1
|
Ghemiș L, Goriuc A, Minea B, Botnariu GE, Mârțu MA, Ențuc M, Cioloca D, Foia LG. Myeloid-Derived Suppressor Cells (MDSCs) and Obesity-Induced Inflammation in Type 2 Diabetes. Diagnostics (Basel) 2024; 14:2453. [PMID: 39518420 PMCID: PMC11544947 DOI: 10.3390/diagnostics14212453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Type 2 diabetes mellitus is a complex metabolic disorder characterized by insulin resistance and, subsequently, decreased insulin secretion. This condition is closely linked to obesity, a major risk factor that boosts the development of chronic systemic inflammation, which, in turn, is recognized for its crucial role in the onset of insulin resistance. Under conditions of obesity, adipose tissue, particularly visceral fat, becomes an active endocrine organ that releases a wide range of pro-inflammatory mediators, including cytokines, chemokines, and adipokines. These mediators, along with cluster of differentiation (CD) markers, contribute to the maintenance of systemic low-grade inflammation, promote cellular signaling and facilitate the infiltration of inflammatory cells into tissues. Emerging studies have indicated the accumulation of a new cell population in the adipose tissue in these conditions, known as myeloid-derived suppressor cells (MDSCs). These cells possess the ability to suppress the immune system, impacting obesity-related chronic inflammation. Given the limited literature addressing the role of MDSCs in the context of type 2 diabetes, this article aims to explore the complex interaction between inflammation, obesity, and MDSC activity. Identifying and understanding the role of these immature cells is essential not only for improving the management of type 2 diabetes but also for the potential development of targeted therapeutic strategies aimed at both glycemic control and the reduction in associated inflammation.
Collapse
Affiliation(s)
- Larisa Ghemiș
- Department of Biochemistry, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania; (L.G.); (A.G.); (B.M.); (L.G.F.)
| | - Ancuța Goriuc
- Department of Biochemistry, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania; (L.G.); (A.G.); (B.M.); (L.G.F.)
| | - Bogdan Minea
- Department of Biochemistry, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania; (L.G.); (A.G.); (B.M.); (L.G.F.)
| | - Gina Eosefina Botnariu
- Department of Internal Medicine II, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Maria-Alexandra Mârțu
- Department of Periodontology, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania;
| | - Melissa Ențuc
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania;
| | - Daniel Cioloca
- Department of Oro-Dental Prevention, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania;
| | - Liliana Georgeta Foia
- Department of Biochemistry, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iasi, Romania; (L.G.); (A.G.); (B.M.); (L.G.F.)
| |
Collapse
|
2
|
Kakkat S, Suman P, Turbat- Herrera EA, Singh S, Chakroborty D, Sarkar C. Exploring the multifaceted role of obesity in breast cancer progression. Front Cell Dev Biol 2024; 12:1408844. [PMID: 39040042 PMCID: PMC11260727 DOI: 10.3389/fcell.2024.1408844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Obesity is a multifaceted metabolic disorder characterized by excessive accumulation of adipose tissue. It is a well-established risk factor for the development and progression of breast cancer. Adipose tissue, which was once regarded solely as a passive energy storage depot, is now acknowledged as an active endocrine organ producing a plethora of bioactive molecules known as adipokines that contribute to the elevation of proinflammatory cytokines and estrogen production due to enhanced aromatase activity. In the context of breast cancer, the crosstalk between adipocytes and cancer cells within the adipose microenvironment exerts profound effects on tumor initiation, progression, and therapeutic resistance. Moreover, adipocytes can engage in direct interactions with breast cancer cells through physical contact and paracrine signaling, thereby facilitating cancer cell survival and invasion. This review endeavors to summarize the current understanding of the intricate interplay between adipocyte-associated factors and breast cancer progression. Furthermore, by discussing the different aspects of breast cancer that can be adversely affected by obesity, this review aims to shed light on potential avenues for new and novel therapeutic interventions.
Collapse
Affiliation(s)
- Sooraj Kakkat
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Prabhat Suman
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Elba A. Turbat- Herrera
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Seema Singh
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| | - Debanjan Chakroborty
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| | - Chandrani Sarkar
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
3
|
Buckley LA, Kulhanek DR, Bruder A, Gisslen T, Paulsen ME. Inflammation as a Sex-Specific Mediator in the Relationship between Maternal and Offspring Obesity in C57Bl/6J Mice. BIOLOGY 2024; 13:399. [PMID: 38927279 PMCID: PMC11200566 DOI: 10.3390/biology13060399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
Maternal obesity is a well-established risk factor for offspring obesity development. The relationship between maternal and offspring obesity is mediated in part by developmental programming of offspring metabolic circuitry, including hypothalamic signaling. Dysregulated hypothalamic inflammation has also been linked to development of obesity. We utilized an established C57Bl/6J mouse model of high-fat, high-sugar diet induced maternal obesity to evaluate the effect of maternal obesity on systemic and hypothalamic TNF-α, IL-6, and IL-1β levels in neonatal and adult offspring. The offspring of dams with obesity demonstrated increased adiposity and decreased activity compared to control offspring. Maternal obesity was associated with decreased plasma TNF-α, IL-6 and IL-1β in adult female offspring and decreased plasma IL-6 in neonatal male offspring. Neonatal female offspring of obese dams had decreased TNF-α gene expression in the hypothalamus compared to control females, while neonatal and adult male offspring of obese dams had decreased IL-6 gene expression in the hypothalamus compared to control males. In summary, our results highlight important sex differences in the inflammatory phenotype of offspring exposed to maternal obesity. Sex-specific immunomodulatory mechanisms should be considered in future efforts to develop therapeutic interventions for obesity prevention and treatment.
Collapse
Affiliation(s)
- Lauren A. Buckley
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55454, USA; (D.R.K.); (A.B.); (T.G.); (M.E.P.)
| | - Debra R. Kulhanek
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55454, USA; (D.R.K.); (A.B.); (T.G.); (M.E.P.)
| | - Adrienne Bruder
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55454, USA; (D.R.K.); (A.B.); (T.G.); (M.E.P.)
| | - Tate Gisslen
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55454, USA; (D.R.K.); (A.B.); (T.G.); (M.E.P.)
