: 10.3390/cells10102582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Aseptic surgical trauma provokes the release of HMGB1, which engages the innate immune response after binding to pattern-recognition receptors on circulating bone marrow-derived monocytes (BM-DM). The initial systemic inflammation, together with HMGB1, disrupts the blood-brain barrier allowing penetration of CCR2-expressing BM-DMs into the hippocampus, attracted by the chemokine MCP-1 that is upregulated by HMGB1. Within the brain parenchyma quiescent microglia are activated and, together with the translocated BM-DMs, release proinflammatory cytokines that disrupt synaptic plasticity and hence memory formation and retention, resulting in postoperative cognitive decline (PCD). Neutralizing antibodies to HMGB1 prevents the inflammatory response to trauma and PCD.
Collapse
Affiliation(s)
- Sarah Saxena
- Department of Anesthesia, University Hospital Center (CHU de Charleroi), 6000 Charleroi, Belgium;
| | - Véronique Kruys
- ULB Immunology Research Center (UIRC), Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, Free University of Brussels (ULB), 6041 Gosselies, Belgium;
| | - Raf De Jongh
- Department of Anesthesia, Fondation Hopale, 62600 Berck-sur-Mer, France;
| | - Joseph Vamecq
- Inserm, CHU Lille, Université de Lille, CHRU Lille, Center of Biology and Pathology (CBP) Pierre-Marie Degand, EA 7364 RADEME, 59000 Lille, France;
- Laboratory of Hormonology, Metabolism-Nutrition & Oncology (HMNO), Department of Biochemistry and Molecular Biology, University of North France, 59000 Lille, France
| | - Mervyn Maze
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA 94143, USA
| |
Collapse
|
16
|
Abstract
The advanced glycosylation end product receptor (RAGE) acts as a recognition receptor and interacts with different types of ligands that form and accumulate in the tissues and circulation, such as diabetes, inflammation, insulin resistance, and obesity. In these environments, RAGE is expressed on the surface of various cells associated with tissue disturbance. This review mainly summarizes the characteristics of RAGE-related signalling, with a particular emphasis on the role of RAGE in the development of obesity. We also briefly describe the phenotypes and characteristics of macrophages and focus on the role of adipose tissue macrophages (ATMs) and the regulatory mechanisms in obesity, diabetes, and other related metabolic diseases. Besides, we will also elaborate on the prospect of new strategies for treating diabetes and obesity-related metabolic diseases by inhibiting RAGE signalling and regulating ATMs recruitment and polarization.
Collapse
Affiliation(s)
- Ziqian Feng
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Luochen Zhu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
17
|
Chen L, Zhu H, Su S, Harshfield G, Sullivan J, Webb C, Blumenthal JA, Wang X, Huang Y, Treiber FA, Kapuku G, Li W, Dong Y. High-Mobility Group Box-1 Is Associated With Obesity, Inflammation, and Subclinical Cardiovascular Risk Among Young Adults: A Longitudinal Cohort Study. Arterioscler Thromb Vasc Biol 2020; 40:2776-2784. [PMID: 32814439 PMCID: PMC7578115 DOI: 10.1161/atvbaha.120.314599] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE We aimed to characterize circulating HMGB1 (high-mobility group box-1) levels, one of the better-characterized damage-associated molecular patterns, with respect to age, sex, and race in the general population, and investigate the longitudinal associations of HMGB1 with inflammatory markers, obesity, and preclinical markers of cardiovascular disease. Approach and Results: The analyses included 489 participants (50% Blacks, aged 24.6±3.3 years at the first visit) with up to 4 follow-up visits (1149 samples) over a maximum of 8.5 years. Systolic blood pressure, diastolic blood pressure, carotid-femoral pulse wave velocity, and carotid intima-media thickness together with plasma HMGB1, hs-CRP (high-sensitivity C-reactive protein), IFN-γ (interferon-γ), IL-6 (interleukin-6), IL-10 (interleukin-10), and TNF-α (tumor necrosis factor-α) were measured at each visit. At baseline, plasma HMGB1 concentrations were higher in Blacks compared with Whites (3.86 versus 3.20 ng/mL, P<0.001), and in females compared with males (3.75 versus 3.30 ng/mL, P=0.005). HMGB1 concentrations increased with age (P=0.007), and higher levels of obesity measures (P<0.001). Without adjustment for age, sex, race, and body mass index, HMGB1 concentrations were positively associated with hs-CRP, IL-6, TNF-α, systolic blood pressure, diastolic blood pressure, and carotid-femoral pulse wave velocity (P<0.05) but not IL-10, IFN-γ or carotid intima-media thickness. After covariate adjustments, the associations of HMGB1 with hs-CRP, and carotid-femoral pulse wave velocity remained statistically significant (P<0.05). CONCLUSIONS This study demonstrates the age, sex, and race differences in circulating HMGB1. The increasing circulating concentrations of HMGB1 with age suggest a potential role of HMGB1 in the pathogenesis of chronic low-grade inflammation, obesity, and subclinical cardiovascular disease risk.
Collapse
Affiliation(s)
- Li Chen
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Haidong Zhu
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Shaoyong Su
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Gregory Harshfield
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jennifer Sullivan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Clinton Webb
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - James A. Blumenthal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Xiaoling Wang
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ying Huang
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Frank A. Treiber
- College of Nursing, Medical University of South Carolina, Charleston, SC, USA
- College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Gaston Kapuku
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Wenjun Li
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yanbin Dong
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
18
|
Peek V, Neumann E, Inoue T, Koenig S, Pflieger FJ, Gerstberger R, Roth J, Matsumura K, Rummel C. Age-Dependent Changes of Adipokine and Cytokine Secretion From Rat Adipose Tissue by Endogenous and Exogenous Toll-Like Receptor Agonists. Front Immunol 2020; 11:1800. [PMID: 32973755 PMCID: PMC7466552 DOI: 10.3389/fimmu.2020.01800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/06/2020] [Indexed: 01/04/2023] Open
Abstract
White adipose tissue but recently also brown adipose tissue have emerged as endocrine organs. Age-associated obesity is accompanied by prolonged and elevated lipopolysaccharide (LPS)-induced sickness symptoms and increased cytokine and adipokine levels in the circulation partially originating from adipose tissue. In the present study, ex vivo fat explants were used to investigate how the exogenous pathogen-associated molecular pattern (PAMP) LPS or the endogenous danger-associated molecular patterns (DAMPs) high mobility group box-1 protein (HMGB1) and biglycan modulate the release of cytokines and adipokines/batokines and, thus, could influence systemic and/or local inflammation. The response of adipose tissue (epididymal, retroperitoneal, subcutaneous, and brown) was compared between young lean and old obese rats (2 vs. 24 months old). LPS induced a strong interleukin (IL)-6 and tumor necrosis factor (TNF) alpha release into the supernatant of all adipose tissue types investigated. HMGB1 (subcutaneous) and biglycan (retroperitoneal) led to an increased release of IL-6 and TNFalpha (HMGB1) and decreased visfatin and adiponectin (biglycan) secretion from epididymal adipose tissue (young rats). Visfatin was also decreased by HMGB1 in retroperitoneal adipose tissue of old rats. We found significantly higher leptin (all fat pads) and adiponectin (subcutaneous) levels in supernatants of adipose tissue from old compared to young rats, whereas visfatin secretion showed the opposite. The expression of the biglycan receptor Toll-like receptor (TLR) 2 as well as the LPS and HMGB1 receptors TLR4 and receptor for advanced glycation end products (RAGE) were reduced with age (TLR4/RAGE) and by stimulation with their ligands (subcutaneous). Overall, we revealed that adipokines/adipose-tissue released cytokines show some modulation of their release caused by mediators of septic (batokines) and sterile inflammation with potential implication for acute and chronic disease. Moreover, aging may increase or decrease the release of fat-derived mediators. These data show that DAMPS and LPS locally modulate cytokine secretion while only DAMPS but not LPS can locally alter adipokine secretion during inflammation.
