101
|
Fooks AN, Beppu LY, Frias AB, D'Cruz LM. Adipose tissue regulatory T cells: differentiation and function. Int Rev Immunol 2022; 42:323-333. [PMID: 35212593 PMCID: PMC9402810 DOI: 10.1080/08830185.2022.2044808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 10/19/2022]
Abstract
Rising obesity levels, worldwide, are resulting in substantial increases in cardiovascular disease, diabetes, kidney disease, musculoskeletal disorders, and certain cancers, and obesity-associated illnesses are estimated to cause ∼4 million deaths worldwide per year. A common theme in this disease epidemic is the chronic systemic inflammation that accompanies obesity. CD4+ Foxp3+ regulatory T cells residing in visceral adipose tissues (VAT Tregs) are a unique immune cell population that play essential functions in restricting obesity-associated systemic inflammation through regulation of adipose tissue homeostasis. The distinct transcriptional program that defines VAT Tregs has been described, but directly linking VAT Treg differentiation and function to improving insulin sensitivity has proven more complex. Here we review new findings which have clarified how VAT Tregs differentiate, and how distinct VAT Treg subsets regulate VAT homeostasis, energy expenditure, and insulin sensitivity.
Collapse
Affiliation(s)
- Allen N Fooks
- Department of Immunology, University of Pittsburgh, Biomedical Science Tower, Pittsburgh, PA, USA
| | - Lisa Y Beppu
- Department of Immunology, University of Pittsburgh, Biomedical Science Tower, Pittsburgh, PA, USA
| | - Adolfo B Frias
- Department of Immunology, University of Pittsburgh, Biomedical Science Tower, Pittsburgh, PA, USA
| | - Louise M D'Cruz
- Department of Immunology, University of Pittsburgh, Biomedical Science Tower, Pittsburgh, PA, USA
| |
Collapse
|
102
|
Zhao X, Liu H, Pan Y, Liu Y, Zhang F, Ao H, Zhang J, Xing K, Wang C. Identification of Potential Candidate Genes From Co-Expression Module Analysis During Preadipocyte Differentiation in Landrace Pig. Front Genet 2022; 12:753725. [PMID: 35178067 PMCID: PMC8843850 DOI: 10.3389/fgene.2021.753725] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Preadipocyte differentiation plays an important role in lipid deposition and affects fattening efficiency in pigs. In the present study, preadipocytes isolated from the subcutaneous adipose tissue of three Landrace piglets were induced into mature adipocytes in vitro. Gene clusters associated with fat deposition were investigated using RNA sequencing data at four time points during preadipocyte differentiation. Twenty-seven co-expression modules were subsequently constructed using weighted gene co-expression network analysis. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses revealed three modules (blue, magenta, and brown) as being the most critical during preadipocyte differentiation. Based on these data and our previous differentially expressed gene analysis, angiopoietin-like 4 (ANGPTL4) was identified as a key regulator of preadipocyte differentiation and lipid metabolism. After inhibition of ANGPTL4, the expression of adipogenesis-related genes was reduced, except for that of lipoprotein lipase (LPL), which was negatively regulated by ANGPTL4 during preadipocyte differentiation. Our findings provide a new perspective to understand the mechanism of fat deposition.
Collapse
Affiliation(s)
- Xitong Zhao
- Beijing Shunxin Agriculture Co., Ltd., Beijing, China.,China Agricultural University, Beijing, China
| | - Huatao Liu
- China Agricultural University, Beijing, China
| | - Yongjie Pan
- Beijing Shunxin Agriculture Co., Ltd., Beijing, China
| | - Yibing Liu
- China Agricultural University, Beijing, China
| | | | - Hong Ao
- Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jibin Zhang
- City of Hope National Medical Center, Duarte, CA, United States
| | - Kai Xing
- Beijing University of Agriculture, Beijing, China
| | | |
Collapse
|
103
|
Jack BU, Mamushi M, Viraragavan A, Dias S, Pheiffer C. Comparing the effects of tumor necrosis factor alpha, lipopolysaccharide and palmitic acid on lipid metabolism and inflammation in murine 3T3-L1 adipocytes. Life Sci 2022; 297:120422. [DOI: 10.1016/j.lfs.2022.120422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 01/12/2023]
|
104
|
Emerging Roles of Adipose Tissue in the Pathogenesis of Psoriasis and Atopic Dermatitis in Obesity. JID INNOVATIONS 2022; 2:100064. [PMID: 35024685 PMCID: PMC8659781 DOI: 10.1016/j.xjidi.2021.100064] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022] Open
Abstract
Obesity is a growing epidemic worldwide, and it is also considered a major environmental factor contributing to the pathogenesis of inflammatory skin diseases, including psoriasis (PSO) and atopic dermatitis (AD). Moreover, obesity worsens the course and impairs the treatment response of these inflammatory skin diseases. Emerging evidence highlights that hypertrophied adipocytes and infiltrated immune cells secrete a variety of molecules, including fatty acids and adipokines, such as leptin, adiponectin, and a panel of cytokines/chemokines that modulate our immune system. In this review, we describe how adipose hypertrophy leads to a chronic low-grade inflammatory state in obesity and how obesity-related inflammatory factors are involved in the pathogenesis of PSO and/or AD. Finally, we discuss the potential role of antimicrobial peptides, mechanical stress and impairment of epidermal barrier function mediated by fast expansion, and dermal fat in modulating skin inflammation. Together, this review summarizes the current literature on how obesity is associated with the pathogenesis of PSO and AD, highlighting the potentially important but overlooked immunomodulatory role of adipose tissue in the skin.
Collapse
Key Words
- AD, atopic dermatitis
- AMP, antimicrobial peptide
- AT, adipose tissue
- BAT, brown adipose tissue
- BMI, body mass index
- CI, confidence interval
- DC, dendritic cell
- DIO, diet-induced obesity
- FFA, free fatty acid
- HFD, high-fat diet
- KC, keratinocyte
- OA, oleic acid
- PA, palmitic acid
- PSO, psoriasis
- SCORAD, SCORing Atopic Dermatitis
- TC, total cholesterol
- TEWL, transepidermal water loss
- TG, triglyceride
- TLR, toll-like receptor
- Th, T helper
- WAT, white adipose tissue
- dFB, dermal fibroblast
- dWAT, dermal white adipose tissue
- sWAT, subcutaneous white adipose tissue
Collapse
|
105
|
Ahmed SM, Nasr MA, Elshenawy SE, Hussein AE, El-Betar AH, Mohamed RH, El-Badri N. BCG vaccination and the risk of COVID 19: A possible correlation. Virology 2022; 565:73-81. [PMID: 34742127 PMCID: PMC8552046 DOI: 10.1016/j.virol.2021.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/01/2021] [Accepted: 10/17/2021] [Indexed: 01/04/2023]
Abstract
Bacillus Calmette-Guérin (BCG) vaccine is currently used to prevent tuberculosis infection. The vaccine was found to enhance resistance to certain types of infection including positive sense RNA viruses. The current COVID-19 pandemic is caused by positive sense RNA, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A higher mortality rate of COVID-19 patients was reported in countries where BCG vaccination is not routinely administered, when compared to the vaccinated ones. We hypothesized that BCG vaccine may control SARS-CoV2 infection via modulating the monocyte immune response. We analyzed GSE104149 dataset to investigate whether human monocytes of BCG-vaccinated individuals acquire resistance to SARS-CoV-2 infection. Differentially expressed genes obtained from the dataset were used to determine enriched pathways, biological processes, and molecular functions for monocytes post BCG vaccination. Our data show that BCG vaccine promotes a more effective immune response of monocytes against SARS-CoV2, but probably not sufficient to prevent the infection.
Collapse
Affiliation(s)
- Sara M Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 12582, 6th of October City, Giza, Egypt
| | - Mohamed A Nasr
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 12582, 6th of October City, Giza, Egypt
| | - Shimaa E Elshenawy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 12582, 6th of October City, Giza, Egypt
| | - Alaa E Hussein
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 12582, 6th of October City, Giza, Egypt
| | - Ahmed H El-Betar
- Department of Urology, Ahmed Maher Teaching Hospital, Cairo, Egypt
| | | | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 12582, 6th of October City, Giza, Egypt.
| |
Collapse
|
106
|
Louis F, Sowa Y, Kitano S, Matsusaki M. High-throughput drug screening models of mature adipose tissues which replicate the physiology of patients' Body Mass Index (BMI). Bioact Mater 2022; 7:227-241. [PMID: 34466729 PMCID: PMC8379425 DOI: 10.1016/j.bioactmat.2021.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/27/2021] [Accepted: 05/07/2021] [Indexed: 12/27/2022] Open
Abstract
Obesity is a complex and incompletely understood disease, but current drug screening strategies mostly rely on immature in vitro adipose models which cannot recapitulate it properly. To address this issue, we developed a statistically validated high-throughput screening model by seeding human mature adipocytes from patients, encapsulated in physiological collagen microfibers. These drop tissues ensured the maintenance of adipocyte viability and functionality for controlling glucose and fatty acids uptake, as well as glycerol release. As such, patients' BMI and insulin sensitivity displayed a strong inverse correlation: the healthy adipocytes were associated with the highest insulin-induced glucose uptake, while insulin resistance was confirmed in the underweight and severely obese adipocytes. Insulin sensitivity recovery was possible with two type 2 diabetes treatments, rosiglitazone and melatonin. Finally, the addition of blood vasculature to the model seemed to more accurately recapitulate the in vivo physiology, with particular respect to leptin secretion metabolism.
Collapse
Affiliation(s)
- Fiona Louis
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
| | - Yoshihiro Sowa
- Kyoto Prefectural University of Medicine, Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kamigyo-ku Kajii-cho, Kawaramachi-Hirokoji, Kyoto, 602-8566, Japan
- Corresponding author. Kyoto, 602-8566, Kamigyo-ku Kajii-cho, Kawaramachi-Hirokoji, Japan.
| | - Shiro Kitano
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- TOPPAN PRINTING CO., LTD., Technical Research Institute, 4-2-3 Takanodaiminami, Sugito-machi, Saitama, 345-8508, Japan
| | - Michiya Matsusaki
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- Osaka University, Graduate School of Engineering, Department of Applied Chemistry, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- Corresponding author. Osaka, 565-0871, 2-1 Yamadaoka, Suita, Japan.
| |
Collapse
|
107
|
Zaidi H, Aksnes T, Åkra S, Eggesbø HB, Byrkjeland R, Seljeflot I, Opstad TB. Abdominal Adipose Tissue Associates With Adiponectin and TNFα in Middle-Aged Healthy Men. Front Endocrinol (Lausanne) 2022; 13:874977. [PMID: 35872989 PMCID: PMC9301307 DOI: 10.3389/fendo.2022.874977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Adipokines are highly active biopeptides involved in glucose metabolism, insulin regulation and the development and progression of obesity and its associated diseases. It includes, among others, adiponectin, visfatin and tumor necrosis factor alpha (TNFα). The sources of adipokines and their associations with glucometabolic variables are not completely understood. AIM In this cross-sectional study, we aimed to investigate whether gene expression levels in subcutaneous adipose tissue (SAT) of selected adipokines and their corresponding circulating levels associate with the amount of AT in superficial (sSAT), deep (dSAT) and visceral AT (VAT), assessed by computed tomography (CT). Any association with glucometabolic variables were also explored. METHODS In 103 healthy Caucasian men, aged 39.5 years, fasting venous blood and SAT samples from the gluteal region were collected. Ninety-four of the participants underwent CT assessment of the abdominal AT, which was divided into VAT, sSAT and dSAT. Circulating levels of adipokines were measured by ELISA and AT gene-expression by PCR. Insulin sensitivity was determined by glucose clamp, assessing glucose disposal rate (GDR). RESULTS Circulating adiponectin and TNFα gene expression correlated inversely and positively to the amount of AT in all three compartments (r=-0.266 to -0.276, p<0.05 for all) and (r=0.323 - 0.368, p<0.05 for all), respectively, with strongest correlations to the amount in sSAT and dSAT. When dividing AT compartments into quartiles, a tendency was observed towards lower circulating adiponectin and higher TNFα gene expression levels, respectively, with increasing amount of sSAT and dSAT. Circulating adiponectin correlated inversely to insulin, C-peptide and waist circumference (r=-456 to -0.373, p<0.001) and positively to GDR (r=0.356, p<0.001). AT-expressed visfatin correlated inversely to insulin and C-peptide (r=-0.370 and r=-0.404, p<0.001). CONCLUSION Increased amount of AT is associated with lower levels of adiponectin and increased levels of TNFα AT expression.