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN 55414, USA
| | - Megan E. Paulsen
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55454, USA; (D.R.K.); (A.B.); (T.G.); (M.E.P.)
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN 55414, USA
| |
Collapse
|
4
|
Dong ZL, Jiao X, Wang ZG, Yuan K, Yang YQ, Wang Y, Li YT, Wang TC, Kan TY, Wang J, Tao HR. D-mannose alleviates intervertebral disc degeneration through glutamine metabolism. Mil Med Res 2024; 11:28. [PMID: 38711073 PMCID: PMC11071241 DOI: 10.1186/s40779-024-00529-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 04/11/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is a multifaceted condition characterized by heterogeneity, wherein the balance between catabolism and anabolism in the extracellular matrix of nucleus pulposus (NP) cells plays a central role. Presently, the available treatments primarily focus on relieving symptoms associated with IVDD without offering an effective cure targeting its underlying pathophysiological processes. D-mannose (referred to as mannose) has demonstrated anti-catabolic properties in various diseases. Nevertheless, its therapeutic potential in IVDD has yet to be explored. METHODS The study began with optimizing the mannose concentration for restoring NP cells. Transcriptomic analyses were employed to identify the mediators influenced by mannose, with the thioredoxin-interacting protein (Txnip) gene showing the most significant differences. Subsequently, small interfering RNA (siRNA) technology was used to demonstrate that Txnip is the key gene through which mannose exerts its effects. Techniques such as colocalization analysis, molecular docking, and overexpression assays further confirmed the direct regulatory relationship between mannose and TXNIP. To elucidate the mechanism of action of mannose, metabolomics techniques were employed to pinpoint glutamine as a core metabolite affected by mannose. Next, various methods, including integrated omics data and the Gene Expression Omnibus (GEO) database, were used to validate the one-way pathway through which TXNIP regulates glutamine. Finally, the therapeutic effect of mannose on IVDD was validated, elucidating the mechanistic role of TXNIP in glutamine metabolism in both intradiscal and orally treated rats. RESULTS In both in vivo and in vitro experiments, it was discovered that mannose has potent efficacy in alleviating IVDD by inhibiting catabolism. From a mechanistic standpoint, it was shown that mannose exerts its anti-catabolic effects by directly targeting the transcription factor max-like protein X-interacting protein (MondoA), resulting in the upregulation of TXNIP. This upregulation, in turn, inhibits glutamine metabolism, ultimately accomplishing its anti-catabolic effects by suppressing the mitogen-activated protein kinase (MAPK) pathway. More importantly, in vivo experiments have further demonstrated that compared with intradiscal injections, oral administration of mannose at safe concentrations can achieve effective therapeutic outcomes. CONCLUSIONS In summary, through integrated multiomics analysis, including both in vivo and in vitro experiments, this study demonstrated that mannose primarily exerts its anti-catabolic effects on IVDD through the TXNIP-glutamine axis. These findings provide strong evidence supporting the potential of the use of mannose in clinical applications for alleviating IVDD. Compared to existing clinically invasive or pain-relieving therapies for IVDD, the oral administration of mannose has characteristics that are more advantageous for clinical IVDD treatment.
Collapse
Affiliation(s)
- Zheng-Lin Dong
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xin Jiao
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zeng-Guang Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Kai Yuan
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi-Qi Yang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yao Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yun-Tao Li
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Tian-Chang Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Tian-You Kan
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jian Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| | - Hai-Rong Tao
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
5
|
Chen CM, Meng XQ, Zhu H, Liu T, Liu Y, Zhou LJ, Zhu GD, Chen XB, Guo XG, Duan SZ. Brown adipocyte mineralocorticoid receptor deficiency impairs metabolic regulation in diet-induced obese mice. J Lipid Res 2023; 64:100449. [PMID: 37734559 PMCID: PMC10622702 DOI: 10.1016/j.jlr.2023.100449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/01/2023] [Accepted: 09/10/2023] [Indexed: 09/23/2023] Open
Abstract
Activation of brown adipose tissue (BAT) contributes to energy dissipation and metabolic health. Although mineralocorticoid receptor (MR) antagonists have been demonstrated to improve metabolism under obesity, the underlying mechanisms remain incompletely understood. We aimed to evaluate the role of BAT MR in metabolic regulation. After 8 weeks of high-fat diet (HFD) feeding, BAT MR KO (BMRKO) mice manifested significantly increased bodyweight, fat mass, serum fasting glucose, and impaired glucose homeostasis compared with littermate control (LC) mice, although insulin resistance and fasting serum insulin were not significantly changed. Metabolic cage experiments showed no change in O2 consumption, CO2 production, or energy expenditure in obese BMRKO mice. RNA sequencing analysis revealed downregulation of genes related to fatty acid metabolism in BAT of BMRKO-HFD mice compared with LC-HFD mice. Moreover, H&E and immunohistochemical staining demonstrated that BMRKO exacerbated HFD-induced macrophage infiltration and proinflammatory genes in epididymal white adipose tissue (eWAT). BMRKO-HFD mice also manifested significantly increased liver weights and hepatic lipid accumulation, an increasing trend of genes related to lipogenesis and lipid uptake, and significantly decreased genes related to lipolytic and fatty acid oxidation in the liver. Finally, the level of insulin-induced AKT phosphorylation was substantially blunted in eWAT but not liver or skeletal muscle of BMRKO-HFD mice compared with LC-HFD mice. These data suggest that BAT MR is required to maintain metabolic homeostasis, likely through its regulation of fatty acid metabolism in BAT and impacts on eWAT and liver.