Collapse
Affiliation(s)
- Verena Peek
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Elena Neumann
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus Liebig University Gießen, Bad Nauheim, Germany
| | - Tomohiro Inoue
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
| | - Sandy Koenig
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Fabian Johannes Pflieger
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Rüdiger Gerstberger
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Joachim Roth
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany.,Joachim Roth and Christoph Rummel, Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Kiyoshi Matsumura
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany.,Joachim Roth and Christoph Rummel, Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| |
Collapse
|
19
|
Kim HS, Han M, Park IH, Park CH, Kwak MS, Shin JS. Sulfatide Inhibits HMGB1 Secretion by Hindering Toll-Like Receptor 4 Localization Within Lipid Rafts. Front Immunol 2020; 11:1305. [PMID: 32655573 PMCID: PMC7324676 DOI: 10.3389/fimmu.2020.01305] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
The high mobility group box 1 (HMGB1) is a well-known late mediator of sepsis, secreted by multiple stimuli, involving pathways, such as the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways, and reactive oxygen species (ROS) under inflammation. Sulfatide, in contrast, is a sphingolipid commonly found in myelin sheets with a disputed immunological role. We sought to determine the immunological characteristics of sulfatide in the periphery by analyzing the secretion of HMGB1 triggered by lipopolysaccharide (LPS) stimulation in Raw 264.7 cells. Suppression of HMGB1 secretion by inhibiting its cytosolic translocation was observed after pre-treatment with sulfatide before LPS stimulation. Further analysis of the downstream molecules of toll-like receptor (TLR) signaling revealed suppression of c-Jun N-terminal kinase (JNK) phosphorylation and p65 translocation. LPS-mediated ROS production was also decreased when sulfatide pre-treatment was provided, caused by the down-regulation of the phosphorylation of activators, such as IRAK4 and TBK1. Investigation of the upstream mechanism that encompasses all the aforementioned inhibitory characteristics unveiled the involvement of lipid rafts. In addition to the co-localization of biotinylated sulfatide and monosialotetrahexosylganglioside, a decrease in LPS-induced co-localization of TLR4 and lipid raft markers was observed when sulfatide treatment was given before LPS stimulation. Overall, sulfatide was found to exert its anti-inflammatory properties by hindering the co-localization of TLR4 and lipid rafts, nullifying the effect of LPS on TLR4 signaling. Similar effects of sulfatide were also confirmed in the LPS-mediated murine experimental sepsis model, showing decreased levels of serum HMGB1, increased survivability, and reduced pathological severity.
Collapse
Affiliation(s)
- Hee Sue Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Myeonggil Han
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - In Ho Park
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Cheol Ho Park
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Man Sup Kwak
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
20
|
Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clément K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest 2020; 129:4032-4040. [PMID: 31498150 DOI: 10.1172/jci129192] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Obesity originates from an imbalance between caloric intake and energy expenditure that promotes adipose tissue expansion, which is necessary to buffer nutrient excess. Patients with higher visceral fat mass are at a higher risk of developing severe complications such as type 2 diabetes and cardiovascular and liver diseases. However, increased fat mass does not fully explain obesity's propensity to promote metabolic diseases. With chronic obesity, adipose tissue undergoes major remodeling, which can ultimately result in unresolved chronic inflammation leading to fibrosis accumulation. These features drive local tissue damage and initiate and/or maintain multiorgan dysfunction. Here, we review the current understanding of adipose tissue remodeling with a focus on obesity-induced adipose tissue fibrosis and its relevance to clinical manifestations.
Collapse
Affiliation(s)
- Geneviève Marcelin
- Nutrition and Obesities: Systemic Approaches (NutriOmics, UMRS U1269), INSERM, Sorbonne Université, Paris, France
| | - Ana Letícia M Silveira
- Nutrition and Obesities: Systemic Approaches (NutriOmics, UMRS U1269), INSERM, Sorbonne Université, Paris, France.,Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laís Bhering Martins
- Nutrition and Obesities: Systemic Approaches (NutriOmics, UMRS U1269), INSERM, Sorbonne Université, Paris, France.,Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adaliene Vm Ferreira
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches (NutriOmics, UMRS U1269), INSERM, Sorbonne Université, Paris, France.,Nutrition Department, Hôpital Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris, Paris, France
| |
Collapse
|
21
|
Pattern Recognition Receptor-Mediated Chronic Inflammation in the Development and Progression of Obesity-Related Metabolic Diseases. Mediators Inflamm 2019; 2019:5271295. [PMID: 31582899 PMCID: PMC6754942 DOI: 10.1155/2019/5271295] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity-induced chronic inflammation is known to promote the development of many metabolic diseases, especially insulin resistance, type 2 diabetes mellitus, nonalcoholic fatty liver disease, and atherosclerosis. Pattern recognition receptor-mediated inflammation is an important determinant for the initiation and progression of these metabolic diseases. Here, we review the major features of the current understanding with respect to obesity-related chronic inflammation in metabolic tissues, focus on Toll-like receptors and nucleotide-binding oligomerization domain-like receptors with an emphasis on how these receptors determine metabolic disease progression, and provide a summary on the development and progress of PRR antagonists for therapeutic intervention.
Collapse
|
22
|
Cao D, Wang W, Li S, Lai W, Huang X, Zhou J, Chen X, Li X. TLR2-Deficiency Promotes Prenatal LPS Exposure-Induced Offspring Hyperlipidemia. Front Physiol 2019; 10:1102. [PMID: 31507457 PMCID: PMC6713936 DOI: 10.3389/fphys.2019.01102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022] Open
Abstract
Toll-like receptor 2 (TLR2), which recognizes several lipopeptides and transduces inflammatory signaling, promotes the pathogenesis of diet-induced dyslipidemia and obesity. TLR2-deficient mice were shown to have improved insulin sensitivity and reduced diet-induced metabolic syndrome. Previous studies demonstrated that prenatal lipopolysaccharide (LPS) exposure causes dyslipidemia accompanied by increased body weight and insulin resistance in offspring. To determine whether TLRs are involved in this complex abnormal phenotype, we analyzed TLR2 and TLR4 expression levels in adipose tissues from offspring with prenatal LPS-exposure (offspring-pLPS) and compared these levels to those of control offspring with prenatal saline-exposure (offspring-pSaline). TLR2 expression was specifically upregulated in the adipose tissue of offspring-pLPS mice. However, unexpectedly, TLR2-deficient offspring-pLPS mice not only presented with an abnormal phenotype comparable to that of wild-type offspring-pLPS mice but also exhibited significantly more severe hyperlipidemia. Our further analyses revealed a dramatic upregulation of TLR4 expression and overactivation of the TLR4/Myd88 signaling pathway in TLR2-deficient offspring-pLPS adipose tissue. Our finding suggests a compensatory genetic interaction between TLR2 and TLR4 in the context of prenatal inflammatory stimulation, and this interaction likely contributes to the prenatal inflammation-induced hyperlipidemia and lipid overload-induced obesity, thus providing a potential mechanism for the fetal origin of adult metabolic diseases.
Collapse
Affiliation(s)
- Dayan Cao
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Wenjia Wang
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Shuhui Li
- Department of Clinical Biochemistry, College of Pharmacy, Army Medical University, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoyong Huang
- Institute of Immunology, PLA, Army Medical University, Chongqing, China
| | - Jianzhi Zhou
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Xin Chen
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| |
Collapse
|
23
|
Mazur-Bialy AI. Superiority of the Non-Glycosylated Form Over the Glycosylated Form of Irisin in the Attenuation of Adipocytic Meta-Inflammation: A Potential Factor in the Fight Against Insulin Resistance. Biomolecules 2019; 9:biom9090394. [PMID: 31438646 PMCID: PMC6770638 DOI: 10.3390/biom9090394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/30/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022] Open
Abstract
Irisin is an adipomyokine that promotes the browning of white adipose tissue and exhibits protective potential against the development of insulin resistance and type 2 diabetes. In our bodies, it occurs in its glycosylated form (G-IR): its activity is still poorly understood, because the majority of studies have used its non-glycosylated counterpart (nG-IR). Glycosylation can affect protein function: therefore, the present study attempted to compare the actions of both forms of irisin toward inflammatory activation of the main component of adipose tissue. The study was carried out in a coculture of 3T3 adipocytes and RAW 264.7 macrophages maintained in the presence of nG-IR or G-IR. The impact on vitality and the expression and release of key inflammatory mediators important for insulin resistance and diabetes development were assessed. The studies showed that both forms effectively inhibited the expression and release of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, macrophage chemotactic protein (MCP)-1, high-mobility group box (HMGB1), leptin, and adiponectin. However, in the case of TNF-α, IL-1β, MCP-1, and HMGB1, the inhibition exerted by nG-IR was more prominent than that by G-IR. In addition, only nG-IR significantly inhibited macrophage migration. Here, nG-IR seemed to be the stronger inhibitor of the development of obesity-related inflammation; however, G-IR also had anti-inflammatory potential.