Collapse
Affiliation(s)
- Hani Zaidi
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- *Correspondence: Hani Zaidi,
| | - Tonje Aksnes
- Section for Interventional Cardiology, Department of Cardiology, Heart-, Lung-, and Vascular-Disease Clinic, Oslo University Hospital, Oslo, Norway
| | - Sissel Åkra
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway
| | - Heidi B. Eggesbø
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Rune Byrkjeland
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trine B. Opstad
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
108
|
He J, Luo J, Yang Q, Yuan F. Eosinophilia in a patient with ankylosing spondylitis treated with prolonged adalimumab only. Arch Med Sci 2022; 18:564-567. [PMID: 35316919 PMCID: PMC8924826 DOI: 10.5114/aoms/146793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Junhong He
- Department of Pharmacy, Ningbo No. 6 Hospital, Ningbo, China
| | - Jing Luo
- Department of Rheumatism and Immunology, Ningbo No. 6 Hospital, Ningbo, China
| | - Qiong Yang
- Department of Pharmacy, Ningbo No. 6 Hospital, Ningbo, China
| | - Fangfang Yuan
- Department of Rheumatism and Immunology, Ningbo No. 6 Hospital, Ningbo, China
| |
Collapse
|
109
|
Lad N, Murphy A, Parenti C, Nelson C, Williams N, Sharpe G, McTernan P. Asthma and obesity: endotoxin another insult to add to injury? Clin Sci (Lond) 2021; 135:2729-2748. [PMID: 34918742 PMCID: PMC8689194 DOI: 10.1042/cs20210790] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022]
Abstract
Low-grade inflammation is often an underlying cause of several chronic diseases such as asthma, obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Defining the mediators of such chronic low-grade inflammation often appears dependent on which disease is being investigated. However, downstream systemic inflammatory cytokine responses in these diseases often overlap, noting there is no doubt more than one factor at play to heighten the inflammatory response. Furthermore, it is increasingly believed that diet and an altered gut microbiota may play an important role in the pathology of such diverse diseases. More specifically, the inflammatory mediator endotoxin, which is a complex lipopolysaccharide (LPS) derived from the outer membrane cell wall of Gram-negative bacteria and is abundant within the gut microbiota, and may play a direct role alongside inhaled allergens in eliciting an inflammatory response in asthma. Endotoxin has immunogenic effects and is sufficiently microscopic to traverse the gut mucosa and enter the systemic circulation to act as a mediator of chronic low-grade inflammation in disease. Whilst the role of endotoxin has been considered in conditions of obesity, cardiovascular disease and T2DM, endotoxin as an inflammatory trigger in asthma is less well understood. This review has sought to examine the current evidence for the role of endotoxin in asthma, and whether the gut microbiota could be a dietary target to improve disease management. This may expand our understanding of endotoxin as a mediator of further low-grade inflammatory diseases, and how endotoxin may represent yet another insult to add to injury.
Collapse
Affiliation(s)
- Nikita Lad
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Alice M. Murphy
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Cristina Parenti
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Carl P. Nelson
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Neil C. Williams
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Graham R. Sharpe
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Philip G. McTernan
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| |
Collapse
|
110
|
Relationship between Physical Activity and the Metabolic, Inflammatory Axis in Pregnant Participants. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182413160. [PMID: 34948770 PMCID: PMC8701987 DOI: 10.3390/ijerph182413160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 01/26/2023]
Abstract
Physical activity (PA) during pregnancy is beneficial for mother and child. Little is known regarding the effects of PA on specific adipokines/myokines and their impact during pregnancy. This study investigates the correlation between PA during late pregnancy, body composition, and maternal levels of leptin, IL-6, and TNF-α at delivery. In a cross-sectional study of 91 pregnant participants (mean age 33.9 ± 4.6 years) without gestational diabetes mellitus or preeclampsia, anthropometric data and blood samples were taken at delivery. PA during the third trimester was measured via the Pregnancy Physical Activity Questionnaire. Activities were ranked by intensity: sedentary (<1.5 metabolic equivalent (METs)), light (1.5–3.0 METs), moderate (3.0–6.0 METs), and vigorous activity (>6.0 METs). Leptin at delivery correlated positively with body composition and negatively with light PA intensity. Sedentary behaviour showed a positive correlation with IL-6 levels at delivery. Moderate activity during the last trimester, sedentary activity levels, and body composition had the greatest influence on maternal IL-6 at delivery. Completed weeks of pregnancy, moderate and light PA, and sedentary activity had the greatest influence on maternal TNF-α at delivery. PA during late pregnancy potentially affects circulating (adipo-)/myokines. Further studies are needed to examine causal relationships and the impact on maternal and new-born health.
Collapse
|
111
|
Pan F, Tian J, Cicuttini F, Jones G. Prospective Association Between Inflammatory Markers and Knee Cartilage Volume Loss and Pain Trajectory. Pain Ther 2021; 11:107-119. [PMID: 34837639 PMCID: PMC8861228 DOI: 10.1007/s40122-021-00341-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/10/2021] [Indexed: 01/15/2023] Open
Abstract
Introduction Inflammation has been suggested to be involved in the pathogenesis of osteoarthritis and pain. We sought to explore the associations between inflammatory serum markers and magnetic resonance imaging-defined long-term structural change and pain trajectory. Methods A total of 169 randomly selected participants (mean age 63 years; 47% female) from a prospective cohort study were included in this study. Circulating levels of interleukin 6 (IL-6), tumour necrosis factor alpha (TNF-α) and high-sensitivity C-reactive protein (CRP) were measured at baseline. A knee MRI scan was performed to measure cartilage volume (CV) and bone marrow lesions (BMLs) at baseline and at 10.7 years. Knee pain at four visits was measured by the WOMAC pain questionnaire, and pain trajectories were identified using group-based trajectory modelling. Linear, log-binomial and multi-nominal logistic regression were used for the analyses. Results IL-6 was associated with lateral but not medial tibial CV loss (β = − 0.25% per annum, per standard deviation [SD] log pg/ml; P < 0.05) in the multivariate analysis. IL-6 was also associated with a ‘Moderate pain’ trajectory (relative risk ratio 1.93 per SD log pg/ml; 95% confidence interval 1.02–3.65) relative to the ‘Minimal pain’ trajectory group. There was no significant association of TNF-α and CRP with CV loss and pain trajectory groups with the exception of a beneficial relationship between CRP and medial tibial CV loss (β = 0.20% per annum, per SD log mg/l). No association between inflammatory markers and change in BML size was observed. Conclusions IL-6 was independently associated with compartment-specific CV loss and worse pain trajectory, but the other markers studied were not, suggesting that components of inflammation are implicated in the pathogenesis of cartilage loss and developing a worse pain course. Supplementary Information The online version contains supplementary material available at 10.1007/s40122-021-00341-1.
Collapse
Affiliation(s)
- Feng Pan
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7000, Australia.
| | - Jing Tian
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7000, Australia
| | - Flavia Cicuttini
- Department of Epidemiology and Preventive Medicine, Monash University Medical School, Commercial Road, Melbourne, VIC, 3181, Australia
| | - Graeme Jones
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7000, Australia
| |
Collapse
|
112
|
Atchan Nwakiban AP, Passarelli A, Da Dalt L, Olivieri C, Demirci TN, Piazza S, Sangiovanni E, Carpentier-Maguire E, Martinelli G, Shivashankara ST, Manjappara UV, Tchamgoue AD, Agbor GA, Kuiate JR, Daglia M, Dell’Agli M, Magni P. Cameroonian Spice Extracts Modulate Molecular Mechanisms Relevant to Cardiometabolic Diseases in SW 872 Human Liposarcoma Cells. Nutrients 2021; 13:nu13124271. [PMID: 34959824 PMCID: PMC8706885 DOI: 10.3390/nu13124271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/27/2022] Open
Abstract
The molecular pathophysiology of cardiometabolic diseases is known to be influenced by dysfunctional ectopic adipose tissue. In addition to lifestyle improvements, these conditions may be managed by novel nutraceutical products. This study evaluatedthe effects of 11 Cameroonian medicinal spice extracts on triglyceride accumulation, glucose uptake, reactive oxygen species (ROS) production and interleukin secretion in SW 872 human adipocytes after differentiation with 100 µM oleic acid. Triglyceride content was significantly reduced by all spice extracts. Glucose uptake was significantly increased by Tetrapleura tetraptera, Aframomum melegueta and Zanthoxylum leprieurii. Moreover, Xylopia parviflora, Echinops giganteus and Dichrostachys glomerata significantly reduced the production of ROS. Concerning pro-inflammatory cytokine secretion, we observed that Tetrapleura tetraptera, Echinops giganteus, Dichrostachys glomerata and Aframomum melegueta reduced IL-6 secretion. In addition, Xylopia parviflora, Monodora myristica, Zanthoxylum leprieurii, and Xylopia aethiopica reduced IL-8 secretion, while Dichrostachys glomerata and Aframomum citratum increased it. These findings highlight some interesting properties of these Cameroonian spice extracts in the modulation of cellular parameters relevant to cardiometabolic diseases, which may be further exploited, aiming to develop novel treatment options for these conditions based on nutraceutical products.
Collapse
Affiliation(s)
| | - Anna Passarelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
| | - Chiara Olivieri
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
| | - Tugba Nur Demirci
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
| | - Stefano Piazza
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
| | - Enrico Sangiovanni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
| | | | - Giulia Martinelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
| | - Shilpa Talkad Shivashankara
- Department of Lipid Science, CSIR-Central Food Technological Research Institute (CFTRI), Mysore 570 020, India; (S.T.S.); (U.V.M.)
| | - Uma Venkateswaran Manjappara
- Department of Lipid Science, CSIR-Central Food Technological Research Institute (CFTRI), Mysore 570 020, India; (S.T.S.); (U.V.M.)
| | - Armelle Deutou Tchamgoue
- Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé 4123, Cameroon; (A.D.T.); (G.A.A.)
| | - Gabriel Agbor Agbor
- Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé 4123, Cameroon; (A.D.T.); (G.A.A.)
| | - Jules-Roger Kuiate
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang P.O. Box 96, Cameroon; (A.P.A.N.); (J.-R.K.)
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Mario Dell’Agli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
- Correspondence: (M.D.); (P.M.); Tel.: +39-0250318398 (M.D.); +39-0250318229 (P.M.)
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.P.); (L.D.D.); (C.O.); (T.N.D.); (S.P.); (E.S.); (G.M.)
- IRCCS MultiMedica, Sesto San Giovanni, 20099 Milan, Italy
- Correspondence: (M.D.); (P.M.); Tel.: +39-0250318398 (M.D.); +39-0250318229 (P.M.)
| |
Collapse
|
113
|
Liu S, Wang L, Ling D, Valencak TG, You W, Shan T. Potential key factors involved in regulating adipocyte dedifferentiation. J Cell Physiol 2021; 237:1639-1647. [PMID: 34796916 DOI: 10.1002/jcp.30637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/26/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
Adipocytes are the key constituents of adipose tissue, and their de-differentiation process has been widely observed in physiological and pathological conditions. For obese people, the promotion of adipocyte de-differentiation or maintenance of an undifferentiated state of adipocytes may help to improve their metabolic condition. Thus, understanding the regulatory mechanisms of adipocyte de-differentiation is necessary for treating metabolic diseases. Attractively, in addition to intracellular signals regulating adipocyte de-differentiation, external factors such as temperature and pressure also affect adipocyte de-differentiation. In this review, we summarize the recent progress in the field and discuss the regulatory roles and mechanisms of involved endogenous and exogenous factors during the process of de-differentiation.