Collapse
Affiliation(s)
- Chu-Mao Chen
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Qian Meng
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hong Zhu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lu-Jun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Guo-Dong Zhu
- Department of Oncology, Guangzhou Geriatric Hospital, Guangzhou, China
| | - Xiao-Bei Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Xu-Guang Guo
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China; Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China; Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China; Department of Teaching Management, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China; Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China.
| |
Collapse
|
6
|
Adipose-specific deletion of the cation channel TRPM7 inhibits TAK1 kinase-dependent inflammation and obesity in male mice. Nat Commun 2023; 14:491. [PMID: 36717580 PMCID: PMC9887063 DOI: 10.1038/s41467-023-36154-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
Chronic inflammation of white adipose tissue is a key link between obesity and the associated metabolic syndrome. Transient receptor potential melastatin-like 7 (TRPM7) is known to be related to inflammation; however, the role of TRPM7 in adipocyte phenotype and function in obesity remains unclear. Here, we observe that the activation of adipocyte TRPM7 plays an essential role in pro-inflammatory responses. Adult male mice are used in our experiments. Adipocyte-specific deficiency in TRPM7 attenuates the pro-inflammatory phenotype, improves glucose homeostasis, and suppresses weight gain in mice fed a high-fat diet. Mechanistically, the pro-inflammatory effect of TRPM7 is dependent on Ca2+ signaling. Ca2+ influx initiated by TRPM7 enhances transforming growth factor-β activated kinase 1 activation via the co-regulation of calcium/calmodulin-dependent protein kinase II and tumor necrosis factor receptor-associated factor 6, leading to exacerbated nuclear factor kappa B signaling. Additionally, obese mice treated with TRPM7 inhibitor are protected against obesity and insulin resistance. Our results demonstrate TRPM7 as a factor in the development of adipose inflammation that regulates insulin sensitivity in obesity.
Collapse
|
7
|
Bacil GP, Romualdo GR, Piagge PMFD, Cardoso DR, Vinken M, Cogliati B, Barbisan LF. Unraveling Hepatic Metabolomic Profiles and Morphological Outcomes in a Hybrid Model of NASH in Different Mouse Strains. Antioxidants (Basel) 2023; 12:antiox12020290. [PMID: 36829849 PMCID: PMC9952348 DOI: 10.3390/antiox12020290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/31/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses nonalcoholic steatohepatitis (NASH) and affects 25% of the global population. Although a plethora of experimental models for studying NASH have been proposed, still scarce findings regarding the hepatic metabolomic/molecular profile. In the present study, we sought to unravel the hepatic metabolomic profile of mice subjected to a hybrid model of NASH, by combining a Western diet and carbon tetrachloride administration, for 8 weeks, in male C57BL/6J and BALB/c mice. In both mouse strains, the main traits of NASH-metabolic (glucose intolerance profile), morphologic (extensive microvesicular steatosis and fibrosis, lobular inflammation, and adipose tissue-related inflammation/hypertrophy), and molecular (impaired Nrf2/NF-κB pathway dynamics and altered metabolomic profile)-were observed. The hepatic metabolomic profile revealed that the hybrid protocol impaired, in both strains, the abundance of branched chain-aromatic amino acids, carboxylic acids, and glycosyl compounds, that might be linked to the Nrf2 pathway activation. Moreover, we observed a strain-dependent hepatic metabolomic signature, in which the tricarboxylic acid metabolites and pyruvate metabolism were dissimilarly modulated in C57BL/6J and BALB/c mice. Thus, we provide evidence that the strain-dependent hepatic metabolomic profile might be linked to the distinct underlying mechanisms of NASH, also prospecting potential mechanistic insights into the corresponding disease.
Collapse
Affiliation(s)
- Gabriel P. Bacil
- Department of Pathology, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu 18618-689, Brazil
| | - Guilherme R. Romualdo
- Department of Pathology, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu 18618-689, Brazil
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil
| | - Priscila M. F. D. Piagge
- Department of Chemistry and Molecular Physics, São Carlos Institute of Chemistry (IQSC), University of São Paulo (USP), São Carlos 13566-590, Brazil
| | - Daniel R. Cardoso
- Department of Chemistry and Molecular Physics, São Carlos Institute of Chemistry (IQSC), University of São Paulo (USP), São Carlos 13566-590, Brazil
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, University of Vrije, 1090 Brussel, Belgium
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil
| | - Luís F. Barbisan
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil
- Correspondence: ; Tel.: +55-14-3880-0469
| |
Collapse
|
8
|
Chen S, Liu H, Zhang J, Zhou B, Zhuang S, He X, Wang T, Wang C. Effects of different levels of rutin on growth performance, immunity, intestinal barrier and antioxidant capacity of broilers. ITALIAN JOURNAL OF ANIMAL SCIENCE 2022. [DOI: 10.1080/1828051x.2022.2116732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Shun Chen
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Huijuan Liu
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Jiaqi Zhang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Binbin Zhou
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Su Zhuang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Xiaofang He
- Co-Innovation Center for School of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, China
| | - Tian Wang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Chao Wang
- College of Animal Science and Technology, National Experimental Teaching Demonstration Centre of Animal Science, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
9
|
Lecoutre S, Lambert M, Drygalski K, Dugail I, Maqdasy S, Hautefeuille M, Clément K. Importance of the Microenvironment and Mechanosensing in Adipose Tissue Biology. Cells 2022; 11:cells11152310. [PMID: 35954152 PMCID: PMC9367348 DOI: 10.3390/cells11152310] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
The expansion of adipose tissue is an adaptive mechanism that increases nutrient buffering capacity in response to an overall positive energy balance. Over the course of expansion, the adipose microenvironment undergoes continual remodeling to maintain its structural and functional integrity. However, in the long run, adipose tissue remodeling, typically characterized by adipocyte hypertrophy, immune cells infiltration, fibrosis and changes in vascular architecture, generates mechanical stress on adipose cells. This mechanical stimulus is then transduced into a biochemical signal that alters adipose function through mechanotransduction. In this review, we describe the physical changes occurring during adipose tissue remodeling, and how they regulate adipose cell physiology and promote obesity-associated dysfunction in adipose tissue.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Mélanie Lambert