Collapse
Affiliation(s)
- Agnieszka Irena Mazur-Bialy
- Department of Ergonomics and Exercise Physiology, Institute of Physiotherapy, Faculty of Health Science, Jagiellonian University Medical College, Grzegorzecka 20, 31-531 Krakow, Poland.
| |
Collapse
|
24
|
Hill AV, Menon R, Perez-Patron M, Carrillo G, Xu X, Taylor BD. High-mobility group box 1 at the time of parturition in women with gestational diabetes mellitus. Am J Reprod Immunol 2019; 82:e13175. [PMID: 31353785 DOI: 10.1111/aji.13175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/28/2019] [Accepted: 07/19/2019] [Indexed: 12/14/2022] Open
Abstract
PROBLEM High-mobility group box 1 (HMGB1), a danger-associated molecular pattern marker, may indicate sterile inflammation through innate immune pathways. HMGB1 is implicated in hyperglycemia and excess glucose in trophoblast. Metabolic dysfunction and dyslipidemia are associated with gestational diabetes mellitus (GDM), but few studies examined associations between HMGB1 and GDM. We determined HMGB1 levels, and the ratio of HMGB1 to innate immune markers, in women with GDM at parturition. METHOD OF STUDY This case-control study of 50 GDM pregnancies and 100 healthy controls utilized data and plasma samples from PeriBank. HMGB1, pentraxin-3, and interleukin (IL)-6 were measured by ELISA. Logistic regression calculated odds ratios (OR) and 95% confidence intervals (CI) adjusting for age, pre-pregnancy body mass index, and type of labor. RESULTS There were no significant associations between HMGB1 and GDM. The ratio of HMGB1 to pentraxin-3 and IL-6 did not alter the odds of GDM. There was a significant statistical interaction between HMGB1 and maternal age (P = .02). When associations were examined by age groups, HMGB1 was associated with reduced odds of HMGB1 among women ≤25 (AOR = 0.007 CI 95% <0.001-0.3). Odds ratios increased as age increased (AOR range 1.2-3.8) but results were not statistically significant. CONCLUSION High-mobility group box 1 was not associated with GDM. However, we found evidence that maternal age was a potential effect modifier of the relationship between HMGB1 and GDM. As there is growing evidence that HMGB1 may play important roles in reproduction, future studies should explore maternal factors that may alter HMGB1 levels.
Collapse
Affiliation(s)
- Ashley V Hill
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX, USA.,Division of Adolescent and Young Adult Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Maria Perez-Patron
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX, USA
| | - Genny Carrillo
- Department of Environmental and Occupational Health, Texas A&M University, College Station, TX, USA
| | - Xiaohui Xu
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX, USA
| | - Brandie D Taylor
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX, USA.,Department of Epidemiology and Biostatistics, College of Public Health, Temple University, Philadelphia, PA, USA
| |
Collapse
|
25
|
Han SY, Choi SH, Shin JS, Lee EJ, Han SH, Yoon JS. High-Mobility Group Box 1 Is Associated with the Inflammatory Pathogenesis of Graves' Orbitopathy. Thyroid 2019; 29:868-878. [PMID: 30973094 DOI: 10.1089/thy.2018.0285] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background: High-mobility group box 1 (HMGB1) has been implicated in the pathogenesis of inflammatory autoimmune diseases. This study investigated the influence and mechanisms of HMGB1 in Graves' orbitopathy (GO). Methods: HMGB1 and its receptors (receptor for advanced glycation end products [RAGE], Toll-like receptor [TLR] 2, and TLR4) mRNA levels were evaluated by real-time polymerase chain reaction (RT-PCR) in GO and non-GO orbital tissues. The mRNA expressions of HMGB1 and its receptors were evaluated in primary cultured orbital fibroblasts from six GO patients and five healthy control subjects under interleukin (IL)-1β or tumor necrosis factor (TNF)-α stimulation using RT-PCR. HMGB1 secretions under IL-1β or TNF-α stimulation were evaluated by enzyme-linked immunosorbent assay (ELISA). The effects of an anti-HMGB1 antibody, RAGE antagonist (FPS-ZM1), and anti-TLR2 antibody on the expressions of IL-1β or TNF-α induced pro-inflammatory cytokines and phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells were evaluated using ELISA and Western blot analysis, respectively. The plasma levels of HMGB1 were compared among patients with active GO (n = 51), inactive GO (n = 48), Graves' disease without GO (n = 30), and healthy control subjects (n = 46) by ELISA. Results: The genes encoding HMGB1 and its receptors, as well as HMGB1 protein expression, were increased in GO orbital tissues compared to non-GO tissues. IL-1β and TNF-α stimulation increased the mRNA levels of HMGB1, RAGE, and TLR2 and the secretion of HMGB1 protein further in GO cells. Anti-HMGB1 antibody, FPS-ZM1, and anti-TLR2 antibody reduced IL-1β- or TNF-α-induced production of pro-inflammatory cytokines and phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells. The plasma levels of HMGB1 were highly increased in patients with active GO, and were significantly correlated with the clinical activity score (r = 0.566, p = 0.002) and levels of thyrotropin binding inhibitory immunoglobulin (r = 0.506, p < 0.001). Conclusions: This study demonstrates an association of HMGB1 and its receptors in the inflammatory mechanisms of GO. HMGB1, RAGE, and TLR2 blockers reduced the production of pro-inflammatory molecules, providing a rationale for blocking the HMGB1 pathway to treat patients with GO. HMGB1 proteins were secreted further in the plasma of patients with active GO, suggesting that HMGB1 can be used as a biomarker of GO activity.
Collapse
Affiliation(s)
- So Young Han
- 1 Department of Ophthalmology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- 2 Institute of Vision Research, Department of Ophthalmology, Severance Hospital; Severance Hospital, Institute of Endocrine Research; Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soo Hyun Choi
- 2 Institute of Vision Research, Department of Ophthalmology, Severance Hospital; Severance Hospital, Institute of Endocrine Research; Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeon-Soo Shin
- 3 Department of Microbiology, BK21 PLUS Project for Medical Science, Institute for Immunology and Immunological Diseases and Severance Biomedical Science Institute; Severance Hospital, Institute of Endocrine Research; Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jig Lee
- 4 Department of Endocrinology, Severance Hospital, Institute of Endocrine Research; Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sueng-Han Han
- 2 Institute of Vision Research, Department of Ophthalmology, Severance Hospital; Severance Hospital, Institute of Endocrine Research; Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Sook Yoon
- 2 Institute of Vision Research, Department of Ophthalmology, Severance Hospital; Severance Hospital, Institute of Endocrine Research; Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
26
|
Erdal M, Altunkaynak BZ, Kocaman A, Alkan I, Öztas E. The role of HMGB1 in liver inflammation in obese rats. Biotech Histochem 2019; 94:449-458. [PMID: 30916587 DOI: 10.1080/10520295.2019.1589573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Obesity is a chronic disease that is characterized by increased body fat owing to imbalance between consumed and expended energy. Inflammation generally is accompanied by accumulation of excess lipid in adipose tissue and liver. High mobility group box-1 (HMGB1) participates in the pathogenesis of inflammatory diseases. We investigated the relation of the number of HMGB1 positive cells to body mass index (BMI), liver inflammation and the number of Kupffer cells. We divided 18 female Wistar albino rats into two groups: group 1, untreated control fed normal commercial rat diet and group 2, obese rats fed a special diet containing 40% fat. The plasma concentrations of cholesterol, glucose, superoxide dismutase enzyme (SOD) and catalase activities were measured for all animals. The numbers of hepatocytes, Kupffer cells and HMGB1 positive cells were counted using stereological methods. The mean numbers of Kupffer cells and HMGB1 positive cells were higher for group 2 than for group 1. The concentrations of plasma cholesterol and glucose levels also were higher in group 2. Plasma levels of SOD and catalase were significantly lower in group 2 compared to group 1. The number of HMGB1 cells was related directly to BMI and inflammation. The role of HMGB1 was demonstrated for the liver of the obese group. We demonstrated the relations among HMGB1, BMI, obesity and inflammation.
Collapse
Affiliation(s)
- M Erdal
- Department of Histology and Embryology, Gulhane Medical School, Ankara, Turkey
| | - B Z Altunkaynak
- Department of Histology and Embryology, Medical School, Istanbul Okan University , Istanbul , Turkey
| | - A Kocaman
- Department of Histology and Embryology, Medical School, Ondokuz Mayis University , Samsun , Turkey
| | - I Alkan
- Department of Histology and Embryology, Medical School, Ondokuz Mayis University , Samsun , Turkey
| | - E Öztas
- Department of Histology and Embryology, Gulhane Medical School, Ankara, Turkey
| |
Collapse
|
27
|
Abstract
We evaluated the effect of in vitro digested milk on mature adipocytes 3T3-L1, paying particular attention to its fatty acid composition, and comparing human (HM), donkey (DM), bovine (BM), ovine (OM), caprine (CM) and formula (FM) milk. Cellular viability, apoptosis, oxidative response and gene expression levels of NF-κB p65, HMGB1, SREBP-1c and FAS were evaluated. Digested milk treatments significantly reduced 3T3-L1 mature adipocytes viability and caspase activity compared with control group, but no significant differences were observed among different sources of digested milk. In all digested milk samples, ROS level was higher than the control, however, the digested human and formula milk showed lower levels of ROS than DM, BM, OM and CM samples. Lower capacity of HM and FM to induce oxidative stress in mature adipocytes was ascribed to the peculiar free fatty acids profile of digested milk samples. All milk treatments elicited a significant over-expression of NF-κB p65 in 3T3-L1 adipocytes compared to the control; the lowest gene expression was found in HM, BM, OM and CM, the highest in FM and an intermediate behavior was shown in DM. All digested milk treatments influenced the gene expression of SRBP-1c with FM and HM showing the highest levels. For FAS expression, BM showed the highest level, OM and CM intermediate and FM, HM and DM the lowest levels, however HM and DM had comparable levels to the control.