Collapse
Affiliation(s)
- Shiqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Defeng Ling
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Teresa G Valencak
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
114
|
Aghamajidi A, Gorgani M, Shahba F, Shafaghat Z, Mojtabavi N. The potential targets in immunotherapy of atherosclerosis. Int Rev Immunol 2021; 42:199-216. [PMID: 34779341 DOI: 10.1080/08830185.2021.1988591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Cardiovascular disease is the most common cause of death, which has the highest mortality rate worldwide. Although a diverse range of inflammatory diseases can affect the cardiovascular system, however, heart failure and stroke occur due to atherosclerosis. Atherosclerosis is a chronic autoinflammatory disease of small to large vessels in which different immune mediators are involved in lipid plaque formation and inflammatory vascular remodeling process. A better understanding of the pathophysiology of atherosclerosis may lead to uncovering immunomodulatory therapies. Despite present diagnostic and therapeutic methods, the lack of immunotherapy in the prevention and treatment of atherosclerosis is perceptible. In this review, we will discuss the promising immunological-based therapeutics and novel preventive approaches for atherosclerosis. This study could provide new insights into a better perception of targeted therapeutic pathways and biological therapies.
Collapse
Affiliation(s)
- Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Melika Gorgani
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.,Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Faezeh Shahba
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Shafaghat
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Mojtabavi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
115
|
Peres Valgas da Silva C, Calmasini F, Alexandre EC, Raposo HF, Delbin MA, Monica FZ, Zanesco A. The effects of mirabegron on obesity-induced inflammation and insulin resistance are associated with brown adipose tissue activation but not beiging in the subcutaneous white adipose tissue. Clin Exp Pharmacol Physiol 2021; 48:1477-1487. [PMID: 34343353 DOI: 10.1111/1440-1681.13566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 01/22/2023]
Abstract
Mirabegron is a selective β₃-adrenergic receptors agonist, which has been recently shown to improve metabolic health in rodents and humans. In this study, we investigated the effects of 2-week mirabegron treatment on the metabolic parameters of mice with a diet-induced obesity (DIO). C57BL/6JUnib mice were divided into control (CTR) and obese (OB) groups treated with vehicle, and an OB group treated with mirabegron (OB + MIRA). The obese groups were fed a high-fat diet for 12 weeks. Mirabegron (10 mg/kg/day) was administrated orally by gavage from weeks 10-12. After 2 weeks of mirabegron treatment, the energy expenditure was assessed with indirect calorimetry. Blood glucose, insulin, glycerol, free fatty acids (FFA), thiobarbituric acid reactive substance (TBAR), and tumour necrosis factor (TNF)-α levels were also assessed, and the HOMA index was determined. Liver tissue, brown adipose tissue (BAT), and inguinal white adipose tissue (iWAT) samples were collected for histological examination. The protein expressions of uncoupling protein 1 (UCP1) and mitochondrial transcription factor A (TFAM) were assessed using western blotting of the BAT and iWAT samples. In this study, mirabegron increased the energy expenditure and decreased adiposity in OB + MIRA. Increased UCP1 expression in BAT without changes in iWAT was also found. Mirabegron decreased circulating levels of FFA, glycerol, insulin, TNF-α, TBARS and HOMA index. DIO significantly increased the lipid deposits in the liver and BAT, but mirabegron partially reversed this change. Our findings indicate that treatment with mirabegron decreased inflammation and improved metabolism in obese mice. This effect was associated with increased BAT-mediated energy expenditure, but not iWAT beiging, which suggests that mirabegron might be useful for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Carmem Peres Valgas da Silva
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Physical Education, Institute of Biosciences, São Paulo State University (UNESP), Rio Claro, Brazil
| | - Fabiano Calmasini
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Eduardo Costa Alexandre
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Helena Fonseca Raposo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Maria Andreia Delbin
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Fabiola Zakia Monica
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Angelina Zanesco
- Department of Physical Education, Institute of Biosciences, São Paulo State University (UNESP), Rio Claro, Brazil
- Medical School, Graduate Program in Environmental Health, Metropolitan University of Santos, Santos, Brazil
| |
Collapse
|
116
|
Yoon YS, Liu W, Van de Velde S, Matsumura S, Wiater E, Huang L, Montminy M. Activation of the adipocyte CREB/CRTC pathway in obesity. Commun Biol 2021; 4:1214. [PMID: 34686752 PMCID: PMC8536733 DOI: 10.1038/s42003-021-02735-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 09/21/2021] [Indexed: 11/09/2022] Open
Abstract
Obesity is a major risk factor for the development of type II diabetes. Increases in adipose tissue mass trigger insulin resistance via the release of pro-inflammatory cytokines from adipocytes and macrophages. CREB and the CRTC coactivators have been found to promote insulin resistance in obesity, although the mechanism is unclear. Here we show that high fat diet feeding activates the CREB/CRTC pathway in adipocytes by decreasing the expression of SIK2, a Ser/Thr kinase that phosphorylates and inhibits CRTCs. SIK2 levels are regulated by the adipogenic factor C/EBPα, whose expression is reduced in obesity. Exposure to PPARγ agonist rescues C/EBPα expression and restores SIK2 levels. CRTC2/3 promote insulin resistance via induction of the chemokines CXCL1/2. Knockout of CRTC2/3 in adipocytes reduces CXCL1/2 expression and improves insulin sensitivity. As administration of CXCL1/2 reverses salutary effects of CRTC2/3 depletion, our results demonstrate the importance of the CREB/CRTC pathway in modulating adipose tissue function.
Collapse
Affiliation(s)
- Young-Sil Yoon
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Weiyi Liu
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Sam Van de Velde
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Shigenobu Matsumura
- Department of Clinical Nutrition, Osaka Prefecture University, Habikino, Habikino City, Osaka, Japan
| | - Ezra Wiater
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Marc Montminy
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
117
|
Quarta S, Scoditti E, Carluccio MA, Calabriso N, Santarpino G, Damiano F, Siculella L, Wabitsch M, Verri T, Favari C, Del Rio D, Mena P, De Caterina R, Massaro M. Coffee Bioactive N-Methylpyridinium Attenuates Tumor Necrosis Factor (TNF)-α-Mediated Insulin Resistance and Inflammation in Human Adipocytes. Biomolecules 2021; 11:biom11101545. [PMID: 34680177 PMCID: PMC8534185 DOI: 10.3390/biom11101545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 12/05/2022] Open
Abstract
Although coffee consumption has been historically associated with negative health outcomes, recent evidence suggests a lower risk of metabolic syndrome, obesity and diabetes among regular coffee drinkers. Among the plethora of minor organic compounds assessed as potential mediators of coffee health benefits, trigonelline and its pyrolysis product N-methylpyridinium (NMP) were preliminary shown to promote glucose uptake and exert anti-adipogenic properties. Against this background, we aimed at characterizing the effects of trigonelline and NMP in inflamed and dysfunctional human adipocytes. Human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes were treated with NMP or, for comparison, trigonelline, for 5 h before stimulation with tumor necrosis factor (TNF)-α. NMP at concentrations as low as 1 µmol/L reduced the stimulated expression of several pro-inflammatory mediators, including C-C Motif chemokine ligand (CCL)-2, C-X-C Motif chemokine ligand (CXCL)-10, and intercellular adhesion Molecule (ICAM)-1, but left the induction of prostaglandin G/H synthase (PTGS)2, interleukin (IL)-1β, and colony stimulating factor (CSF)1 unaffected. Furthermore, NMP restored the downregulated expression of adiponectin (ADIPOQ). These effects were functionally associated with downregulation of the adhesion of monocytes to inflamed adipocytes. Under the same conditions, NMP also reversed the TNF-α-mediated suppression of insulin-stimulated Ser473 Akt phosphorylation and attenuated the induction of TNF-α-stimulated lipolysis restoring cell fat content. In an attempt to preliminarily explore the underlying mechanisms of its action, we show that NMP restores the expression of the master regulator of adipocyte differentiation peroxisome proliferator-activated receptor (PPAR)γ and downregulates activation of the pro-inflammatory mitogen-activated protein jun N-terminal kinase (JNK). In conclusion, NMP reduces adipose dysfunction in pro-inflammatory activated adipocytes. These data suggest that bioactive NMP in coffee may improve the inflammatory and dysmetabolic milieu associated with obesity.
Collapse
Affiliation(s)
- Stefano Quarta
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy; (S.Q.); (F.D.); (L.S.); (T.V.)
| | - Egeria Scoditti
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (E.S.); (M.A.C.); (N.C.)
| | - Maria Annunziata Carluccio
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (E.S.); (M.A.C.); (N.C.)
| | - Nadia Calabriso
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (E.S.); (M.A.C.); (N.C.)
| | - Giuseppe Santarpino
- Cardiovascular Center, Paracelsus Medical University, 90471 Nuremberg, Germany;
- GVM Care & Research, Città di Lecce Hospital, 73100 Lecce, Italy
- Cardiac Surgery Unit, Department of Experimental and Clinical Medicine, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Fabrizio Damiano
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy; (S.Q.); (F.D.); (L.S.); (T.V.)
| | - Luisa Siculella
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy; (S.Q.); (F.D.); (L.S.); (T.V.)
| | - Martin Wabitsch
- Division of Pediatric Endocrinology, Diabetes and Obesity, Department of Pediatrics and Adolescent Medicine, University of Ulm, 89075 Ulm, Germany;
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy; (S.Q.); (F.D.); (L.S.); (T.V.)
| | - Claudia Favari
- Human Nutrition Unit, Department of Food and Drugs, University of Parma, 43125 Parma, Italy; (C.F.); (D.D.R.); (P.M.)
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food and Drugs, University of Parma, 43125 Parma, Italy; (C.F.); (D.D.R.); (P.M.)
| | - Pedro Mena
- Human Nutrition Unit, Department of Food and Drugs, University of Parma, 43125 Parma, Italy; (C.F.); (D.D.R.); (P.M.)
| | - Raffaele De Caterina
- Cardiology Division, Pisa University Hospital, 56126 Pisa, Italy
- Fondazione Villa Serena per la Ricerca, Città Sant’Angelo, 65013 Pescara, Italy
- Correspondence: (R.D.C.); (M.M.); Tel.: +39-050-996-751 (R.D.C.); +39-083-229-8860 (M.M.)
| | - Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (E.S.); (M.A.C.); (N.C.)
- Correspondence: (R.D.C.); (M.M.); Tel.: +39-050-996-751 (R.D.C.); +39-083-229-8860 (M.M.)
| |
Collapse
|
118
|
Yoshida H. [Preventive and Ameliorating Effects of Food Factors on Obesity-related Diseases by Regulating Inflammation]. YAKUGAKU ZASSHI 2021; 141:1161-1171. [PMID: 34602513 DOI: 10.1248/yakushi.21-00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Japan is currently a super-aging society, and lifestyle-related diseases that increase in incidence with age and the related rise in national medical expenses are major social problems. Preventive medicine and self-medication are becoming more important. Recently, various in vitro and in vivo studies have shown that food-derived natural compounds may contribute to the prevention and treatment of obesity-related diseases, such as diabetes mellitus. This report reviews our previous studies on the usefulness of the citrus flavonoid naringenin for obesity-related diseases. We showed that naringenin exerts an anti-diabetic effect by regulating inflammation pathways involving adipocytes and adipose tissue, and also showed an interaction between naringenin and anti-diabetic drugs. Because natural compounds are generally inexpensive and safe, they have the advantage of being easily applied to clinical applications. However, more detailed studies, such as clinical trials in humans, are required. Further research and scientific evidence will be required for the proper use of food factors in disease prevention and treatment.