- Labex Inflamex, Université Sorbonne Paris Nord, INSERM, F-93000 Bobigny, France;
| | - Krzysztof Drygalski
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet Hospital, C2-94, 14186 Stockholm, Sweden;
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR 7622), IBPS, Sorbonne Université, F-75005 Paris, France;
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, F-75013 Paris, France
- Correspondence: or
| |
Collapse
|
10
|
Núñez-Ruiz A, Sánchez-Brena F, López-Pacheco C, Acevedo-Domínguez NA, Soldevila G. Obesity modulates the immune macroenvironment associated with breast cancer development. PLoS One 2022; 17:e0266827. [PMID: 35472214 PMCID: PMC9041840 DOI: 10.1371/journal.pone.0266827] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/28/2022] [Indexed: 11/19/2022] Open
Abstract
Growing evidence demonstrates a strong correlation between obesity and an increased risk of breast cancer, although the mechanisms involved have not been completely elucidated. Some reports have described a crosstalk between adipocytes, cancer cells, and immune cells within the tumor microenvironment, however, it is currently unknown whether obesity can promote tumor growth by inducing systemic alterations of the immune cell homeostasis in peripheral lymphoid organs and adipose tissue. Here, we used the E0771 breast cancer cell line in a mouse model of diet-induced obesity to analyze the immune subpopulations present in the tumors, visceral adipose tissue (VAT), and spleen of lean and obese mice. Our results showed a significant reduction in the frequency of infiltrating CD8+ T cells and a decreased M1/M2 macrophage ratio, indicative of the compromised anti-tumoral immune response reported in obesity. Despite not finding differences in the percentage or numbers of intratumoral Tregs, phenotypic analysis showed that they were enriched in CD39+, PD-1+ and CCR8+ cells, compared to the draining lymph nodes, confirming the highly immunosuppressive profile of infiltrating Tregs reported in established tumors. Analysis of peripheral T lymphocytes showed that tumor development in obese mice was associated to a significant increase in the percentage of peripheral Tregs, which supports the systemic immunosuppressive effect caused by the tumor. Interestingly, evaluation of immune subpopulations in the VAT showed that the characteristic increase in the M1/M2 macrophage ratio reported in obesity, was completely reversed in tumor-bearing mice, resembling the M2-polarized profile found in the microenvironment of the growing tumor. Importantly, VAT Tregs, which are commonly decreased in obese mice, were significantly increased in the presence of breast tumors and displayed significantly higher levels of Foxp3, indicating a regulatory feedback mechanism triggered by tumor growth. Altogether, our results identify a complex reciprocal relationship between adipocytes, immune cells, and the tumor, which may modulate the immune macroenvironment that promotes breast cancer development in obesity.
Collapse
Affiliation(s)
- Aleida Núñez-Ruiz
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | - Flor Sánchez-Brena
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | - Cynthia López-Pacheco
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
- Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | | | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
- * E-mail:
| |
Collapse
|
11
|
Lauria F, Iacomino G, Russo P, Venezia A, Marena P, Ahrens W, De Henauw S, Eiben G, Foraita R, Hebestreit A, Kourides Y, Molnár D, Moreno LA, Veidebaum T, Siani A. Circulating miRNAs Are Associated with Inflammation Biomarkers in Children with Overweight and Obesity: Results of the I.Family Study. Genes (Basel) 2022; 13:632. [PMID: 35456438 PMCID: PMC9030192 DOI: 10.3390/genes13040632] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 01/22/2023] Open
Abstract
Increasing data suggest that overnutrition-induced obesity may trigger an inflammatory process in adipose tissue and upturn in the innate immune system. Numerous players have been involved in governing the inflammatory response, including epigenetics. Among epigenetic players, miRNAs are emerging as crucial regulators of immune cell development, immune responses, autoimmunity, and inflammation. In this study, we aimed at identifying the involvement of candidate miRNAs in relation to inflammation-associated biomarkers in a subsample of European children with overweight and obesity participating in the I.Family study. The study sample included individuals with increased adiposity since this condition contributes to the early occurrence of chronic low-grade inflammation. We focused on the acute-phase reagent C-reactive protein (CRP) as the primary outcome and selected cytokines as plausible biomarkers of inflammation. We found that chronic low-grade CRP elevation shows a highly significant association with miR-26b-3p and hsa-miR-576-5p in boys. Furthermore, the association of CRP with hsa-miR-10b-5p and hsa-miR-31-5p is highly significant in girls. We also observed major sex-related associations of candidate miRNAs with selected cytokines. Except for IL-6, a significant association of hsa-miR-26b-3p and hsa-miR-576-5p with TNF-α, IL1-Ra, IL-8, and IL-15 levels was found exclusively in boys. The findings of this exploratory study suggest sex differences in the association of circulating miRNAs with inflammatory response biomarkers, and indicate a possible role of miRNAs among the candidate epigenetic mechanisms related to the process of low-grade inflammation in childhood obesity.
Collapse
Affiliation(s)
- Fabio Lauria
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy; (F.L.); (P.R.); (A.V.); (P.M.); (A.S.)
| | - Giuseppe Iacomino
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy; (F.L.); (P.R.); (A.V.); (P.M.); (A.S.)
| | - Paola Russo
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy; (F.L.); (P.R.); (A.V.); (P.M.); (A.S.)
| | - Antonella Venezia
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy; (F.L.); (P.R.); (A.V.); (P.M.); (A.S.)
| | - Pasquale Marena
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy; (F.L.); (P.R.); (A.V.); (P.M.); (A.S.)
| | - Wolfgang Ahrens
- Leibniz Institute for Prevention Research and Epidemiology-BIPS, Achterstraße 30, 28359 Bremen, Germany; (W.A.); (R.F.); (A.H.)
| | - Stefaan De Henauw
- Department of Public Health and Primary Care, University Hospital 4K3 C. Heymanslaan, 10, 9000 Ghent, Belgium;
| | - Gabriele Eiben
- Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 3, 41390 Göteborg, Sweden;
| | - Ronja Foraita
- Leibniz Institute for Prevention Research and Epidemiology-BIPS, Achterstraße 30, 28359 Bremen, Germany; (W.A.); (R.F.); (A.H.)