Collapse
|
28
|
Porcine reproductive and respiratory syndrome virus induces concurrent elevation of High Mobility Group Box-1 protein and pro-inflammatory cytokines in experimentally infected piglets. Cytokine 2019; 113:21-30. [DOI: 10.1016/j.cyto.2018.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/01/2023]
|
29
|
Pahlavani M, Wijayatunga NN, Kalupahana NS, Ramalingam L, Gunaratne PH, Coarfa C, Rajapakshe K, Kottapalli P, Moustaid-Moussa N. Transcriptomic and microRNA analyses of gene networks regulated by eicosapentaenoic acid in brown adipose tissue of diet-induced obese mice. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1523-1531. [PMID: 30261280 PMCID: PMC6298436 DOI: 10.1016/j.bbalip.2018.09.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/01/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022]
Abstract
Brown adipose tissue (BAT) dissipates chemical energy as heat via thermogenesis and protects against obesity by increasing energy expenditure. However, regulation of BAT by dietary factors remains largely unexplored at the mechanistic level. We investigated the effect of eicosapentaenoic acid (EPA) on BAT metabolism. Male C57BL/6J (B6) mice were fed either a high-fat diet (HF, 45% kcal fat) or HF diet supplemented with EPA (HF-EPA, 6.75% kcal EPA) for 11 weeks. RNA sequencing (RNA-Seq) and microRNA (miRNA) profiling were performed on RNA from BAT using Illumina HiSeq and Illumina Genome Analyzer NextSeq, respectively. We conducted pathway analyses using ingenuity pathway analysis software (IPA®) and validated some genes and miRNAs using qPCR. We identified 479 genes that were differentially expressed (2-fold change, n = 3, P ≤ 0.05) in BAT from HF compared to HF-EPA. Genes negatively correlated with thermogenesis such as hypoxia inducible factor 1 alpha subunit inhibitor (Hif1an), were downregulated by EPA. Pathways related to thermogenesis such as peroxisome proliferator-activated receptor (PPAR) were upregulated by EPA while pathways associated with obesity and inflammation such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) were downregulated by EPA. MiRNA profiling identified nine and six miRNAs that were upregulated and downregulated by EPA, respectively (log2 fold change > 1.25, n = 3, P ≤ 0.05). Key regulatory miRNAs which were involved in thermogenesis, such as miR-455-3p and miR-129-5p were validated using qPCR. In conclusion, the depth of transcriptomic and miRNA profiling revealed novel mRNA-miRNA interaction networks in BAT which are involved in thermogenesis, and regulated by EPA.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States
| | - Nadeeja N Wijayatunga
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States
| | - Nishan S Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States; Department of Physiology, University of Peradeniya, Sri Lanka
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, United States
| | - Cristian Coarfa
- Department of Molecular and Cell Biology, Houston, TX, United States
| | - Kimal Rajapakshe
- Department of Molecular and Cell Biology, Houston, TX, United States
| | - Pratibha Kottapalli
- Center for Biotechnology and Genomics, Texas Tech University, Lubbock, TX, United States
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States.
| |
Collapse
|
30
|
Horodyska J, Reyer H, Wimmers K, Trakooljul N, Lawlor PG, Hamill RM. Transcriptome analysis of adipose tissue from pigs divergent in feed efficiency reveals alteration in gene networks related to adipose growth, lipid metabolism, extracellular matrix, and immune response. Mol Genet Genomics 2018; 294:395-408. [PMID: 30483895 DOI: 10.1007/s00438-018-1515-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 11/13/2018] [Indexed: 12/14/2022]
Abstract
Adipose tissue is hypothesized to play a vital role in regulation of feed efficiency (FE; efficiency in converting energy and nutrients into tissue), of which improvement will simultaneously reduce environmental impact and feed cost per pig. The objective of the present study was to sequence the subcutaneous adipose tissue transcriptome in FE-divergent pigs (n = 16) and identify relevant biological processes underpinning observed differences in FE. We previously demonstrated that high-FE pigs were associated with lower fatness when compared to their counterparts. Here, ontology analysis of a total of 209 annotated genes that were differentially expressed at a p < 0.01 revealed establishment of a dense extracellular matrix and inhibition of capillary formation as one underlying mechanism to achieve suppressed adipogenesis. Moreover, mechanisms ensuring an efficient utilization of lipids in high-FE pigs might be orchestrated by upstream regulators including CEBPA and EGF. Consequently, high-FE adipose tissue could exhibit more efficient cholesterol disposal, whilst inhibition of inflammatory and immune response in high-FE pigs may be an indicator of an optimally functioning adipose tissue. Taken together, adipose tissue growth, extracellular matrix formation, lipid metabolism and inflammatory and immune response are key biological events underpinning the differences in FE. Further investigations focusing on elucidating these processes would assist the animal production industry in optimizing strategies related to nutrient utilization and product quality.
Collapse
Affiliation(s)
- Justyna Horodyska
- Teagasc, Food Research Centre, Ashtown, Dublin 15, Ireland.,Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | - Henry Reyer
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany.,Faculty of Agricultural and Environmental Sciences, University Rostock, Rostock, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | - Peadar G Lawlor
- Teagasc, Pig Development Department, AGRIC, Moorepark, Fermoy, Co. Cork, Ireland
| | - Ruth M Hamill
- Teagasc, Food Research Centre, Ashtown, Dublin 15, Ireland.
| |
Collapse
|
31
|
Hundertmark J, Krenkel O, Tacke F. Adapted Immune Responses of Myeloid-Derived Cells in Fatty Liver Disease. Front Immunol 2018; 9:2418. [PMID: 30405618 PMCID: PMC6200865 DOI: 10.3389/fimmu.2018.02418] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered to be one of the most frequent chronic liver diseases worldwide and is associated with an increased risk of developing liver cirrhosis and hepatocellular carcinoma. Hepatic macrophages, mainly comprising monocyte derived macrophages and tissue resident Kupffer cells, are characterized by a high diversity and plasticity and act as key regulators during NAFLD progression, in conjunction with other infiltrating myeloid cells like neutrophils or dendritic cells. The activation and polarization of myeloid immune cells is influenced by dietary components, inflammatory signals like danger-associated molecular patterns (DAMPs) or cytokines as well as gut-derived inflammatory factors such as pathogen-associated molecular patterns (PAMPs). The functionality of myeloid leukocytes in the liver is directly linked to their inflammatory polarization, which is shaped by local and systemic inflammatory mediators such as cytokines, chemokines, PAMPs, and DAMPs. These environmental signals provoke intracellular adaptations in myeloid cells, including inflammasome and transcription factor activation, inflammatory signaling pathways, or switches in cellular metabolism. Dietary changes and obesity also promote a dysbalance in intestinal microbiota, which can facilitate intestinal permeability and bacterial translocation. The aim of this review is to highlight recent findings on the activating pathways of innate immune cells during the progression of NAFLD, dissecting local hepatic and systemic signals, dietary and metabolic factors as well as pathways of the gut-liver axis. Understanding the mechanism by which plasticity of myeloid-derived leukocytes is related to metabolic changes and NAFLD progression may provide options for new therapeutic approaches.