Collapse
Affiliation(s)
- Hiroki Yoshida
- Department of Biochemistry, Graduate School of Clinical Pharmacy, Kyushu University of Health and Welfare
| |
Collapse
|
119
|
Abstract
Tumour necrosis factor (TNF) is a classical, pleiotropic pro-inflammatory cytokine. It is also the first 'adipokine' described to be produced from adipose tissue, regulated in obesity and proposed to contribute to obesity-associated metabolic disease. In this review, we provide an overview of TNF in the context of metabolic inflammation or metaflammation, its discovery as a metabolic messenger, its sites and mechanisms of action and some critical considerations for future research. Although we focus on TNF and the studies that elucidated its immunometabolic actions, we highlight a conceptual framework, generated by these studies, that is equally applicable to the complex network of pro-inflammatory signals, their biological activity and their integration with metabolic regulation, and to the field of immunometabolism more broadly.
Collapse
Affiliation(s)
- Jaswinder K Sethi
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK.
- Institute for Life Sciences, University of Southampton, Southampton, UK.
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Harvard-MIT Broad Institute, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
- The Joslin Diabetes Center, Boston, MA, USA.
| |
Collapse
|
120
|
Role of Inflammatory Cytokines, Growth Factors and Adipokines in Adipogenesis and Insulin Resistance. Inflammation 2021; 45:31-44. [PMID: 34536157 PMCID: PMC8449520 DOI: 10.1007/s10753-021-01559-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 01/06/2023]
Abstract
Obesity, manifested by increased adiposity, represents a main cause of morbidity in the developed countries, causing increased risk of insulin resistance and type 2 diabetes mellitus. Recruitment of macrophages and activation of innate immunity represent the initial insult, which can be further exacerbated through secretion of chemokines and adipocytokines from activated macrophages and other cells within the adipose tissue. These events can impact adipogenesis, causing dysfunction of the adipose tissue and increased risk of insulin resistance. Various factors mediate adiposity and related insulin resistance including inflammatory and non-inflammatory factors such as pro and anti-inflammatory cytokines, adipokines and growth factors. In this review we will discuss the role of these factors in adipogenesis and development of insulin resistance and type 2 diabetes mellitus in the context of obesity. Understanding the molecular mechanisms that mediate adipogenesis and insulin resistance could help the development of novel therapeutic strategies for individuals at higher risk of insulin resistance and type 2 diabetes mellitus.
Collapse
|
121
|
Velikova TV, Kabakchieva PP, Assyov YS, Georgiev TА. Targeting Inflammatory Cytokines to Improve Type 2 Diabetes Control. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7297419. [PMID: 34557550 PMCID: PMC8455209 DOI: 10.1155/2021/7297419] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is one of the most common chronic metabolic disorders in adulthood worldwide, whose pathophysiology includes an abnormal immune response accompanied by cytokine dysregulation and inflammation. As the T2D-related inflammation and its progression were associated with the balance between pro and anti-inflammatory cytokines, anticytokine treatments might represent an additional therapeutic option for T2D patients. This review focuses on existing evidence for antihyperglycemic properties of disease-modifying antirheumatic drugs (DMARDs) and anticytokine agents (anti-TNF-α, anti-interleukin-(IL-) 6, -IL-1, -IL-17, -IL-23, etc.). Emphasis is placed on their molecular mechanisms and on the biological rationale for clinical use. Finally, we briefly summarize the results from experimental model studies and promising clinical trials about the potential of anticytokine therapies in T2D, discussing the effects of these drugs on systemic and islet inflammation, beta-cell function, insulin secretion, and insulin sensitivity.
Collapse
Affiliation(s)
- Tsvetelina V. Velikova
- Department of Clinical Immunology, University Hospital “Lozenetz”, Sofia University “St. Kliment Ohridski”, Sofia 1407, Bulgaria
| | - Plamena P. Kabakchieva
- Clinic of Endocrinology, University Hospital “Alexandrovska, ” Department of Internal Medicine, Medical Faculty, Medical University of Sofia, Sofia 1431, Bulgaria
- Clinic of Internal Medicine, Naval Hospital-Varna, Military Medical Academy, Varna 9010, Bulgaria
| | - Yavor S. Assyov
- Clinic of Endocrinology, University Hospital “Alexandrovska, ” Department of Internal Medicine, Medical Faculty, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Tsvetoslav А. Georgiev
- Clinic of Rheumatology, University Hospital “St. Marina, ” First Department of Internal Medicine, Medical Faculty, Medical University-Varna, Varna 9010, Bulgaria
| |
Collapse
|
122
|
Psoriasis and Cardiovascular Disease: Novel Mechanisms and Evolving Therapeutics. Curr Atheroscler Rep 2021; 23:67. [PMID: 34468875 PMCID: PMC9744099 DOI: 10.1007/s11883-021-00963-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Psoriasis is a chronic inflammatory skin condition that is associated with increased cardiovascular risk compared to those without psoriasis. This review will cover emerging mechanisms of cardiovascular risk, key pathways targeted with biologic therapies, and the current evidence on therapies to modulate this risk in patients with psoriasis. RECENT FINDINGS Recent scientific work has highlighted mechanisms that contribute to this enhanced risk, including the role of vascular endothelial dysfunction, platelet activation, dyslipidemia, and increased cardiometabolic comorbidities. Newer biologic and targeted synthetic therapies have transformed psoriasis treatment with high rates of clinical remission and durable skin disease control now possible. Epidemiological evidence suggests that many of these therapies may lower cardiovascular risk in psoriasis, although prospective interventional data is lacking (or mixed). Recently, caution has also been raised that some treatments may negatively affect cardiovascular risk. Overall, the current data suggests a positive or neutral ability to reduce cardiovascular risk for TNF, IL-17A, and IL-12/23p40 inhibitors, but current evidence remains conflicting for anti-IL-23/p19 and JAK inhibitors. More studies that include prospective cohorts, larger number of patients, treatment duration, and validated surrogate outcomes are needed to better evaluate the role of biologic therapies on cardiovascular risk in psoriasis.
Collapse
|
123
|
Husson H, Bukanov NO, Moreno S, Smith MM, Richards B, Zhu C, Picariello T, Park H, Wang B, Natoli TA, Smith LA, Zanotti S, Russo RJ, Madden SL, Klinger KW, Modur V, Ibraghimov-Beskrovnaya O. Correction of cilia structure and function alleviates multi-organ pathology in Bardet-Biedl syndrome mice. Hum Mol Genet 2021; 29:2508-2522. [PMID: 32620959 PMCID: PMC7471507 DOI: 10.1093/hmg/ddaa138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/22/2020] [Accepted: 07/01/2020] [Indexed: 12/21/2022] Open
Abstract
Bardet–Biedl syndrome (BBS) is a pleiotropic autosomal recessive ciliopathy affecting multiple organs. The development of potential disease-modifying therapy for BBS will require concurrent targeting of multi-systemic manifestations. Here, we show for the first time that monosialodihexosylganglioside accumulates in Bbs2−/− cilia, indicating impairment of glycosphingolipid (GSL) metabolism in BBS. Consequently, we tested whether BBS pathology in Bbs2−/− mice can be reversed by targeting the underlying ciliary defect via reduction of GSL metabolism. Inhibition of GSL synthesis with the glucosylceramide synthase inhibitor Genz-667161 decreases the obesity, liver disease, retinal degeneration and olfaction defect in Bbs2−/− mice. These effects are secondary to preservation of ciliary structure and signaling, and stimulation of cellular differentiation. In conclusion, reduction of GSL metabolism resolves the multi-organ pathology of Bbs2−/− mice by directly preserving ciliary structure and function towards a normal phenotype. Since this approach does not rely on the correction of the underlying genetic mutation, it might translate successfully as a treatment for other ciliopathies.
Collapse
Affiliation(s)
- Hervé Husson
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Nikolay O Bukanov
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Sarah Moreno
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Mandy M Smith
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | | | - Cheng Zhu
- Translational Sciences, Sanofi, Framingham, MA 01701, USA
| | - Tyler Picariello
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Hyejung Park
- Pre-Development Sciences, Sanofi, Waltham, MA 02451, USA
| | - Bing Wang
- Pre-Development Sciences, Sanofi, Waltham, MA 02451, USA
| | - Thomas A Natoli
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Laurie A Smith
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Stefano Zanotti
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Ryan J Russo
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | | | | | - Vijay Modur
- Rare Diseases Development, Sanofi, Cambridge, MA 02142, USA
| | | |
Collapse
|
124
|
Jo S, Fang S. Therapeutic Strategies for Diabetes: Immune Modulation in Pancreatic β Cells. Front Endocrinol (Lausanne) 2021; 12:716692. [PMID: 34484126 PMCID: PMC8415970 DOI: 10.3389/fendo.2021.716692] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022] Open
Abstract
Increased incidence of type I and type II diabetes has been prevailed worldwide. Though the pathogenesis of molecular mechanisms remains still unclear, there are solid evidence that disturbed immune homeostasis leads to pancreatic β cell failure. Currently, autoimmunity and uncontrolled inflammatory signaling pathways have been considered the major factors in the pathogenesis of diabetes. Many components of immune system have been reported to implicate pancreatic β cell failure, including helper T cells, cytotoxic T cells, regulatory T cells and gut microbiota. Immune modulation of those components using small molecules and antibodies, and fecal microbiota transplantation are undergoing in many clinical trials for the treatment of type I and type II diabetes. In this review we will discuss the basis of molecular pathogenesis focusing on the disturbed immune homeostasis in type I and type II diabetes, leading to pancreatic β cell destruction. Finally, we will introduce current therapeutic strategies and clinical trials by modulation of immune system for the treatment of type I and type II diabetes patients.
Collapse
Affiliation(s)
- Sugyeong Jo
- Department of Medical Science, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sungsoon Fang
- Department of Medical Science, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
125
|
Michicotl-Meneses MM, Thompson-Bonilla MDR, Reyes-López CA, García-Pérez BE, López-Tenorio II, Ordaz-Pichardo C, Jaramillo-Flores ME. Inflammation Markers in Adipose Tissue and Cardiovascular Risk Reduction by Pomegranate Juice in Obesity Induced by a Hypercaloric Diet in Wistar Rats. Nutrients 2021; 13:2577. [PMID: 34444736 PMCID: PMC8402035 DOI: 10.3390/nu13082577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 02/07/2023] Open
Abstract
Pomegranate juice (Punica granatum) has been used since ancient times in traditional medicine (Unani Medicine, Ayurveda); its main compounds are anthocyanins and ellagic acid, which have anti-inflammatory, antioxidant, hepatoprotective, and cardiovascular health effects. The objective was to evaluate the effect of pomegranate juice on inflammation, blood pressure, and vascular and physiological markers associated with obesity induced by a high-fat diet in a murine model. The results show that pomegranate juice reduces the concentration of low-density lipoprotein cholesterol (cLDL) 39% and increases the concentration of high-density lipoprotein cholesterol (cHDL) by 27%, leading to a 12%-18% decrease in the risk of cardiovascular diseases (CVD). In addition to reducing blood pressure by 24%, it also had an antiatherogenic effect by decreasing sE-selectin levels by 42%. On the other hand, the juice significantly increased adiponectin levels in adipose tissue, decreased levels of inflammation markers (tumor necrosis factor-α (TNF-α), plasminogen activator inhibitor-1 (PAI-1), interleukin-17A (IL-17A), interleukin-6 (IL-6), interleukin-1β (IL-1β)), and inhibited the monocyte chemoattractant protein-1 (MCP-1). Pomegranate juice requires clinical studies to prove its immunoregulatory and therapeutic effects on cardiovascular and atherogenic risks.