| | - Antje Hebestreit
- Leibniz Institute for Prevention Research and Epidemiology-BIPS, Achterstraße 30, 28359 Bremen, Germany; (W.A.); (R.F.); (A.H.)
| | - Yiannis Kourides
- Research and Education Institute of Child Health, 138 Limassol Ave, #205, Strovolos 2015, Cyprus;
| | - Dénes Molnár
- Department of Pediatrics, Medical School, University of Pécs, H-7624 Pecs, Hungary;
| | - Luis A. Moreno
- University of Zaragoza, Domingo Miral s/n, 50009 Zaragoza, Spain;
| | - Toomas Veidebaum
- National Institute for Health Development, Hiiu 42, 11619 Tallinn, Estonia;
| | - Alfonso Siani
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy; (F.L.); (P.R.); (A.V.); (P.M.); (A.S.)
| |
Collapse
|
12
|
The aetiology and molecular landscape of insulin resistance. Nat Rev Mol Cell Biol 2021; 22:751-771. [PMID: 34285405 DOI: 10.1038/s41580-021-00390-6] [Citation(s) in RCA: 252] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 02/07/2023]
Abstract
Insulin resistance, defined as a defect in insulin-mediated control of glucose metabolism in tissues - prominently in muscle, fat and liver - is one of the earliest manifestations of a constellation of human diseases that includes type 2 diabetes and cardiovascular disease. These diseases are typically associated with intertwined metabolic abnormalities, including obesity, hyperinsulinaemia, hyperglycaemia and hyperlipidaemia. Insulin resistance is caused by a combination of genetic and environmental factors. Recent genetic and biochemical studies suggest a key role for adipose tissue in the development of insulin resistance, potentially by releasing lipids and other circulating factors that promote insulin resistance in other organs. These extracellular factors perturb the intracellular concentration of a range of intermediates, including ceramide and other lipids, leading to defects in responsiveness of cells to insulin. Such intermediates may cause insulin resistance by inhibiting one or more of the proximal components in the signalling cascade downstream of insulin (insulin receptor, insulin receptor substrate (IRS) proteins or AKT). However, there is now evidence to support the view that insulin resistance is a heterogeneous disorder that may variably arise in a range of metabolic tissues and that the mechanism for this effect likely involves a unified insulin resistance pathway that affects a distal step in the insulin action pathway that is more closely linked to the terminal biological response. Identifying these targets is of major importance, as it will reveal potential new targets for treatments of diseases associated with insulin resistance.
Collapse
|
13
|
The relationship between Schistosoma and glycolipid metabolism. Microb Pathog 2021; 159:105120. [PMID: 34358648 DOI: 10.1016/j.micpath.2021.105120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 02/02/2023]
Abstract
Diabetes and obesity have become the most popular metabolic diseases in the world. A large number of previous studies have shown that glucose and lipid metabolism disorder is an important risk factor and a main cause of diabetes and obesity. Schistosoma is a parasite transmitted by freshwater snails. It can induce a series of inflammatory and immune reactions after infecting the human body, causing schistosomiasis. However, in recent years, studies have found that Schistosoma infection or Schistosoma related products can improve or prevent some immune and inflammatory diseases, such as severe asthma, inflammatory bowel disease, diabetes and so on. Further experiments have also revealed that Schistosoma can promote the secretion of anti-inflammatory factors and regulate the glucose and lipid metabolism in the host body by polarizing immune cells such as T cells, B cells and dendritic cells (DCs). In this review, we summarize studies that investigated Schistosoma and Schistosoma-derived products and their relationship with glycolipid metabolism and related diseases, highlighting potential protective mechanisms.
Collapse
|
14
|
Wang R, Li DF, Hu YF, Liao Q, Jiang TT, Olatunji OJ, Yang K, Zuo J. Qing-Luo-Yin Alleviated Monocytes/Macrophages-Mediated Inflammation in Rats with Adjuvant-Induced Arthritis by Disrupting Their Interaction with (Pre)-Adipocytes Through PPAR-γ Signaling. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3105-3118. [PMID: 34295151 PMCID: PMC8291661 DOI: 10.2147/dddt.s320599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/09/2021] [Indexed: 12/29/2022]
Abstract
Background The Chinese herbal formula Qing-Luo-Yin (QLY) has been successfully used in rheumatoid arthritis treatment for decades. It exhibits notable immune and metabolism regulatory properties. Thereby, we investigated its effects on the interplay between (pre)-adipocytes and monocytes/macrophages under adjuvant-induced arthritis (AIA) circumstances. Methods Fat reservoir and histological characteristics of white fat tissues (WAT) in AIA rats receiving QLY treatment were examined upon sacrifice. Metabolic parameters, clinical indicators, and oxidative stress levels were determined using corresponding kits, while mRNA/protein expression was investigated by PCR and immunoblotting methods. M1 macrophage distribution in WAT was assessed by flow cytometry. The effects of QLY on (pre)-adipocytes were further validated by experiments in vitro. Results Compared with normal healthy controls, body weight and circulating triglyceride were declined in AIA rats, but serological levels of free fatty acids and low-density lipoprotein cholesterol were increased. mRNA IL-1β and iNOS expression in white blood cells and rheumatoid factor, C-reactive protein, anti-cyclic citrullinated peptide antibody, MCP-1 and IL-1β production in serum/WAT were up-regulated. Obvious CD86+CD11b+ macrophages were enriched in WAT. Meanwhile, expression of PPAR-γ and SIRT1 and secretion of adiponectin and leptin in these AIA rats were impaired. QLY restored all these pathological changes. Of note, it significantly stimulated PPAR-γ expression in the treated AIA rats. Accordingly, QLY-containing serum promoted SCD-1, PPAR-γ, and SIRT1 expression in pre-adipocytes cultured in vitro. AIA rats-derived peripheral blood mononuclear cells suppressed PPAR-γ and SCD-1 expression in co-cultured pre-adipocytes, but serum from AIA rats receiving QLY treatment did not exhibit this potential. The changes on PPAR-γ expression eventually resulted in varied adipocyte differentiation statuses. PPAR-γ selective inhibitor T0070907 abrogated QLY-induced MCP-1 production decline in LPS-primed pre-adipocytes and reduced adiponectin secretion. Conclusion QLY was potent in promoting PPAR-γ expression and consequently disrupted inflammatory feedback in WAT by altering monocytes/macrophages polarization and adipocytes differentiation.