Collapse
Affiliation(s)
- Jana Hundertmark
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Oliver Krenkel
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| |
Collapse
|
32
|
Chen Y, He X, Yuan X, Hong J, Bhat O, Li G, Li PL, Guo J. NLRP3 Inflammasome Formation and Activation in Nonalcoholic Steatohepatitis: Therapeutic Target for Antimetabolic Syndrome Remedy FTZ. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2901871. [PMID: 30140364 PMCID: PMC6081604 DOI: 10.1155/2018/2901871] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/16/2018] [Accepted: 04/26/2018] [Indexed: 12/14/2022]
Abstract
The Nod-like receptor protein 3 (NLRP3) inflammasome activation not only serves as an intracellular machinery triggering inflammation but also produces uncanonical effects beyond inflammation such as changing cell metabolism and increasing cell membrane permeability. The present study was designed to test whether this NLRP3 inflammasome activation contributes to the "two-hit" injury during nonalcoholic steatohepatitis (NASH) and whether it can be a therapeutic target for the action of Fufang Zhenzhu Tiaozhi (FTZ), a widely used herbal remedy for hyperlipidemia and metabolic syndrome in China. We first demonstrated that NLRP3 inflammasome formation and activation as well as lipid deposition occurred in the liver of mice on the high-fat diet (HFD), as shown by increased NLRP3 aggregation, enhanced production of IL-1β and high mobility group box 1 (HMGB1), and remarkable lipid deposition in liver cells. FTZ extracts not only significantly reduced the NLRP3 inflammasome formation and activation but also attenuated the liver steatosis and fibrogenic phenotype changed. In in vitro studies, palmitic acid (PA) was found to increase colocalization of NLRP3 components and enhanced caspase-1 activity in hepatic stellate cells (HSCs), indicating enhanced formation and activation of NLRP3 inflammasomes by PA. PA also increased lipid deposition. Nlrp3 siRNA can reverse this effect by silencing the NLRP3 inflammasome and both with FTZ. In FTZ-treated cells, not only inflammasome formation and activation was substantially attenuated but also lipid deposition in HSCs was blocked. This inhibition of FTZ on lipid deposition was similar to the effects of glycyrrhizin, an HMGB1 inhibitor. Mechanistically, stimulated membrane raft redox signaling platform formation and increased O2•- production by PA to activate NLRP3 inflammasomes in HSCs was blocked by FTZ treatment. It is concluded that FTZ extracts inhibit NASH by its action on both inflammatory response and liver lipid metabolism associated with NLRP3 inflammasome formation and activation.
Collapse
Affiliation(s)
- Yu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xingxiang He
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
| | - Xinxu Yuan
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jinni Hong
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Owais Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jiao Guo
- Department of Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Joint Laboratory of Guangdong Province and Hongkong and Macao Regions on Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
33
|
High-Fat Diet Increases HMGB1 Expression and Promotes Lung Inflammation in Mice Subjected to Mechanical Ventilation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7457054. [PMID: 29619146 PMCID: PMC5830287 DOI: 10.1155/2018/7457054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/16/2017] [Accepted: 12/21/2017] [Indexed: 12/24/2022]
Abstract
This study aims to evaluate the effects of a high-fat diet and mechanical ventilation on the pulmonary and systemic inflammatory response in C57BL/6 mice. Male C57BL/6 mice were divided into two groups: one received a standard diet, and the other received a high-fat diet. After 10 weeks, the groups were further divided into two groups each: control group (CG), mechanical ventilation group (MVG), diet group (DG), and diet mechanical ventilation group (DMVG). MVG and DMVG underwent mechanical ventilation for 60 minutes. All animals were euthanized for subsequent analysis. Animals receiving a high-fat diet presented higher body mass, adipose index, and greater adipocyte area. In the lung, the expression of HMGB1 was greater in DG and DMVG than in CG and MVG. CCL2 and IL-22 levels in MVG and DMVG were increased compared to those in CG and DG, whereas IL-10 and IL-17 were decreased. Superoxide dismutase activity was higher in MVG and DMVG than in CG. Catalase activity was lower in DG than in CG, and in MV groups, it was lower than that in CG and DG. MV and obesity promote inflammation and pulmonary oxidative stress in adult C57BL/6 mice.
Collapse
|
34
|
Zhang J, Zhang L, Zhang S, Yu Q, Xiong F, Huang K, Wang CY, Yang P. HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose tissue in obesity. Mol Cell Endocrinol 2017; 454:103-111. [PMID: 28619625 DOI: 10.1016/j.mce.2017.06.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/17/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022]
Abstract
Obesity has emerged as an imminent global public health concern over the past several decades. It has now become evident that obesity is characterized by the persistent and low-grade inflammation in the adipose tissue, and serves as an independent risk factor for many metabolic disorders such as diabetes and cardiovascular disease. Particularly, adipocytes originated from obese mice and humans likely predominate necrosis upon stressful insults, leading to passive release of cellular contents including the high mobility group box 1 (HMGB1) into the extracellular milieu. Extracellular HMGB1 acts as an innate alarmin to stimulate the activation of resident immune cells in the adipose tissue. Upon activation, those resident immune cells actively secrete additional HMGB1, which in turn activates/recruits additional immune cells, and induces adipocyte death. This review summarizes those novel discoveries in terms of HMGB1 in the initiation and maintenance of chronic inflammatory state in adipose tissue in obesity, and discusses its potential application in clinical settings.
Collapse
Affiliation(s)
- Jing Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Lei Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Shu Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Qilin Yu
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Fei Xiong
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
| | - Ping Yang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
| |
Collapse
|
35
|
Corrêa LH, Corrêa R, Farinasso CM, de Sant'Ana Dourado LP, Magalhães KG. Adipocytes and Macrophages Interplay in the Orchestration of Tumor Microenvironment: New Implications in Cancer Progression. Front Immunol 2017; 8:1129. [PMID: 28970834 PMCID: PMC5609576 DOI: 10.3389/fimmu.2017.01129] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022] Open
Abstract
Inflammation has been known as one of the main keys to the establishment and progression of cancers. Chronic low-grade inflammation is also a strategic condition that underlies the causes and development of metabolic syndrome and obesity. Moreover, obesity has been largely related to poor prognosis of tumors by modulating tumor microenvironment with secretion of several inflammatory mediators by tumor-associated adipocytes (TAAs), which can modulate and recruit tumor-associated macrophages. Thus, the understanding of cellular and molecular mechanisms that underlay and link inflammation, obesity, and cancer is crucial to identify potential targets that interfere with this important route. Knowledge about the exact role of each component of the tumor microenvironment is not yet fully understood, but the new insights in literature highlight the essential role of adipocytes and macrophages interplay as key factor to determine the fate of cancer progression. In this review article, we focus on the functions of adipocytes and macrophages orchestrating cellular and molecular mechanisms that lead to inflammatory modulation in tumor microenvironment, which will be crucial to cancer establishment. We also emphasized the mechanisms by which the tumor promotes itself by recruiting and polarizing macrophages, discussing the role of adipocytes in this process. In addition, we discuss here the newest possible anticancer therapeutic treatments aiming to retard the development of the tumor based on what is known about cancer, adipocyte, and macrophage polarization.
Collapse
Affiliation(s)
- Luís Henrique Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Rafael Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Cecília Menezes Farinasso
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | | | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
36
|
Rider P, Voronov E, Dinarello CA, Apte RN, Cohen I. Alarmins: Feel the Stress. THE JOURNAL OF IMMUNOLOGY 2017; 198:1395-1402. [PMID: 28167650 DOI: 10.4049/jimmunol.1601342] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/27/2016] [Indexed: 12/20/2022]
Abstract
Over the last decade, danger-associated molecular pattern molecules, or alarmins, have been recognized as signaling mediators of sterile inflammatory responses after trauma and injury. In contrast with the accepted passive release models suggested by the "danger hypothesis," it was recently shown that alarmins can also directly sense and report damage by signaling to the environment when released from live cells undergoing physiological stress, even without loss of subcellular compartmentalization. In this article, we review the involvement of alarmins such as IL-1α, IL-33, IL-16, and high-mobility group box 1 in cellular and physiological stress, and suggest a novel activity of these molecules as central initiators of sterile inflammation in response to nonlethal stress, a function we denote "stressorins." We highlight the role of posttranslational modifications of stressorins as key regulators of their activity and propose that targeted inhibition of stressorins or their modifiers could serve as attractive new anti-inflammatory treatments for a broad range of diseases.