Collapse
Affiliation(s)
- Maria Monica Michicotl-Meneses
- Departamento de Ingeniería Bioquímica, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Mexico City 07738, Mexico; (M.M.M.-M.); (I.I.L.-T.)
| | - María del Rocío Thompson-Bonilla
- Laboratorio de Medicina Genómica, Investigación Biomédica y Traslacional, ISSSTE, Hospital Regional “1° de Octubre”, Mexico City 07760, Mexico;
| | - César A. Reyes-López
- Laboratorio de Bioquímica Estructural, Instituto Politécnico Nacional, Escuela Nacional de Medicina y Homeopatía, Mexico City 07320, Mexico;
| | - Blanca Estela García-Pérez
- Laboratorio de Microbiología General, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Mexico City 11340, Mexico;
| | - Itzel I. López-Tenorio
- Departamento de Ingeniería Bioquímica, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Mexico City 07738, Mexico; (M.M.M.-M.); (I.I.L.-T.)
| | - Cynthia Ordaz-Pichardo
- Laboratorio de Biología Celular y Productos Naturales, Instituto Politécnico Nacional, Escuela Nacional de Medicina y Homeopatía, Mexico City 07320, Mexico;
| | - María Eugenia Jaramillo-Flores
- Departamento de Ingeniería Bioquímica, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Mexico City 07738, Mexico; (M.M.M.-M.); (I.I.L.-T.)
| |
Collapse
|
126
|
Zhang L, Zhang XT, Jin P, Zhao H, Liu X, Sheng Q. Effects of oral administration of Spirulina platensis and probiotics on serum immunity indexes, colonic immune factors, fecal odor, and fecal flora in mice. Anim Sci J 2021; 92:e13593. [PMID: 34289202 DOI: 10.1111/asj.13593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/01/2022]
Abstract
To evaluate the effects of Spirulina platensis and probiotics on growth, immunity indexes, fecal flora, and fecal odor in mice, 40 mice were randomly allotted to four groups, and each was administrated with nothing, S. platensis, probiotics, or both for 28 days, respectively. Then, many indexes were measured. The results showed that S. platensis was more effective (P < 0.001) than probiotics in improving mice's feed conversion ration (FCR). In immunity, probiotics administration increased (P < 0.042) serum IgE, IgM, IFN-γ, colonic AHR, TLR4, and NF-κB protein expression and decreased (P < 0.039) serum IL-1α, IL-21, IL-22, and colonic ARNT gene expression. However, the S. platensis showed weaker effect, which increased (P < 0.025) the serum IgE, IgM, TNF-α, and the colonic AHR and NF-κB protein expression, and decreased (P < 0.01) serum IL-21. Probiotics consumption decreased the fecal odor by decreasing (P < 0.02) fecal Escherichia coli, indole-3-acetic acid (IAA), and skatole contents, and the S. platensis decreased (P = 0.04) the IAA. These results indicated that oral administration of probiotics, S. platensis, or both of them in mice probably benefited body's immunity and reduced fecal odor. However, their mechanisms were still unclear and need further study.
Collapse
Affiliation(s)
- Lingyan Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Jinan, China
| | - Xing Tao Zhang
- The Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, China
| | - Pingting Jin
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Jinan, China
| | - Hongbo Zhao
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Jinan, China
| | - Xue Liu
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qingkai Sheng
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Jinan, China.,College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
127
|
Dilworth L, Facey A, Omoruyi F. Diabetes Mellitus and Its Metabolic Complications: The Role of Adipose Tissues. Int J Mol Sci 2021; 22:ijms22147644. [PMID: 34299261 PMCID: PMC8305176 DOI: 10.3390/ijms22147644] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Many approaches have been used in the effective management of type 2 diabetes mellitus. A recent paradigm shift has focused on the role of adipose tissues in the development and treatment of the disease. Brown adipose tissues (BAT) and white adipose tissues (WAT) are the two main types of adipose tissues with beige subsets more recently identified. They play key roles in communication and insulin sensitivity. However, WAT has been shown to contribute significantly to endocrine function. WAT produces hormones and cytokines, collectively called adipocytokines, such as leptin and adiponectin. These adipocytokines have been proven to vary in conditions, such as metabolic dysfunction, type 2 diabetes, or inflammation. The regulation of fat storage, energy metabolism, satiety, and insulin release are all features of adipose tissues. As such, they are indicators that may provide insights on the development of metabolic dysfunction or type 2 diabetes and can be considered routes for therapeutic considerations. The essential roles of adipocytokines vis-a-vis satiety, appetite, regulation of fat storage and energy, glucose tolerance, and insulin release, solidifies adipose tissue role in the development and pathogenesis of diabetes mellitus and the complications associated with the disease.
Collapse
Affiliation(s)
- Lowell Dilworth
- Department of Pathology, Mona Campus, University of the West Indies, Kingston 7, Jamaica;
| | - Aldeam Facey
- Mona Academy of Sport, Mona Campus, University of the West Indies, Kingston 7, Jamaica;
| | - Felix Omoruyi
- Department of Life Sciences, Texas A&M University, Corpus Christi, TX 78412, USA
- Correspondence:
| |
Collapse
|
128
|
Aladhami AK, Unger CA, Ennis SL, Altomare D, Ji H, Hope MC, Velázquez KT, Enos RT. Macrophage tumor necrosis factor-alpha deletion does not protect against obesity-associated metabolic dysfunction. FASEB J 2021; 35:e21665. [PMID: 34131955 PMCID: PMC8716064 DOI: 10.1096/fj.202100543rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 01/11/2023]
Abstract
The pro-inflammatory cytokine, tumor necrosis factor-alpha (TNF-α), has been suggested to be a key factor in the induction of obesity-associated metabolic dysfunction. However, the role that macrophage-derived TNF-α has on regulating metabolic perturbations in obesity is not completely understood. Therefore, we utilized the TNF-αFlox/Flox(F/F) , LyzMcre± mouse model to determine the impact that macrophage TNF-α deletion has on the development of high-fat diet (HFD)-induced obesity. At 10 weeks of age, male littermates were randomly assigned to 1 of 4 groups: TNF-αF/F low-fat diet (TNF-αF/F LFD), TNF-αF/F,LyzMCre LFD, TNF-αF/F HFD, or TNF-αF/F,LyzMCre HFD (n = 16-28/group) and were fed their respective diets for 18 weeks. Body weight was assessed throughout the course of the experiment. Body composition, hepatic lipid accumulation, and metabolic outcomes were also examined. A microarray gene expression experiment was performed from RNA isolated from epididymal adipose tissue of the HFD-fed groups (n = 10/group) and results were verified via qRT-PCR for all groups. Macrophage-derived TNF-α deletion significantly reduced adipose tissue TNF-α gene expression and circulating TNF-α and downregulated genes linked to the toll-like receptor (TLR) and NFκB signaling pathways. However, macrophage TNF-α deletion had no effect on hindering the development of obesity, hepatic lipid accumulation, or improving glucose metabolism or insulin sensitivity. In conclusion, macrophage-derived TNF-α is not a causative factor for the induction of obesity-associated metabolic dysfunction.
Collapse
Affiliation(s)
- Ahmed K. Aladhami
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
- Nursing College, University of Baghdad, Baghdad, Iraq
| | - Christian A. Unger
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Shannon L. Ennis
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Diego Altomare
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| | - Hao Ji
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| | - Marion C. Hope
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Kandy T. Velázquez
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Reilly T. Enos
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
129
|
Slaughter VL, Rumsey JW, Boone R, Malik D, Cai Y, Sriram NN, Long CJ, McAleer CW, Lambert S, Shuler ML, Hickman JJ. Validation of an adipose-liver human-on-a-chip model of NAFLD for preclinical therapeutic efficacy evaluation. Sci Rep 2021; 11:13159. [PMID: 34162924 PMCID: PMC8222323 DOI: 10.1038/s41598-021-92264-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease and strongly correlates with the growing incidence of obesity and type II diabetes. We have developed a human-on-a-chip model composed of human hepatocytes and adipose tissue chambers capable of modeling the metabolic factors that contribute to liver disease development and progression, and evaluation of the therapeutic metformin. This model uses a serum-free, recirculating medium tailored to represent different human metabolic conditions over a 14-day period. The system validated the indirect influence of adipocyte physiology on hepatocytes that modeled important aspects of NAFLD progression, including insulin resistant biomarkers, differential adipokine signaling in different media and increased TNF-α-induced steatosis observed only in the two-tissue model. This model provides a simple but unique platform to evaluate aspects of an individual factor's contribution to NAFLD development and mechanisms as well as evaluate preclinical drug efficacy and reassess human dosing regimens.
Collapse
Affiliation(s)
- Victoria L Slaughter
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - John W Rumsey
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Rachel Boone
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Duaa Malik
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Yunqing Cai
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | | | - Christopher J Long
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | | | - Stephen Lambert
- College of Medicine, University of Central Florida, 6850 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Michael L Shuler
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - J J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA.
| |
Collapse
|
130
|
De Luca M, Mandala M, Rose G. Towards an understanding of the mechanoreciprocity process in adipocytes and its perturbation with aging. Mech Ageing Dev 2021; 197:111522. [PMID: 34147549 DOI: 10.1016/j.mad.2021.111522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/29/2021] [Accepted: 06/15/2021] [Indexed: 12/25/2022]
Abstract
Adipose tissue (AT) is a complex organ, with multiple functions that are essential for maintaining metabolic health. A feature of AT is its capability to expand in response to physiological challenges, such as pregnancy and aging, and during chronic states of positive energy balance occurring throughout life. AT grows through adipogenesis and/or an increase in the size of existing adipocytes. One process that is required for healthy AT growth is the remodeling of the extracellular matrix (ECM), which is a necessary step to restore mechanical homeostasis and maintain tissue integrity and functionality. While the relationship between mechanobiology and adipogenesis is now well recognized, less is known about the role of adipocyte mechanosignaling pathways in AT growth. In this review article, we first summarize evidence linking ECM remodelling to AT expansion and how its perturbation is associated to a metabolically unhealthy phenotype. Subsequently, we highlight findings suggesting that molecules involved in the dynamic, bidirectional process (mechanoreciprocity) enabling adipocytes to sense changes in the mechanical properties of the ECM are interconnected to pathways regulating lipid metabolism. Finally, we discuss processes through which aging may influence the ability of adipocytes to appropriately respond to alterations in ECM composition.
Collapse
Affiliation(s)
- Maria De Luca
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Maurizio Mandala
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, 87036, Italy
| | - Giuseppina Rose
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, 87036, Italy
| |
Collapse
|
131
|
Caravà E, Moretto P, Caon I, Parnigoni A, Passi A, Karousou E, Vigetti D, Canino J, Canobbio I, Viola M. HA and HS Changes in Endothelial Inflammatory Activation. Biomolecules 2021; 11:biom11060809. [PMID: 34072476 PMCID: PMC8229641 DOI: 10.3390/biom11060809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular diseases are a group of disorders caused by the presence of a combination of risk factors, such as tobacco use, unhealthy diet and obesity, physical inactivity, etc., which cause the modification of the composition of the vessel’s matrix and lead to the alteration of blood flow, matched with an inflammation condition. Nevertheless, it is not clear if the inflammation is a permissive condition or a consequent one. In order to investigate the effect of inflammation on the onset of vascular disease, we treated endothelial cells with the cytokine TNF-α that is increased in obese patients and is reported to induce cardiometabolic diseases. The inflammation induced a large change in the extracellular matrix, increasing the pericellular hyaluronan and altering the heparan sulfate Syndecans sets, which seems to be related to layer permeability but does not influence cell proliferation or migration nor induce blood cell recruitment or activation.
Collapse
Affiliation(s)
- Elena Caravà
- Quantix Italia S.r.l., 20121 Milano, Italy;
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (P.M.); (I.C.); (A.P.); (A.P.); (E.K.); (D.V.)
| | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (P.M.); (I.C.); (A.P.); (A.P.); (E.K.); (D.V.)
| | - Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (P.M.); (I.C.); (A.P.); (A.P.); (E.K.); (D.V.)
| | - Arianna Parnigoni
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (P.M.); (I.C.); (A.P.); (A.P.); (E.K.); (D.V.)