Collapse
Affiliation(s)
- Rui Wang
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People's Republic of China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People's Republic of China
| | - Dan-Feng Li
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People's Republic of China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People's Republic of China
| | - Yi-Fang Hu
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People's Republic of China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People's Republic of China
| | - Qiang Liao
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People's Republic of China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People's Republic of China
| | - Tian-Tian Jiang
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People's Republic of China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People's Republic of China
| | - Opeyemi Joshua Olatunji
- Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, 90112, Thailand
| | - Kui Yang
- Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People's Republic of China.,Department of Pharmacy, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People's Republic of China
| | - Jian Zuo
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People's Republic of China.,Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People's Republic of China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241000, People's Republic of China
| |
Collapse
|
15
|
Correlation between TNF-α- serum level and TNF-α -308 gene polymorphism in Iraqi patients with type 2 diabetes mellitus. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
16
|
Chinniah R, Sevak V, Pandi S, Ravi PM, Vijayan M, Kannan A, Karuppiah B. HLA-DRB1 genes and the expression dynamics of HLA CIITA determine the susceptibility to T2DM. Immunogenetics 2021; 73:291-305. [PMID: 33754173 DOI: 10.1007/s00251-021-01212-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/01/2021] [Indexed: 12/18/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a disease with polygenic inheritance. The expression of major histocompatibility complex class II genes are regulated by several trans-activators. We have studied the expression of HLA-DRB1, RFX, CIITA-P1, PIV transactivators, immunophenotyping of cells, SNPs in CIITA-168 (A/G) and IFN-γ + 874 (T/A) in T2DM patients and controls (n = 201 each). We observed increased frequencies of DRB1*03, DRB1*04 and DRB1*07 and decreased frequencies of DRB1*10, DRB1*14, and DRB1*15 alleles among patients. Significant up-regulations of HLA-DRB1 genes were observed in patients (p < 0.0001). Down-regulated expressions were documented in DRB1*03-homo (p < 0.002) and DRB1*04-homo (p < 0.009) patients. No significant differences were observed for CIITA-P1 expression except DRB1*04-pooled (p < 0.0113). The CIITA-PIV was up-regulated in overall (p < 0.0001), DRB1*03-pooled (p < 0.0006), DRB1*03-hetero (p < 0.0006) and DRB1*03-homo (p < 0.001) T2DM patients. However, significant down-regulations were documented for DRB1*04-pooled (p < 0.040), DRB1*04-hetero (p < 0.060), and DRB1*04-homo (p < 0.027) combinations. Further, significant down-regulations of RFX5 were observed in overall (p < 0.0006), DRB1*04-pooled (p < 0.0022), and DRB1*04-hetero (p < 0.0004) combinations. Immunophenotyping studies revealed significant increase of CD45+ CD14-, CD19+, CD14- and CD8 cells and elevated level of expression of IFN-γ (p < 0.0001) in patients. A significant increase of TT (p < 3.35 × 10-6) and decrease of TA (p < 4.57 × 10-4) genotypes of IFN-γ + 874 (T/A) and an increase of GG (p < 0.001) and decrease of AG (p < 8.24 × 10-5) genotypes of CIITA-168 A/G SNPs were observed. The combinatorial analysis revealed susceptible associations for DRB1*03 + AA, *03 + AG, *03 + GG and *04 + GG and protective associations for DRB1*10 + AG, *10 + GG, *15 + AG, and *14 + GG combinations. Thus, the present study corroborated the effect of differential expressions of promoters of risk alleles in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Rathika Chinniah
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India
| | - Vandit Sevak
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India
| | - Sasiharan Pandi
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India
| | - Padma Malini Ravi
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Arun Kannan
- The Madurai Institute of Diabetes and Endocrine Practice Research, Madurai, Tamil Nadu, 625 001, India
| | - Balakrishnan Karuppiah
- Department of Immunology, School of Biological Sciences, Madurai, Tamil Nadu, 625021, India.
| |
Collapse
|
17
|
Yu J, Sun H, Zhu J, Wei X, Shi H, Shen B, Ren L, He Y, Zhang R, Zhang M, Peng H. Asymptomatic Hyperuricemia and Metabolically Unhealthy Obesity: A Cross-Sectional Analysis in the Tianning Cohort. Diabetes Metab Syndr Obes 2021; 14:1367-1374. [PMID: 33790604 PMCID: PMC8006809 DOI: 10.2147/dmso.s301363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE The relationship between obesity and hyperuricemia has been demonstrated by many studies. However, whether or to what extent metabolic condition influents the association between obesity and hyperuricemia was not clear. Here, we aimed to examine the association between obese-metabolic phenotype and hyperuricemia in a large sample of Chinese adults. METHODS According to BMI and metabolic syndrome, obese-metabolic phenotype was defined as metabolically unhealthy obesity (MUO), metabolically healthy obesity (MHO), metabolically unhealthy non-obesity (MUNO) and metabolically healthy non-obesity (MHNO)in the Tianning cohort (N=5072). We conducted a cross-sectional analysis between obese-metabolic phenotype and hyperuricemia, followed by a Mendelian Randomization analysis using GWAS summary data to confirm the causality between uric acid and BMI. RESULTS The average level of serum UA showed 41.87-higher μmol/L in participants with MHO (β=41.87, P<0.001) and 63.18-higher μmol/L in participants with MUO (β=63.18, P<0.001), compared to those with MHNO. Compared to participants with MHNO, those with MUO had the highest likelihood to have hyperuricemia (OR=4.56, P<0.001), followed by those with MHO (OR=3.32, P<0.001). Mendelian randomization analysis indicated that uric acid was more likely to be a consequence of BMI (β=0.059, P=6.54×10-154). CONCLUSION MUO, in comparison with MHO, was significantly associated with hyperuricemia in Chinese adults.