Collapse
Affiliation(s)
- Peleg Rider
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | | | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Idan Cohen
- Faculty of Medicine, Galilee Medical Center, Nahariya Hospital, 22100 Nahariya, Israel
| |
Collapse
|
37
|
The Pathogenesis of Obesity-Associated Adipose Tissue Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:221-245. [PMID: 28585201 DOI: 10.1007/978-3-319-48382-5_9] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Ahmad R, Kochumon S, Thomas R, Atizado V, Sindhu S. Increased adipose tissue expression of TLR8 in obese individuals with or without type-2 diabetes: significance in metabolic inflammation. JOURNAL OF INFLAMMATION-LONDON 2016; 13:38. [PMID: 27980459 PMCID: PMC5146894 DOI: 10.1186/s12950-016-0147-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 12/05/2016] [Indexed: 12/29/2022]
Abstract
Background The innate immune Toll-like receptors (TLRs) 2/4 are important players in chronic low-grade inflammation called metabolic inflammation in obesity and type-2 diabetes (T2D). While TLR2/4 expression changes associated with metabolic inflammation are known, the adipose tissue expression of endocytic TLR8, which is expressed by all major macrophage subsets, remain unclear. We, therefore, determined the TLR8 mRNA/protein expression in the adipose tissue samples from lean, overweight, and obese individuals with or without T2D. Methods Subcutaneous fat biopsy samples were collected from 49 non-diabetic (23 obese, 17 overweight, and nine lean) and 45 T2D (32 obese, ten overweight, and three lean) individuals. TLR8 gene expression was determined using real-time RT-PCR and TLR8 protein expression was assessed by both immunohistochemistry and confocal microscopy. The changes in TLR8 expression were compared with those of macrophage markers, proinflammatory cytokines/chemokines, and surface TLRs/adapter proteins. The data were analyzed using t-test/Mann-Whitney U-test, Pearson’s correlation, and multiple regression test. Results The data show that in obese non-diabetic/T2D individuals, TLR8 gene expression was significantly upregulated as compared with lean individuals which correlated with body mass index (BMI) and body fat percentage in non-diabetic population (P < 0.05). As expected, TLR8 adipose tissue protein expression in non-diabetic/T2D obese individuals was also higher than that of overweight/lean counterparts. In non-diabetic/T2D individuals, TLR8 gene expression associated (P < 0.05) with the expression of CD68, CD11c, CD86, and CD163 macrophage markers. Also, in these individuals, TLR8 gene expression correlated positively (P < 0.05) with adipose tissue expression of TNF-α, IL-18, and IL-8 as well as with systemic CRP levels (in non-diabetics). TLR8 expression was also associated with TLR4/TLR2 and MyD88 expression in the adipose tissue. Conclusions The elevated adipose tissue expression of TLR8 in obesity/T2D has consensus with inflammatory signatures and may thus represent an immune marker of metabolic inflammation. Electronic supplementary material The online version of this article (doi:10.1186/s12950-016-0147-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rasheed Ahmad
- Immunology & Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Shihab Kochumon
- Immunology & Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Reeby Thomas
- Immunology & Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Valerie Atizado
- Tissue Bank Core Facility, Dasman Diabetes Institute (DDI), Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Sardar Sindhu
- Immunology & Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| |
Collapse
|
39
|
High Mobility Group Box-1: A Missing Link between Diabetes and Its Complications. Mediators Inflamm 2016; 2016:3896147. [PMID: 27847406 PMCID: PMC5099456 DOI: 10.1155/2016/3896147] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/03/2016] [Indexed: 12/13/2022] Open
Abstract
High mobility group box-1 (HMGB-1), a damage-associated molecular pattern, can be actively or passively released from various cells under different conditions and plays a pivotal role in the pathogenesis of inflammation and angiogenesis-dependent diseases. More and more evidence suggests that inflammation, in addition to its role in progression of diabetes, also promotes initiation and development of diabetic complications. In this review, we focus on the role of HMGB-1 in diabetes-related complications and the therapeutic strategies targeting HMGB-1 in diabetic complications.
Collapse
|
40
|
Gunasekaran MK, Virama-Latchoumy AL, Girard AC, Planesse C, Guérin-Dubourg A, Ottosson L, Andersson U, Césari M, Roche R, Hoareau L. TLR4-dependant pro-inflammatory effects of HMGB1 on human adipocyte. Adipocyte 2016; 5:384-388. [PMID: 27994953 PMCID: PMC5160392 DOI: 10.1080/21623945.2016.1245818] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/03/2016] [Accepted: 10/03/2016] [Indexed: 10/20/2022] Open
Abstract
Chronic low grade inflammation is one of the major metabolic disorders in case of obesity and associated pathologies. By its important secretion function, the role of adipose tissue in this metabolic low grade inflammation is well known. Recently, it was demonstrated that the alarmin high mobility group box protein 1 (HMGB1) is involved in obesity-related pathologies by its increased serum levels in obese compared to normal weight individuals, and by its pro-inflammatory effects. However, the role of HMGB1 on adipocytes inflammation is poorly documented and we propose to investigate this point. Primary culture of human subcutaneous adipocytes were performed from human adipose tissue samples. Cells were treated with recombinant HMGB1 with/without anti-TLR4 antibody and inhibitors of NF-κB and P38 MAPK. Supernatants were collected for IL-6 and MCP-1 ELISA. HMGB1 initiates Toll-like receptor 4 (TLR4)-dependent activation of inflammation through the downstream NF-κB and P38 MAPK signaling pathway to upregulate the secretion of the pro-inflammatory cytokine IL-6. HMGB1 has pro-inflammatory effects on adipocytes. This reinforces the role of TLR4 in adipose tissue inflammation and antagonizing the HMGB1 inflammatory pathway could bring on new therapeutic targets to counteract obesity-associated pathologies.
Collapse
Affiliation(s)
- Manoj Kumar Gunasekaran
- Inserm, UMR 1188, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
| | - Anne-Laurence Virama-Latchoumy
- Inserm, UMR 1188, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
| | - Anne-Claire Girard
- Inserm, UMR 1188, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
- Centre Hospitalier Universitaire (CHU) de La Réunion
| | - Cynthia Planesse
- Inserm, UMR 1188, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
- Stemcis, plateforme CYROI, Sainte-Clotilde, France
| | | | - Lars Ottosson
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stokholm, Sweden
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stokholm, Sweden
| | - Maya Césari
- Inserm, UMR 1188, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
| | - Régis Roche
- Stemcis, plateforme CYROI, Sainte-Clotilde, France
| | - Laurence Hoareau
- Inserm, UMR 1188, Diabéte athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
- Stemcis, plateforme CYROI, Sainte-Clotilde, France
| |
Collapse
|
41
|
Shimizu T, Yamakuchi M, Biswas KK, Aryal B, Yamada S, Hashiguchi T, Maruyama I. HMGB1 is secreted by 3T3-L1 adipocytes through JNK signaling and the secretion is partially inhibited by adiponectin. Obesity (Silver Spring) 2016; 24:1913-21. [PMID: 27430164 DOI: 10.1002/oby.21549] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/13/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Obesity is a chronic inflammatory disease, and adipocytes contribute to obesity-associated inflammation by releasing inflammatory mediators. High mobility group box 1 (HMGB1), a highly conserved DNA-binding protein, mainly localized to cell nuclei, has been recently recognized as an innate pro-inflammatory mediator when released extracellularly. It was hypothesized that HMGB1 is an adipocytokine that acts as an innate pro-inflammatory mediator in white adipose tissue (WAT) of patients with obesity and is associated with insulin resistance. Additionally, it was hypothesized that HMGB1 secretion is regulated by adiponectin. METHODS 3T3-L1 cells were differentiated into mature adipocytes. After tumor necrosis factor-α (TNF-α) stimulation, HMGB1 in culture media was measured. Localizations of HMGB1 in 3T3-L1 adipocytes and human WAT were examined by immunostaining. RESULTS HMGB1 was secreted from TNF-α-induced 3T3-L1 adipocytes through JNK signaling. HMGB1-activated MAP kinases (ERK1/2, JNK) and suppressed insulin-stimulated Akt phosphorylation in 3T3-L1 adipocytes. The cytoplasm in 3T3-L1 adipocytes and adipocytes of WAT from a patient with obesity was intensely stained with HMGB1. Adiponectin partially inhibited TNF-α-induced HMGB1 secretion from 3T3-L1 adipocytes. CONCLUSIONS These findings suggest that HMGB1 is a pro-inflammatory adipocytokine involved in WAT inflammation and insulin resistance in patients with obesity, which may contribute to the progression of metabolic syndrome, and that adiponectin protects against HMGB1-induced adipose tissue inflammation.
Collapse
Affiliation(s)
- Toshiaki Shimizu
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kamal Krishna Biswas
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Bibek Aryal
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
42
|
Pahwa R, Jialal I. The role of the high-mobility group box1 protein-Toll like receptor pathway in diabetic vascular disease. J Diabetes Complications 2016; 30:1186-91. [PMID: 27037040 DOI: 10.1016/j.jdiacomp.2016.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Increased Toll like receptors (TLRs) especially 2 and 4 have been demonstrated in obesity, metabolic syndrome (MetS) and diabetes resulting in increased cellular inflammation. Since we have shown increased TLR2 and 4 activities in both T1DM and T2DM and MetS, we wanted to elucidate the mechanisms of this sterile inflammation. In T1DM, T2DM and MetS we have shown that high mobility group box 1 protein (HMGB-1), a non-histone DNA binding protein is increased and could be a potential activator of TLRs since it has previously shown to activate TLR2, 4 and 9. We examined the role of HMGB-1 in patients and animal models of diabetes and MetS to determine how important it is as an activator of TLR mediated inflammation and its role in diabetic vascular complications. METHODS A Medline search was conducted using the terms HMGB-1, TLRs and diabetes. RESULTS HMGB-1 levels are increased in patients with diabetes and MetS, and associated with increased biomediators of inflammation. Furthermore data supported a role of HMGB-1 in both diabetic microvascular and macrovascular complications. CONCLUSIONS HMGB-1 interaction with TLRs is implicated in diabetic complications and could be important therapeutic target.