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (P.M.); (I.C.); (A.P.); (A.P.); (E.K.); (D.V.)
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (P.M.); (I.C.); (A.P.); (A.P.); (E.K.); (D.V.)
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (P.M.); (I.C.); (A.P.); (A.P.); (E.K.); (D.V.)
| | - Jessica Canino
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (J.C.); (I.C.)
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy; (J.C.); (I.C.)
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (P.M.); (I.C.); (A.P.); (A.P.); (E.K.); (D.V.)
- Correspondence: ; Tel.: +39-0332-397143
| |
Collapse
|
132
|
Peripheral Blood Mononuclear Cells Oxidative Stress and Plasma Inflammatory Biomarkers in Adults with Normal Weight, Overweight and Obesity. Antioxidants (Basel) 2021; 10:antiox10050813. [PMID: 34065281 PMCID: PMC8161114 DOI: 10.3390/antiox10050813] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 12/28/2022] Open
Abstract
Background: Obesity is an important pathology in public health worldwide. Obese patients are characterized by higher cardiovascular risk and a pro-inflammatory profile. Objective: To assess the oxidative stress in peripheral blood mononuclear cells (PBMCs) and inflammatory biomarkers in plasma in adults with normal weight, overweight and obesity. Methods: One hundred and fifty adults (55-80-years-old; 60% women) from the Balearic Islands, Spain, were recruited and classified according to body mass index (BMI). Anthropometric measurements were carried out, fasting blood samples were collected and plasma and PBMCs were obtained. Biochemical parameters, hemogram, antioxidant enzyme activities and protein levels, reactive oxygen species production (ROS), malondialdehyde (MDA), and cytokine (tumour necrosis factor, TNFα, and interleukin 6, IL-6) levels were measured. Results: Glycaemia, triglyceridemia, abdominal obesity, and waist-to-height ratio (WHtR) were higher, and HDL-cholesterol was lower in obese patients. MDA and TNFα plasma levels were higher in the obese compared to normal-weight group, while the levels of IL-6 were higher in both obese and overweight subjects with respect to normal-weight peers. The activities of all antioxidant enzymes in PBMCs as well as the production ROS progressively increased with BMI. The protein levels of catalase in PBMCs were higher in obese and glutathione reductase in obese and overweight subjects compared to normal-weight peers. No other differences were observed. Conclusion: The current results show that overweight and obesity are related to an increase in pro-oxidant and proinflammatory status in plasma and PBMCs. The studied biomarkers may be useful for monitoring the progression/reversal of obesity.
Collapse
|
133
|
Kralova E, Marusakova M, Hadova K, Krenek P, Klimas J. Dapagliflozin elevates plasma high-density lipoprotein levels and influences visceral fat gene expression in streptozotocin-induced diabetes mellitus. J Pharm Pharmacol 2021; 73:778-784. [PMID: 33749792 DOI: 10.1093/jpp/rgab005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/13/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Dapagliflozin (Dapa) could potentially be used to treat type 1 diabetes mellitus. We tested the hypothesis that it would influence blood lipid levels and visceral fat accumulation in a rodent diabetic model. METHODS We used three groups of male Wistar rats: Controls, streptozotocin (STZ)-treated rats and STZ-treated orally with Dapa (STZ+Dapa), 10 mg/kg/day for six weeks. Blood glucose and serum lipids levels were determined. Plasma levels of lipases (hormone-sensitive lipase, HSL and lipoprotein lipase, LPL), adipokines (leptin and adiponectin) and proinflammatory cytokines [tumour necrosis factor-alpha (TNFα) and interleukin-6 (IL-6)] were determined by ELISA assays. mRNA levels in the perirenal fat were determined by real-time PCR. KEY FINDINGS Dapa suppressed STZ-related hyperglycemia by 20% (P < 0.05) and increased serum HDL when compared to the controls and the STZ-only treated rats (both P < 0.05). STZ treatment caused elevations of other serum lipids that were resistant to Dapa treatment. Dapa treatment also increased both plasma and visceral fat mRNA levels of leptin, LPL and IL-6, while decreasing plasma and fat expressions of HSL and TNFα compared to the STZ-only treated rats (all P < 0.05). CONCLUSIONS Our results suggest that Dapa, in addition to its antidiabetic effect, also influences the function of adipose tissue which could be beneficial in the treatment of diabetes.
Collapse
Affiliation(s)
- Eva Kralova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Margareta Marusakova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Katarina Hadova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Peter Krenek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
134
|
Adipocyte, Immune Cells, and miRNA Crosstalk: A Novel Regulator of Metabolic Dysfunction and Obesity. Cells 2021; 10:cells10051004. [PMID: 33923175 PMCID: PMC8147115 DOI: 10.3390/cells10051004] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is characterized as a complex and multifactorial excess accretion of adipose tissue (AT) accompanied with alterations in the immune response that affects virtually all age and socioeconomic groups around the globe. The abnormal accumulation of AT leads to several metabolic diseases, including nonalcoholic fatty liver disorder (NAFLD), low-grade inflammation, type 2 diabetes mellitus (T2DM), cardiovascular disorders (CVDs), and cancer. AT is an endocrine organ composed of adipocytes and immune cells, including B-Cells, T-cells and macrophages. These immune cells secrete various cytokines and chemokines and crosstalk with adipokines to maintain metabolic homeostasis and low-grade chronic inflammation. A novel form of adipokines, microRNA (miRs), is expressed in many developing peripheral tissues, including ATs, T-cells, and macrophages, and modulates the immune response. miRs are essential for insulin resistance, maintaining the tumor microenvironment, and obesity-associated inflammation (OAI). The abnormal regulation of AT, T-cells, and macrophage miRs may change the function of different organs including the pancreas, heart, liver, and skeletal muscle. Since obesity and inflammation are closely associated, the dysregulated expression of miRs in inflammatory adipocytes, T-cells, and macrophages suggest the importance of miRs in OAI. Therefore, in this review article, we have elaborated the role of miRs as epigenetic regulators affecting adipocyte differentiation, immune response, AT browning, adipogenesis, lipid metabolism, insulin resistance (IR), glucose homeostasis, obesity, and metabolic disorders. Further, we will discuss a set of altered miRs as novel biomarkers for metabolic disease progression and therapeutic targets for obesity.
Collapse
|
135
|
Dwaib HS, AlZaim I, Eid AH, Obeid O, El-Yazbi AF. Modulatory Effect of Intermittent Fasting on Adipose Tissue Inflammation: Amelioration of Cardiovascular Dysfunction in Early Metabolic Impairment. Front Pharmacol 2021; 12:626313. [PMID: 33897419 PMCID: PMC8062864 DOI: 10.3389/fphar.2021.626313] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiometabolic syndrome (CMS) is a cluster of maladaptive cardiovascular, renal, thrombotic, inflammatory, and metabolic disorders. It confers a high risk of cardiovascular mortality and morbidity. CMS is triggered by major shifts in lifestyle and dietary habits with increased consumption of refined, calorie-dense diets. Evidence indicates that diet-induced CMS is linked to Adipose tissue (AT) inflammation. This led to the proposal that adipose inflammation may be involved in metabolic derangements, such as insulin resistance and poor glycemic control, as well as the contribution to the inflammatory process predisposing patients to increased cardiovascular risk. Therefore, in the absence of direct pharmacological interventions for the subclinical phase of CMS, time restricted feeding regimens were anticipated to alleviate early metabolic damage and subsequent comorbidities. These regimens, referred to as intermittent fasting (IF), showed a strong positive impact on the metabolic state of obese and non-obese human subjects and animal models, positive AT remodeling in face of overnutrition and high fat diet (HFD) consumption, and improved CV outcomes. Here, we summarize the available evidence on the role of adipose inflammation in triggering cardiovascular impairment in the context of diet induced CMS with an emphasis on the involvement of perivascular adipose tissue. As well, we propose some possible molecular pathways linking intermittent fasting to the ameliorative effect on adipose inflammation and cardiovascular dysfunction under such circumstances. We highlight a number of targets, whose function changes in perivascular adipose tissue inflammation and could be modified by intermittent fasting acting as a novel approach to ameliorate the inflammatory status.
Collapse
Affiliation(s)
- Haneen S. Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Omar Obeid
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Al-Alamein International University, Alamein, Egypt
| |
Collapse
|
136
|
An J, Lee H, Lee S, Song Y, Kim J, Park IH, Kong H, Kim K. Modulation of Pro-inflammatory and Anti-inflammatory Cytokines in the Fat by an Aloe Gel-based Formula, QDMC, Is Correlated with Altered Gut Microbiota. Immune Netw 2021; 21:e15. [PMID: 33996171 PMCID: PMC8099612 DOI: 10.4110/in.2021.21.e15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
Abnormal inflammatory responses are closely associated with intestinal microbial dysbiosis. Oral administration of Qmatrix-diabetes-mellitus complex (QDMC), an Aloe gel-based formula, has been reported to improve inflammation in type 2 diabetic mice; however, the role of the gut microbiota in ameliorating efficacy of QDMC remains unclear. We investigated the effect of QDMC on the gut microbiota in a type 2 diabetic aged mouse model that was administered a high-fat diet. Proinflammatory (TNF-α and IL-6) and anti-inflammatory (IL-4 and IL-10) cytokine levels in the fat were normalized via oral administration of QDMC, and relative abundances of Bacteroides, Butyricimonas, Ruminococcus, and Mucispirillum were simultaneously significantly increased. The abundance of these bacteria was correlated to the expression levels of cytokines. Our findings suggest that the immunomodulatory activity of QDMC is partly mediated by the altered gut microbiota composition.
Collapse
Affiliation(s)
- Jinho An
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| | - Heetae Lee
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| | - Sungwon Lee
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| | - Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| | - Jiyeon Kim
- Climacteric Natural Products Research Institute, Sahmyook University, Seoul 01795, Korea
| | - Il Ho Park
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| | - Hyunseok Kong
- Climacteric Natural Products Research Institute, Sahmyook University, Seoul 01795, Korea.,College of Animal Biotechnology and Resource, Sahmyook University, Seoul 01795, Korea
| | - Kyungjae Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| |
Collapse
|
137
|
Kashiwagi K, Takayama M, Ichikawa H, Takaishi H, Iwao Y, Kanai T. A significant association of non-obese non-alcoholic fatty liver disease with osteosarcopenic obesity in females 50 years and older. Clin Nutr ESPEN 2021; 42:166-172. [PMID: 33745573 DOI: 10.1016/j.clnesp.2021.01.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/27/2021] [Accepted: 01/30/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Osteosarcopenic obesity (OSO) encompassing obesity, sarcopenia and osteopenia, is due to redistribution or infiltration of fat into muscle and bone. This cross-sectional study evaluated the association between OSO and non-alcoholic fatty liver disease (NAFLD). METHODS Obesity, sarcopenia and osteopenia was defined using the percentage of body fat mass, reduced muscle mass, and the percentage of young adult mean < 80%, measured by dual-energy x-ray absorptiometry, respectively. Non-obese and obese NAFLD was diagnosed by ultrasound and body mass index cut-off point (25 kg/m2). A total of 619 subjects ≥ 50 years who completed health checkups were divided into obesity group including OSO and sarcopenic obesity (SO) alone phenotype, and non-obesity group that did not belong to any phenotype, including standard (St). RESULTS Overall osteopenia and OSO were detected in only 10% and 1% in males, compared with 45% and 9% in females, respectively. Multivariate analysis for females demonstrated a significant association of OSO with non-obese NAFLD (odds ratio = 3.737, 95% confidence interval = 1.365-10.233, P = 0.010), while the association between SO alone and non-obese NAFLD was equivocal. The OSO phenotype had a significantly higher proportion of slower walking speed and weaker grip strength, compared to the St phenotype. The proportion of OSO increased with age in contrast to constant prevalence of non-obese NAFLD. CONCLUSION Non-obese NAFLD had a significant association with OSO in females, independent of plausible confounders. These results suggest that non-obese NAFLD might be an independent risk factor for OSO.