Collapse
Affiliation(s)
- Jia Yu
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Hongyan Sun
- Center for Disease Prevention and Control of Tianning District, Changzhou, People’s Republic of China
| | - Jinhua Zhu
- Center for Disease Prevention and Control of Wujiang District, Suzhou, People’s Republic of China
| | - Xintong Wei
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Hongfei Shi
- Center for Disease Prevention and Control of Tianning District, Changzhou, People’s Republic of China
| | - Bin Shen
- Center for Disease Prevention and Control of Wujiang District, Suzhou, People’s Republic of China
| | - Liyun Ren
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Yan He
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Rongyan Zhang
- Center for Disease Prevention and Control of Wujiang District, Suzhou, People’s Republic of China
| | - Mingzhi Zhang
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Hao Peng
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, People’s Republic of China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, People’s Republic of China
- Correspondence: Hao Peng Department of Epidemiology, School of Public Health, Medical College of Soochow University, 199 Renai Road, Industrial Park, Suzhou, 215123, People’s Republic of ChinaTel +86 512 6588 0078Fax +86 512 6588 0052 Email
| |
Collapse
|
18
|
Neutrophils and Macrophages as Targets for Development of Nanotherapeutics in Inflammatory Diseases. Pharmaceutics 2020; 12:pharmaceutics12121222. [PMID: 33348630 PMCID: PMC7766591 DOI: 10.3390/pharmaceutics12121222] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 11/27/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Neutrophils and macrophages are major components of innate systems, playing central roles in inflammation responses to infections and tissue injury. If they are out of control, inflammation responses can cause the pathogenesis of a wide range of diseases, such as inflammatory disorders and autoimmune diseases. Precisely regulating the functions of neutrophils and macrophages in vivo is a potential strategy to develop immunotherapies to treat inflammatory diseases. Advances in nanotechnology have enabled us to design nanoparticles capable of targeting neutrophils or macrophages in vivo. This review discusses the current status of how nanoparticles specifically target neutrophils or macrophages and how they manipulate leukocyte functions to inhibit their activation for inflammation resolution or to restore their defense ability for pathogen clearance. Finally, we present a novel concept of hijacking leukocytes to deliver nanotherapeutics across the blood vessel barrier. This review highlights the challenges and opportunities in developing nanotherapeutics to target leukocytes for improved treatment of inflammatory diseases.
Collapse
|
19
|
Effects of the Glutamine Administration on T Helper Cell Regulation and Inflammatory Response in Obese Mice Complicated with Polymicrobial Sepsis. Mediators Inflamm 2020; 2020:8869017. [PMID: 33223959 PMCID: PMC7671796 DOI: 10.1155/2020/8869017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
This study investigated the impacts of GLN on inflammation and T cell dysregulation in obese mice complicated with sepsis. Mice were divided into normal control (NC) and high-fat diet groups. The high-fat diet provided 60% of energy from fat and was administered for 10 weeks to induce obesity. Mice fed with a high-fat diet were then assigned to sham (SH) and sepsis with saline (SS) or GLN (SG) groups. The SH group was subjected to laparotomy, while the sepsis group underwent cecal ligation and puncture (CLP). The SS group was intravenously injected with saline. The SG group was intravenously administered GLN after CLP. Mice were sacrificed at 12, 24, or 48 h post-CLP, respectively. Results demonstrated that in the presence of obesity, sepsis drove CD4+ T cells toward the helper T (Th)2 and Th17 lineages. Also, expressions of inflammatory cytokines and macrophage infiltration markers in adipose tissues and lungs were elevated. Treatment of obese mice with GLN after sepsis reversed Th polarization and downregulated macrophage infiltration and inflammatory cytokine, whereas the tight junction-associated protein expression increased in the lungs. These findings suggest that the intravenous administration of GLN to obese mice after sepsis modulated a more balanced Th cell lineage, alleviated inflammation, and attenuated lung injury.
Collapse
|
20
|
Fischer AW, Behrens J, Sass F, Schlein C, Heine M, Pertzborn P, Scheja L, Heeren J. Brown adipose tissue lipoprotein and glucose disposal is not determined by thermogenesis in uncoupling protein 1-deficient mice. J Lipid Res 2020; 61:1377-1389. [PMID: 32769145 DOI: 10.1194/jlr.ra119000455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Adaptive thermogenesis is highly dependent on uncoupling protein 1 (UCP1), a protein expressed by thermogenic adipocytes present in brown adipose tissue (BAT) and white adipose tissue (WAT). Thermogenic capacity of human and mouse BAT can be measured by positron emission tomography-computed tomography quantifying the uptake of 18F-fluodeoxyglucose or lipid tracers. BAT activation is typically studied in response to cold exposure or treatment with β-3-adrenergic receptor agonists such as CL316,243 (CL). Currently, it is unknown whether cold-stimulated uptake of glucose or lipid tracers is a good surrogate marker of UCP1-mediated thermogenesis. In metabolic studies using radiolabeled tracers, we found that glucose uptake is increased in mildly cold-activated BAT of Ucp1 -/- versus WT mice kept at subthermoneutral temperature. Conversely, lower glucose disposal was detected after full thermogenic activation achieved by sustained cold exposure or CL treatment. In contrast, uptake of lipoprotein-derived fatty acids into chronically activated thermogenic adipose tissues was substantially increased in UCP1-deficient mice. This effect is linked to higher sympathetic tone in adipose tissues of Ucp1 -/- mice, as indicated by elevated levels of thermogenic genes in BAT and WAT. Thus, glucose and lipoprotein handling does not necessarily reflect UCP1-dependent thermogenic activity, but especially lipid uptake rather mirrors sympathetic activation of adipose tissues.