Collapse
Affiliation(s)
- Roma Pahwa
- Laboratory of Atherosclerosis and Metabolic Research, Department of Pathology and InternalMedicine, University of California Davis Medical Center, Sacramento, CA
| | - Ishwarlal Jialal
- Laboratory of Atherosclerosis and Metabolic Research, Department of Pathology and InternalMedicine, University of California Davis Medical Center, Sacramento, CA; Medical Services (Endocrinology and Metabolism), VA Medical Center, Mather, CA.
| |
Collapse
|
43
|
Azzarà A, Pirillo C, Giovannini C, Federico G, Scarpato R. Different repair kinetic of DSBs induced by mitomycin C in peripheral lymphocytes of obese and normal weight adolescents. Mutat Res 2016; 789:9-14. [PMID: 27174706 DOI: 10.1016/j.mrfmmm.2016.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 03/21/2016] [Accepted: 05/01/2016] [Indexed: 01/13/2023]
Abstract
In 2013, 42 million children under the age of 5 years were overweight or obese. In the context of obesity, we recently showed that (1) peripheral lymphocytes of obese children/adolescents had an 8-fold increase in double strand breaks (DSBs), expressed as g-H2AX foci, than normal weight adolescents, and (2) 30% of the damage was retained into chromosome mutations. Thus, we investigated DSBs repair efficiency in a group of obese adolescents assessing the kinetic of H2AX phosphorylation in mitomycin C (MMC)-treated lymphocytes harvested 2 h- or 4 h-post mutagen treatment. According to our previous studies, these harvesting times represent the peak of DSBs induction and the time in which an appreciable DSBs reduction was observed. In addition, we evaluated the expression of the high mobility group box-1 protein (HMGB1), a chromatin remodelling protein involved in DSBs repair and obesity. Compared to normal weight adolescents, obese subjects 1) showed higher levels of g-H2AX foci at either 2 h- (0.239±0.041 vs. 0.473±0.048, P=0.0016) or 4 h- (0.150±0.026 vs. 0.255±0.030, P=0.0198) post mutagen treatment, and 2) have repaired a greater amount of the initial lesions (0.088±0.033 vs. 0.218±0.045, P=0.0408). Concordantly, 1) HMGB1 levels of obese individuals increased and decreased at 2h- or 4 h-post mutagen treatment, respectively, and 2) the opposite occurred for the normal weight adolescents where the protein was down-expressed at 2h and over-expressed at 4h. In conclusion, lymphocytes of obese and normal weight adolescents showed a distinct temporal kinetic of repairing MMC-induced DSBs, together with a different expression of HMGB1. The finding that obesity may modulate the repair of DNA damage induced in lymphocytes by genotoxic agents should be confirmed by further experiments.
Collapse
Affiliation(s)
- Alessia Azzarà
- Unità di Genetica, Dipartimento di Biologia, Pisa University, Pisa, Italy
| | - Chiara Pirillo
- Unità di Genetica, Dipartimento di Biologia, Pisa University, Pisa, Italy
| | | | - Giovanni Federico
- Unità di Endocrinologia Pediatrica e Diabete, Dipartimento di Medicina Clinica e Sperimentale Pisa University, Pisa, Italy
| | - Roberto Scarpato
- Unità di Genetica, Dipartimento di Biologia, Pisa University, Pisa, Italy; Research Center of Nutraceuticals and Food for Health, University of Pisa, Pisa, Italy.
| |
Collapse
|
44
|
Abstract
Inflammation originating from the adipose tissue is considered to be one of the main driving forces for the development of insulin resistance and type 2 diabetes in obese individuals. Although a plethora of different immune cells shapes adipose tissue inflammation, this review is specifically focused on the contribution of macrophages that reside in adipose tissue in lean and obese conditions. Both conventional and tissue-specific functions of adipose tissue macrophages (ATMs) in lean and obese adipose tissue are discussed and linked with metabolic and inflammatory changes that occur during the development of obesity. Furthermore, we will address various circulating and adipose tissue-derived triggers that may be involved in shaping the ATM phenotype and underlie ATM function in lean and obese conditions. Finally, we will highlight how these changes affect adipose tissue inflammation and may be targeted for therapeutic interventions to improve insulin sensitivity in obese individuals.
Collapse
Affiliation(s)
- Lily Boutens
- Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Nutrition, Metabolism and Genomics Group, Wageningen University, Bomenweg 2, 6703 HD, Wageningen, the Netherlands
| | - Rinke Stienstra
- Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
- Nutrition, Metabolism and Genomics Group, Wageningen University, Bomenweg 2, 6703 HD, Wageningen, the Netherlands.
| |
Collapse
|
45
|
miR-155 Deletion in Female Mice Prevents Diet-Induced Obesity. Sci Rep 2016; 6:22862. [PMID: 26953132 PMCID: PMC4782173 DOI: 10.1038/srep22862] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 02/22/2016] [Indexed: 12/28/2022] Open
Abstract
Obesity is a growing epidemic in developed countries. Obese individuals are susceptible to comorbidities, including cardiovascular disease and metabolic disorder. Increasing the ability of adipose tissue to expend excess energy could improve protection from obesity. One promising target is microRNA (miR)-155-5p. We demonstrate that deletion of miR-155 (-5p and -3p) in female mice prevents diet-induced obesity. Body weight gain did not differ between wild-type (WT) and miR-155 knockout (KO) mice fed control diet (CD); however, miR-155 KO mice fed high-fat diet (HFD) gained 56% less body weight and 74% less gonadal white adipose tissue (WAT) than WT mice. Enhanced WAT thermogenic potential, brown adipose tissue differentiation, and/or insulin sensitivity might underlie this obesity resistance. Indeed, miR-155 KO mice on HFD had 21% higher heat release than WT HFD mice. Compared to WT adipocytes, miR-155 KO adipocytes upregulated brown (Ucp1, Cidea, Pparg) and white (Fabp4, Pnpla2, AdipoQ, Fasn) adipogenic genes, and glucose metabolism genes (Glut4, Irs1). miR-155 deletion abrogated HFD-induced adipocyte hypertrophy and WAT inflammation. Therefore, miR-155 deletion increases adipogenic, insulin sensitivity, and energy uncoupling machinery, while limiting inflammation in WAT, which together could restrict HFD-induced fat accumulation. Our results identify miR-155 as a novel candidate target for improving obesity resistance.
Collapse
|
46
|
Hosick PA, AlAmodi AA, Hankins MW, Stec DE. Chronic treatment with a carbon monoxide releasing molecule reverses dietary induced obesity in mice. Adipocyte 2016; 5:1-10. [PMID: 27144091 PMCID: PMC4836479 DOI: 10.1080/21623945.2015.1038443] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 03/31/2015] [Accepted: 03/31/2015] [Indexed: 01/23/2023] Open
Abstract
Chronic, low level treatment with a carbon monoxide releasing molecule (CO-RM), CORM-A1, has been shown to prevent the development of obesity in response to a high fat diet. The objective of this study was to test the hypothesis that chronic, low level treatment with this CO-RM can reverse established obesity via a mechanism independent of food intake. Dietary induced obese mice were treated with CORM-A1, the inactive compound iCORM-A1, or saline every 48 hours for 30 weeks while maintained on a high fat (60%) diet. Chronic treatment with CORM-A1 resulted in a 33% decrease from initial body weight over the 30 week treatment period while treatment with iCORM and saline were associated with 18 and 25% gain in initial body weight over the same time frame. Chronic treatment with CORM-A1 did not affect food intake or activity but resulted in a significant increase in metabolism. CORM-A1 treatment also resulted in lower fasting blood glucose, improvement in insulin sensitivity and decreased heptatic steatosis. Chronic treatment with CO releasing molecules can reverse dietary induced obesity and normalize insulin resistance independent of changes in food intake or activity. These findings are likely though a mechanism which increases metabolism.