Collapse
Affiliation(s)
| | - Michiyo Takayama
- Center for Preventive Medicine, Keio University Hospital, Tokyo, Japan
| | - Hitoshi Ichikawa
- Center for Preventive Medicine, Keio University Hospital, Tokyo, Japan
| | - Hiromasa Takaishi
- Center for Preventive Medicine, Keio University Hospital, Tokyo, Japan
| | - Yasushi Iwao
- Center for Preventive Medicine, Keio University Hospital, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
138
|
The complex role of adipokines in obesity, inflammation, and autoimmunity. Clin Sci (Lond) 2021; 135:731-752. [PMID: 33729498 PMCID: PMC7969664 DOI: 10.1042/cs20200895] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
The global obesity epidemic is a major contributor to chronic disease and disability in the world today. Since the discovery of leptin in 1994, a multitude of studies have characterized the pathological changes that occur within adipose tissue in the obese state. One significant change is the dysregulation of adipokine production. Adipokines are an indispensable link between metabolism and optimal immune system function; however, their dysregulation in obesity contributes to chronic low-grade inflammation and disease pathology. Herein, I will highlight current knowledge on adipokine structure and physiological function, and focus on the known roles of these factors in the modulation of the immune response. I will also discuss adipokines in rheumatic and autoimmune diseases.
Collapse
|
139
|
Maissan P, Mooij EJ, Barberis M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. BIOLOGY 2021; 10:194. [PMID: 33806509 PMCID: PMC7999230 DOI: 10.3390/biology10030194] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Sirtuins are a family of highly conserved NAD+-dependent proteins and this dependency links Sirtuins directly to metabolism. Sirtuins' activity has been shown to extend the lifespan of several organisms and mainly through the post-translational modification of their many target proteins, with deacetylation being the most common modification. The seven mammalian Sirtuins, SIRT1 through SIRT7, have been implicated in regulating physiological responses to metabolism and stress by acting as nutrient sensors, linking environmental and nutrient signals to mammalian metabolic homeostasis. Furthermore, mammalian Sirtuins have been implicated in playing major roles in mammalian pathophysiological conditions such as inflammation, obesity and cancer. Mammalian Sirtuins are expressed heterogeneously among different organs and tissues, and the same holds true for their substrates. Thus, the function of mammalian Sirtuins together with their substrates is expected to vary among tissues. Any therapy depending on Sirtuins could therefore have different local as well as systemic effects. Here, an introduction to processes relevant for the actions of Sirtuins, such as metabolism and cell cycle, will be followed by reasoning on the system-level function of Sirtuins and their substrates in different mammalian tissues. Their involvement in the healthy metabolism and metabolic disorders will be reviewed and critically discussed.
Collapse
Affiliation(s)
- Parcival Maissan
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Eva J. Mooij
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| |
Collapse
|
140
|
Souza ATP, Freitas GP, Lopes HB, Totoli GGC, Tarone AG, Marostica-Junior MR, Rosa AL, Beloti MM. Jabuticaba peel extract modulates adipocyte and osteoblast differentiation of MSCs from healthy and osteoporotic rats. J Bone Miner Metab 2021; 39:163-173. [PMID: 32889573 DOI: 10.1007/s00774-020-01152-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/20/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The jabuticaba peel extract (JPE) contains bioactive compounds that regulate fat metabolism. Because the negative correlation between fat accumulation and bone formation in bone marrow, we hypothesized that JPE inhibits adipocyte as well as favors osteoblast differentiation of mesenchymal stromal cells (MSCs) under healthy and osteoporotic conditions, a disease that display an imbalance between adipocyte and osteoblast differentiation resulting in reduced bone mass. MATERIAL AND METHODS To test these hypotheses, bone marrow MSCs were harvested from healthy and osteoporotic rats and cultured in adipogenic and osteogenic media with three concentrations of JPE, 0.25, 5 and 10 µg/ml, and vehicle (control). After selecting the most efficient concentrations of JPE, we used them to evaluate adipocyte and osteoblast differentiation of MSCs from both sources. RESULTS We observed that, in general, JPE inhibited adipocyte differentiation of MSCs with more pronounced effects in cells from healthy than osteoporotic rats. In addition, JPE increased osteoblast differentiation, exhibiting a slightly higher osteogenic potential on MSCs from osteoporotic compared to healthy condition. CONCLUSION Our results demonstrated that JPE drives MSCs to inhibit adipocyte differentiation and toward osteoblast differentiation under healthy and osteoporotic conditions. These findings pave the way for further translational studies to investigate the therapeutic possibilities of JPE in both prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Alann Thaffarell Portilho Souza
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Av Do Café S/N, Ribeirão Preto, SP, 14040-904, Brazil
| | - Gileade Pereira Freitas
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Av Do Café S/N, Ribeirão Preto, SP, 14040-904, Brazil
| | - Helena Bacha Lopes
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Av Do Café S/N, Ribeirão Preto, SP, 14040-904, Brazil
| | - Gabriela Guaraldo Campos Totoli
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Av Do Café S/N, Ribeirão Preto, SP, 14040-904, Brazil
| | - Adriana Gadioli Tarone
- School of Food Engineering, University of Campinas, Rua Monteiro Lobato 80, Campinas, 13083-862, SP, Brazil
| | | | - Adalberto Luiz Rosa
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Av Do Café S/N, Ribeirão Preto, SP, 14040-904, Brazil
| | - Marcio Mateus Beloti
- Bone Research Lab, School of Dentistry of Ribeirão Preto, University of São Paulo, Av Do Café S/N, Ribeirão Preto, SP, 14040-904, Brazil.
| |
Collapse
|
141
|
Oliva-Olivera W, Castellano-Castillo D, von Meyenn F, Cardona F, Lönnberg T, Tinahones FJ. Human adipose tissue-derived stem cell paracrine networks vary according metabolic risk and after TNFα-induced death: An analysis at the single-cell level. Metabolism 2021; 116:154466. [PMID: 33333081 DOI: 10.1016/j.metabol.2020.154466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Adipose tissue-derived stem cells (ASCs) might play an important role in adipose microenvironment remodelling during tissue expansion through their response to hypoxia. We examined the cytokine profiles of hypoxic visceral ASCs (hypox-visASCs) from subjects with different metabolic risk, the interactions between cytokines as well as the impact of TNFα-induced death in the behavior of surviving hypoxic subcutaneous ASCs (hypox-subASCs) both at bulk population and single-cell level. MATERIALS/METHODS Visceral adipose tissue was processed to isolate the ASCs from 33 subjects grouped into normal weight, obese with and without metabolic syndrome. Multiplex assay was used to simultaneously measure multiple inflammatory, anti-inflammatory and angiogenic cytokines in hypox-visASCs from these patients and to elucidate cytokine profiles of hypox-subASCs upon stimulation with IL1β or TNFα and after TNFα-induced death. qPCR and single-cell RNA-sequencing were also performed to elucidate transcriptional impact in surviving hypox-subASCs after TNFα-induced apoptosis. RESULTS Hypox-visASCs from subjects without metabolic syndrome showed greater secretion levels of inflammatory, anti-inflammatory and angiogenic cytokines compared with those from patients with metabolic syndrome. While IL-1β stimulation was sufficient to increase the secretion levels of these cytokines in hypox-subASCs, TNFα-induced apoptosis also increased their levels and impacted on the expression levels of extracellular matrix proteins, acetyl-CoA producing enzymes and redox-balance proteins in surviving hypox-subASCs. TNFα-induced apoptosis under different glucose concentrations caused selective impoverishment of cell clusters and differentially influenced gene expression profiles of surviving hypox-subASCs. CONCLUSIONS Immunoregulatory and angiogenic functions of hypox-visASCs from patients with metabolic syndrome could be insufficient to promote healthy adipose tissue expansion. TNFα-induced apoptosis may impact on functionality of hypox-subASC populations, whose differential metabolic sensitivity to death could serve to manipulate individual populations selectively in order to elucidate their role in shaping adipose heterogeneity and treating metabolic disorders.
Collapse
Affiliation(s)
- Wilfredo Oliva-Olivera
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital (IBIMA), Malaga University, Malaga, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III (ISCIII), Malaga, Spain.
| | | | - Ferdinand von Meyenn
- Institute of Food, Nutrition and Health, ETH Zurich, CH-8603 Schwerzenbach, Switzerland
| | - Fernando Cardona
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital (IBIMA), Malaga University, Malaga, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III (ISCIII), Malaga, Spain
| | - Tapio Lönnberg
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
| | - Francisco J Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital (IBIMA), Malaga University, Malaga, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III (ISCIII), Malaga, Spain
| |
Collapse
|
142
|
Cooper PO, Haas MR, Noonepalle SKR, Shook BA. Dermal Drivers of Injury-Induced Inflammation: Contribution of Adipocytes and Fibroblasts. Int J Mol Sci 2021; 22:1933. [PMID: 33669239 PMCID: PMC7919834 DOI: 10.3390/ijms22041933] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023] Open
Abstract
Irregular inflammatory responses are a major contributor to tissue dysfunction and inefficient repair. Skin has proven to be a powerful model to study mechanisms that regulate inflammation. In particular, skin wound healing is dependent on a rapid, robust immune response and subsequent dampening of inflammatory signaling. While injury-induced inflammation has historically been attributed to keratinocytes and immune cells, a vast body of evidence supports the ability of non-immune cells to coordinate inflammation in numerous tissues and diseases. In this review, we concentrate on the active participation of tissue-resident adipocytes and fibroblasts in pro-inflammatory signaling after injury, and how altered cellular communication from these cells can contribute to irregular inflammation associated with aberrant wound healing. Furthering our understanding of how tissue-resident mesenchymal cells contribute to inflammation will likely reveal new targets that can be manipulated to regulate inflammation and repair.
Collapse
Affiliation(s)
| | | | | | - Brett A. Shook
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.O.C.); (M.R.H.); (S.k.R.N.)
| |
Collapse
|
143
|
Cancer Related Anemia: An Integrated Multitarget Approach and Lifestyle Interventions. Nutrients 2021; 13:nu13020482. [PMID: 33535496 PMCID: PMC7912724 DOI: 10.3390/nu13020482] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer is often accompanied by worsening of the patient's iron profile, and the resulting anemia could be a factor that negatively impacts antineoplastic treatment efficacy and patient survival. The first line of therapy is usually based on oral or intravenous iron supplementation; however, many patients remain anemic and do not respond. The key might lie in the pathogenesis of the anemia itself. Cancer-related anemia (CRA) is characterized by a decreased circulating serum iron concentration and transferrin saturation despite ample iron stores, pointing to a more complex problem related to iron homeostatic regulation and additional factors such as chronic inflammatory status. This review explores our current understanding of iron homeostasis in cancer, shedding light on the modulatory role of hepcidin in intestinal iron absorption, iron recycling, mobilization from liver deposits, and inducible regulators by infections and inflammation. The underlying relationship between CRA and systemic low-grade inflammation will be discussed, and an integrated multitarget approach based on nutrition and exercise to improve iron utilization by reducing low-grade inflammation, modulating the immune response, and supporting antioxidant mechanisms will also be proposed. Indeed, a Mediterranean-based diet, nutritional supplements and exercise are suggested as potential individualized strategies and as a complementary approach to conventional CRA therapy.
Collapse
|
144
|
Strobel HA, Gerton T, Hoying JB. Vascularized adipocyte organoid model using isolated human microvessel fragments. Biofabrication 2021; 13. [PMID: 33513595 DOI: 10.1088/1758-5090/abe187] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022]
Abstract
Tissue organoids are proving valuable for modeling tissue health and disease in a variety of applications. This is due, in part, to the dynamic cell-cell interactions fostered within the 3D tissue-like space. To this end, the more that organoids recapitulate the different cell-cell interactions found in native tissue, such as that between parenchyma and the microvasculature, the better the fidelity of the model. The microvasculature, which is comprised of a spectrum of cell types, provides not only perfusion in its support of tissue health, but also important cellular interactions and biochemical dynamics important in tissue phenotype and function. Here, we incorporate whole, intact human microvessel fragments isolated from adipose tissue into organoids to form both MSC and adipocyte vascularized organoids. Isolated microvessels retain their native structure and cell composition, providing a more complete representation of the microvasculature within the organoids. Microvessels expanded via sprouting angiogenesis within organoids comprised of either MSCs or MSC-derived adipocytes and grew out of the organoids when placed in a 3D collagen matrix. In MSC organoids, a ratio of 50 MSCs to 1 microvessel fragment created the optimal vascularization response. We developed a new differentiation protocol that enabled the differentiation of MSCs into adipocytes while simultaneously promoting microvessel angiogenesis. The adipocyte organoids contained vascular networks, were responsive in a lipolysis assay, and expressed the functional adipocyte markers adiponectin and PPARγ. The presence of microvessels promoted insulin receptor expression by adipocytes and modified IL-6 secretion following a TNF-alpha challenge. Overall, we demonstrate a robust method for vascularizing high cell-density organoids with potential implications for other tissues as well.
Collapse
Affiliation(s)
- Hannah A Strobel
- Advanced Solutions Life Sciences, 500 N Commercial Street, Suite 200, Manchester, Manchester, New Hampshire, 03101, UNITED STATES
| | - Thomas Gerton
- Advanced Solutions Life Sciences, 500 N Commercial Street, Suite 200, Manchester, Manchester, New Hampshire, 03101, UNITED STATES
| | - James B Hoying
- Advanced Solutions Life Sciences, 500 N Commercial St, United States, Manchester, New Hampshire, 03101, UNITED STATES
| |
Collapse
|
145
|
Haas de Mello A, Ferreira GK, Rezin GT. Abnormal mitochondrial metabolism in obesity and insulin resistance. CLINICAL BIOENERGETICS 2021:83-92. [DOI: 10.1016/b978-0-12-819621-2.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
146
|
Yang YH, Hao YM, Liu XF, Gao X, Wang BZ, Takahashi K, Du L. Docosahexaenoic acid-enriched phospholipids and eicosapentaenoic acid-enriched phospholipids inhibit tumor necrosis factor-alpha-induced lipolysis in 3T3-L1 adipocytes by activating sirtuin 1 pathways. Food Funct 2021; 12:4783-4796. [PMID: 34100500 DOI: 10.1039/d1fo00157d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Some chronic diseases such as cancer-associated cachexia (CAC) and obesity are associated with the overproduction of tumor necrosis factor-alpha (TNF-α) that stimulates excess lipolysis in adipocytes. Our previous studies have shown that docosahexaenoic acid-enriched phospholipids (DHA-PL) and eicosapentaenoic acid-enriched phospholipids (EPA-PL) ameliorated CAC and obesity-related metabolic disorders. To identify the molecular mechanisms involved, we examined the impact and the associated signaling pathways of DHA-PL and EPA-PL on TNF-α-induced lipolysis in 3T3-L1 adipocytes. The present results revealed that DHA-PL and EPA-PL inhibited the TNF-α-induced increase of glycerol release and protected lipid droplets. In addition, DHA-PL and EPA-PL increased DHA and EPA contents in the phospholipid fraction of adipocytes, respectively. Moreover, DHA-PL and EPA-PL enhanced sirtuin 1 (SIRT1) deacetylase activity and its protein expression. By activating SIRT1, DHA-PL and EPA-PL upregulated the G0/G1 switch gene 2 protein level to inhibit adipose triglyceride lipase activity, activate AMP-activated protein kinase to reverse the downregulation of perilipin expression and phosphorylation of hormone-sensitive lipase (HSL) at Ser565 and prevent the phosphorylation of HSL at Ser660. Furthermore, DHA-PL and EPA-PL improved glucose uptake and glucose transporter type 4 translocation to the plasma membrane in TNF-α-treated adipocytes. Thus, it was concluded that DHA-PL and EPA-PL inhibit TNF-α-induced lipolysis in 3T3-L1 adipocytes by activating the SIRT1 pathways.
Collapse
Affiliation(s)
- Yu-Hong Yang
- School of Food Science & Engineering, Qilu University of Technology (Shandong Academy of Sciences), No. 3501, Daxue Road, Jinan, Shandong 250353, China
| | - Yi-Ming Hao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| | - Xiao-Fang Liu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Sustainable Development of Polar Fishery, Ministry of Agriculture and Rural Affairs, No. 106 Nanjing Road, Qingdao, Shandong 266071, China
| | - Xiang Gao
- College of Life Sciences, Qingdao University, No. 308 Ningxia Road, Qingdao, Shandong 266071, China
| | - Bao-Zhen Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| | - Koretaro Takahashi
- Faculty of Engineering, Kitami Institute of Technology, 165 Koen-cho, Kitami, Hokkaido, 090-8507, Japan
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| |
Collapse
|
147
|
TNF-TNFR2 Signal Plays a Decisive Role in the Activation of CD4 +Foxp3 + Regulatory T Cells: Implications in the Treatment of Autoimmune Diseases and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:257-272. [PMID: 33523452 DOI: 10.1007/978-981-15-6407-9_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The puzzling biphasic or dual roles of tumor necrosis factor α (TNF) in the inflammatory and immune responses are likely to be mediated by distinct signaling pathways transduced by one of its two receptors, e.g., TNF receptor type I (TNFR1) and TNF receptor type II (TNFR2). Unlike TNFR1 that is ubiquitously expressed on almost all types of cells, the expression of TNFR2 is rather restricted to certain types of cells, such as T lymphocytes. There is now compelling evidence that TNFR2 is preferentially expressed by CD4+Foxp3+ regulatory T cells (Tregs), and TNFR2 plays a decisive role in the activation, expansion, in vivo function, and phenotypical stability of Tregs. In this chapter, the current understanding of the molecular basis and signaling pathway of TNF-TNFRs signal is introduced. Latest studies that have further supported and substantiated the pivotal role of TNF-TNFR2 interaction in Tregs biology and its molecular basis are discussed. The research progress regarding TNFR2-targeting treatment for autoimmune diseases and cancer is analyzed. Future study should focus on the further understanding of molecular mechanism underlying Treg-stimulatory effect of TNFR2 signal, as well as on the translation of research findings into therapeutic benefits of human patients with autoimmune diseases, allergy, allograft rejection, and cancer.
Collapse
|
148
|
Maftei D, Lattanzi R, Vincenzi M, Squillace S, Fullone MR, Miele R. The balance of concentration between Prokineticin 2β and Prokineticin 2 modulates the food intake by STAT3 signaling. BBA ADVANCES 2021; 1:100028. [PMID: 37082024 PMCID: PMC10074905 DOI: 10.1016/j.bbadva.2021.100028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/31/2022] Open
Abstract
The secreted bioactive peptide prokineticin 2 (PK2) is a potent adipokine and its central and peripheral administration reduces food intake in rodents. The pk2 gene has two splice variants, PK2 and PK2L (PK2 long form), which is cleaved into an active peptide, PK2β, that preferentially binds prokineticin receptor 1 (PKR1). We investigated the role of PK2β in the regulation of food intake. We demonstrated that intraperitoneal injection of PK2β, in contrast to PK2, did not reduce food intake in mice. Exposure of hypotalamic explants to PK2, but not PK2β, induced phosphorylation of STAT3 and ERK. We also evidenced that in adipocytes from PKR1 knock-out mice, a model of obesity, there were higher PK2β levels than PK2 inducing a decreased activation of STAT3 and ERK. Our results suggest that variations in PK2 and PK2β levels, due to modulation of pk2 gene splicing processes, affect food intake in mice.
Collapse
Affiliation(s)
- Daniela Maftei
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
- Corresponding author: Roberta Lattanzi, Department of Physiology and Pharmacology “Vittorio Erspamer” Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy.
| | - Martina Vincenzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Silvia Squillace
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Maria Rosaria Fullone
- Department of Biochemical Sciences “A. Rossi Fanelli” and CNR-Institute of Molecular Biology and Pathology, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Department of Biochemical Sciences “A. Rossi Fanelli” and CNR-Institute of Molecular Biology and Pathology, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| |
Collapse
|
149
|
Gater DR, Farkas GJ, Tiozzo E. Pathophysiology of Neurogenic Obesity After Spinal Cord Injury. Top Spinal Cord Inj Rehabil 2021; 27:1-10. [PMID: 33814879 PMCID: PMC7983633 DOI: 10.46292/sci20-00067] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Individuals with a spinal cord injury (SCI) have a unique physiology characterized by sarcopenia, neurogenic osteoporosis, neurogenic anabolic deficiency, sympathetic dysfunction, and blunted satiety associated with their SCI, all of which alter energy balance and subsequently body composition. The distinct properties of "neurogenic obesity" place this population at great risk for metabolic dysfunction, including systemic inflammation, hyperglycemia, dyslipidemia, and hypertension. The purpose of this article is to demonstrate the relationship between neurogenic obesity and the metabolic syndrome after SCI, highlighting the mechanisms associated with adipose tissue pathology and those respective comorbidities. Additionally, representative studies of persons with SCI will be provided to elucidate the severity of the problem and to prompt greater vigilance among SCI specialists as well as primary care providers in order to better manage the epidemic from a public health perspective.
Collapse
Affiliation(s)
- David R. Gater
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Gary J. Farkas
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
| | - Eduard Tiozzo
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
150
|
Saeidi A, Haghighi MM, Kolahdouzi S, Daraei A, Abderrahmane AB, Essop MF, Laher I, Hackney AC, Zouhal H. The effects of physical activity on adipokines in individuals with overweight/obesity across the lifespan: A narrative review. Obes Rev 2021; 22:e13090. [PMID: 32662238 DOI: 10.1111/obr.13090] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
This narrative review summarizes current knowledge on the effects of physical activity (PA) on adipokine levels in individuals with overweight and obesity. Approximately 90 investigations including randomized control, cross-sectional and longitudinal studies that reported on the effects of a single session of PA (acute) or long-term PA (chronic) on adipokine levels in individuals with overweight/obesity were reviewed. The findings support the notion that there is consensus on the benefits of chronic exercise training-regardless of the mode (resistance vs. aerobic), intensity and cohort (healthy vs. diabetes)-on adipokine levels (such as tumour necrosis factor-alpha, interleukin-6, adiponectin, visfatin, omentin-1 and leptin). However, several confounding factors (frequency, intensity, time and type of exercise) can alter the magnitude of the effects of an acute exercise session. Available evidence suggests that PA, as a part of routine lifestyle behaviour, improves obesity complications by modulating adipokine levels. However, additional research is needed to help identify the most effective interventions to elicit the most beneficial changes in adipokine levels in individuals with overweight/obesity.
Collapse
Affiliation(s)
- Ayoub Saeidi
- Department of Physical Education, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Marjan Mosalman Haghighi
- Faculty of Medicine and Health, Cardiology Centre, The University of Sydney, The Children's Hospital at Westmead, Sydney, Australia
| | - Sarkawt Kolahdouzi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Mazandaran, Babolsar, Iran
| | - Ali Daraei
- Department of Biological Sciences in Sport, Faculty of Sports Sciences and Health, Shahid Beheshti University, Tehran, Iran
| | | | - M Faadiel Essop
- Centre for Cardio-metabolic Research in Africa (CARMA), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, Canada
| | - Anthony C Hackney
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill, NC, USA
| | - Hassane Zouhal
- Movement, Sport and Health Sciences Laboratory (M2S), UFR-STAPS, University of Rennes 2-ENS Rennes, Rennes, France
| |
Collapse
|