Collapse
|
47
|
Garcia-Martinez I, Shaker ME, Mehal WZ. Therapeutic Opportunities in Damage-Associated Molecular Pattern-Driven Metabolic Diseases. Antioxid Redox Signal 2015; 23:1305-15. [PMID: 26055926 PMCID: PMC4685500 DOI: 10.1089/ars.2015.6383] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Sterile inflammation is a common finding present in various metabolic disorders. This type of inflammation is mediated by damage-associated molecular patterns (DAMPs) that are released upon cellular injury to activate pattern recognition receptors on innate immune cells and amplify organ damage. RECENT ADVANCES In the last decade, DAMPs, such as high-mobility group protein B1, nucleic acids (DNA, RNA), adenosine triphosphate, and other metabolites, were found to contribute to the inflammatory response in diabetes, gout, obesity, steatohepatitis, and atherosclerosis. Varied receptors, including Toll-like receptors (TLRs), the purinergic P2X(7) receptors, and nucleotide-binding domain, and leucine-rich repeat protein 3 (NLRP3)-inflammasome sense DAMPs and DAMP-like molecules and release the proinflammatory cytokines, interleukin (IL)-1β and IL-18. CRITICAL ISSUES Available therapeutic approaches that interfered with the signaling of TLRs, P2X(7), NLRP3-inflammasome, and IL-1β showed encouraging results in metabolic diseases, which will be also highlighted in this review. FUTURE DIRECTIONS It is important to understand the origination of DAMPs and how they contribute to the inflammatory response in metabolic disorders to develop selective and efficient therapeutics for intervention.
Collapse
Affiliation(s)
- Irma Garcia-Martinez
- 1 Section of Digestive Diseases, Department of Internal Medicine, Yale University , New Haven, Connecticut
| | - Mohamed E Shaker
- 1 Section of Digestive Diseases, Department of Internal Medicine, Yale University , New Haven, Connecticut.,2 Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University , Mansoura, Egypt
| | - Wajahat Z Mehal
- 1 Section of Digestive Diseases, Department of Internal Medicine, Yale University , New Haven, Connecticut
| |
Collapse
|
48
|
Wang H, Qu H, Deng H. Plasma HMGB-1 Levels in Subjects with Obesity and Type 2 Diabetes: A Cross-Sectional Study in China. PLoS One 2015; 10:e0136564. [PMID: 26317615 PMCID: PMC4552731 DOI: 10.1371/journal.pone.0136564] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/05/2015] [Indexed: 12/22/2022] Open
Abstract
Object To detect the levels of plasma High-Mobility Group Box-1(HMGB1) in Chinese subject with obesity and type 2 diabetes mellitus (T2DM), and to investigate the correlations between plasma HMGB1 concentration and parameters of body fat, insulin resistance (IR) metabolism and inflammation. Methods This study recruited 79 normal glucose tolerance (NGT) subjects and 76 newly diagnosed T2DM patients. NGT and T2DM groups were divided into normal weight (NW) and obese (OB)subgroups respectively. Anthropometric parameters such as height, weight, waist circumference, hip circumference and blood pressure were measured. Plasma concentrations of HMGB1, IL-6, fasting plasma glucose (FPG), 2 hours post challenge plasma glucose (2hPG), serum lipid, glycated hemoglobin (HbA1C) and fasting insulin (FINS) were examined. The homeostasis model assessment (HOMA) was performed to assess IR status. Results Plasma HMGB1 levels were higher in T2DM group than that in NGT group. The concentrations of serum HMGB1 were also higher in subjects with OB than those in subjects with NW both in NGT and T2DM groups. Plasma levels of HMGB1 were positively correlated with waist hip ratio (WHR), blood pressure, FPG, FINS, HOMA-IR, TG, IL-6 and negatively correlated with HOMA-βand high-density lipoprotein-cholesterol (HDL-c) independent of age, gender and BMI. Plasma levels of HMGB1 were significantly correlated with diabetes in fully adjusted models. Conclusion Plasma HMGB1 levels were increased in Chinese subjects with pure T2DM, which might be caused by IR. Serum HMGB1 participated in the pathological process of obesity and T2DM via its proinflammatory effect.
Collapse
Affiliation(s)
- Hang Wang
- M.D. Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Qu
- M.D. Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huacong Deng
- M.D. Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
49
|
Jialal I, Devaraj S, Bettaieb A, Haj F, Adams-Huet B. Increased adipose tissue secretion of Fetuin-A, lipopolysaccharide-binding protein and high-mobility group box protein 1 in metabolic syndrome. Atherosclerosis 2015; 241:130-7. [PMID: 25978344 DOI: 10.1016/j.atherosclerosis.2015.04.814] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/24/2015] [Accepted: 04/24/2015] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Adipose Tissue (AT) dysregulation contributes to the pro-inflammatory state and insulin resistance of Metabolic Syndrome (MetS). We examined AT secretion of the hepatokine, Fetuin-A, LBP, sCD14 and HMGB-1, and toll-like receptor 2 and 4 protein levels in MetS and controls. DESIGN AND METHODS Secreted levels of Fetuin-A, LBP, HMGB-1 and sCD14 and TLR2 and TLR4 protein in AT of controls and MetS patients were assayed. Also mRNA and protein for Fetuin-A, LBP, sCD14 and HMGB-1 were studied in subcutaneous fat depot of mice and during adipocyte differentiation. RESULTS Secretion of Fetuin-A, LBP and HMGB-1 from AT were significantly increased in MetS (n = 28) compared to controls (n = 25), even after adjustment for adiposity. There were no significant differences in sCD14. Both LBP and Fetuin-A correlated significantly with HOMA-IR and increased significantly with increasing features of MetS. There was a significant increase in AT TLR2 and TLR4 protein in MetS compared to controls. Expression of Fetuin-A and LBP were significantly higher in subcutaneous white adipose tissue of HFD fed mice as well as in ob/ob mice compared to C57BL6/J control mice (n = 6 per group). Additionally mRNA and protein levels of FetA, LBP and HMGB-1 increased during differentiation of 3T3-L1 adipocytes. CONCLUSIONS We make the novel observation of increased secretion of Fetuin A, LBP and HMGB-1 from AT and hypothesize that these engage TLRs in AT and other tissues contributing to the pro-inflammatory state and insulin resistance of MetS.
Collapse
Affiliation(s)
- Ishwarlal Jialal
- Laboratory for Atherosclerosis and Metabolic Research, Department of Pathology, Division of Endocrinology, Diabetes and Metabolism, University of California Davis Medical Center, Sacramento, CA, USA; Medical Services, VA Medical Center, Mather, CA, USA.
| | - Sridevi Devaraj
- Department of Pathology & Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Fawaz Haj
- Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Beverley Adams-Huet
- Department of Clinical Sciences, Division of Biostatistics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
50
|
Song F, Hurtado del Pozo C, Rosario R, Zou YS, Ananthakrishnan R, Xu X, Patel PR, Benoit VM, Yan SF, Li H, Friedman RA, Kim JK, Ramasamy R, Ferrante AW, Schmidt AM. RAGE regulates the metabolic and inflammatory response to high-fat feeding in mice. Diabetes 2014; 63:1948-65. [PMID: 24520121 PMCID: PMC4030112 DOI: 10.2337/db13-1636] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In mammals, changes in the metabolic state, including obesity, fasting, cold challenge, and high-fat diets (HFDs), activate complex immune responses. In many strains of rodents, HFDs induce a rapid systemic inflammatory response and lead to obesity. Little is known about the molecular signals required for HFD-induced phenotypes. We studied the function of the receptor for advanced glycation end products (RAGE) in the development of phenotypes associated with high-fat feeding in mice. RAGE is highly expressed on immune cells, including macrophages. We found that high-fat feeding induced expression of RAGE ligand HMGB1 and carboxymethyllysine-advanced glycation end product epitopes in liver and adipose tissue. Genetic deficiency of RAGE prevented the effects of HFD on energy expenditure, weight gain, adipose tissue inflammation, and insulin resistance. RAGE deficiency had no effect on genetic forms of obesity caused by impaired melanocortin signaling. Hematopoietic deficiency of RAGE or treatment with soluble RAGE partially protected against peripheral HFD-induced inflammation and weight gain. These findings demonstrate that high-fat feeding induces peripheral inflammation and weight gain in a RAGE-dependent manner, providing a foothold in the pathways that regulate diet-induced obesity and offering the potential for therapeutic intervention.
Collapse
Affiliation(s)
- Fei Song
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Carmen Hurtado del Pozo
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Rosa Rosario
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Yu Shan Zou
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Radha Ananthakrishnan
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Xiaoyuan Xu
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, NY
| | - Payal R Patel
- Program in Molecular Medicine and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Vivian M Benoit
- Program in Molecular Medicine and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Shi Fang Yan
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Huilin Li
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University School of Medicine, New York, NY
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center and Department of Biomedical Informatics, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Jason K Kim
- Program in Molecular Medicine and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Anthony W Ferrante
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, NY
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| |
Collapse
|