Collapse
Affiliation(s)
- Alexander W Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janina Behrens
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederike Sass
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Schlein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Pertzborn
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
Lecoutre S, Kwok KHM, Petrus P, Lambert M, Breton C. Epigenetic Programming of Adipose Tissue in the Progeny of Obese Dams. Curr Genomics 2020; 20:428-437. [PMID: 32477000 PMCID: PMC7235387 DOI: 10.2174/1389202920666191118092852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 01/13/2023] Open
Abstract
According to the Developmental Origin of Health and Disease (DOHaD) concept, maternal obesity and the resulting accelerated growth in neonates predispose offspring to obesity and associated metabolic diseases that may persist across generations. In this context, the adipose tissue has emerged as an important player due to its involvement in metabolic health, and its high potential for plasticity and adaptation to environmental cues. Recent years have seen a growing interest in how maternal obesity induces long-lasting adipose tissue remodeling in offspring and how these modifications could be transmitted to subsequent generations in an inter- or transgenerational manner. In particular, epigenetic mechanisms are thought to be key players in the developmental programming of adipose tissue, which may partially mediate parts of the transgenerational inheritance of obesity. This review presents data supporting the role of maternal obesity in the developmental programming of adipose tissue through epigenetic mechanisms. Inter- and transgenerational effects on adipose tissue expansion are also discussed in this review.
Collapse
Affiliation(s)
- Simon Lecoutre
- University of Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F-59000 Lille, France.,Department of Medicine (H7), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Kelvin H M Kwok
- Department of Biosciences and Nutrition, Karolinska Insitutet, 141 86 Stockholm, Sweden
| | - Paul Petrus
- Department of Medicine (H7), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Mélanie Lambert
- Department of Medicine (H7), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Christophe Breton
- University of Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, F-59000 Lille, France
| |
Collapse
|
22
|
Petrus P, Lecoutre S, Dollet L, Wiel C, Sulen A, Gao H, Tavira B, Laurencikiene J, Rooyackers O, Checa A, Douagi I, Wheelock CE, Arner P, McCarthy M, Bergo MO, Edgar L, Choudhury RP, Aouadi M, Krook A, Rydén M. Glutamine Links Obesity to Inflammation in Human White Adipose Tissue. Cell Metab 2020; 31:375-390.e11. [PMID: 31866443 DOI: 10.1016/j.cmet.2019.11.019] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 08/27/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
Abstract
While obesity and associated metabolic complications are linked to inflammation of white adipose tissue (WAT), the causal factors remain unclear. We hypothesized that the local metabolic environment could be an important determinant. To this end, we compared metabolites released from WAT of 81 obese and non-obese women. This identified glutamine to be downregulated in obesity and inversely associated with a pernicious WAT phenotype. Glutamine administration in vitro and in vivo attenuated both pro-inflammatory gene and protein levels in adipocytes and WAT and macrophage infiltration in WAT. Metabolomic and bioenergetic analyses in human adipocytes suggested that glutamine attenuated glycolysis and reduced uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) levels. UDP-GlcNAc is the substrate for the post-translational modification O-linked β-N-acetylglucosamine (O-GlcNAc) mediated by the enzyme O-GlcNAc transferase. Functional studies in human adipocytes established a mechanistic link between reduced glutamine, O-GlcNAcylation of nuclear proteins, and a pro-inflammatory transcriptional response. Altogether, glutamine metabolism is linked to WAT inflammation in obesity.
Collapse
Affiliation(s)
- Paul Petrus
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden
| | - Simon Lecoutre
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden
| | - Lucile Dollet
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Clotilde Wiel
- Department of Biosciences and Nutrition (BioNut), H2, Karolinska Institutet, Stockholm 141 86, Sweden
| | - André Sulen
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Hui Gao
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden
| | - Beatriz Tavira
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden
| | - Jurga Laurencikiene
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden
| | - Olav Rooyackers
- Perioperative Medicine and Intensive Care, B31, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Antonio Checa
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Iyadh Douagi
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden
| | - Craig E Wheelock
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Peter Arner
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden
| | - Mark McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK; Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Martin O Bergo
- Department of Biosciences and Nutrition (BioNut), H2, Karolinska Institutet, Stockholm 141 86, Sweden
| | - Laurienne Edgar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Myriam Aouadi
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Stockholm 141 86, Sweden.
| |
Collapse
|
23
|
Iacomino G, Lauria F, Venezia A, Iannaccone N, Russo P, Siani A. microRNAs in Obesity and Metabolic Diseases. OBESITY AND DIABETES 2020:71-95. [DOI: 10.1007/978-3-030-53370-0_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
24
|
Yu C, Niu X, Du Y, Chen Y, Liu X, Xu L, Iwakura Y, Ma X, Li Y, Yao Z, Deng W. IL-17A promotes fatty acid uptake through the IL-17A/IL-17RA/p-STAT3/FABP4 axis to fuel ovarian cancer growth in an adipocyte-rich microenvironment. Cancer Immunol Immunother 2019; 69:115-126. [PMID: 31802182 DOI: 10.1007/s00262-019-02445-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 11/27/2019] [Indexed: 01/13/2023]
Abstract
Pro-inflammatory cytokines are crucial mediators of cancer development, representing potential targets for cancer therapy. The molecular mechanism of a vital pro-inflammatory cytokine, IL-17A, in cancer progression and its potential use in therapy through influencing fatty acid (FA) metabolism, especially FA uptake of cancer cells, remains unknown. In the present study, we used IL-17A and ovarian cancer (OvCa), a representative of both obesity-related and inflammation-related cancers, to explore the interactions among IL-17A, cancer cells and adipocytes (which can provide FAs). We found that in the presence of palmitic acid (PA), IL-17A could directly increase the cellular uptake of PA, leading to the proliferation of OvCa cells via the IL-17A/IL-17RA/p-STAT3/FABP4 axis rather than via CD36. Moreover, in vivo experiments using an orthotopic implantation model in IL-17A-deficient mice demonstrated that endogenous IL-17A could fuel OvCa growth and metastasis with increased expression of FABP4 and p-STAT3. Furthermore, analysis of clinical specimens supported the above findings. Our data not only provide useful insights into the clinical intervention of the growth and metastasis of the tumors (such as OvCa) that are prone to growth and metastasis in an adipocyte-rich microenvironment (ARM) but also provides new insights into the roles of IL-17A in tumor progression and immunomodulatory therapy of OvCa.
Collapse
Affiliation(s)
- Chunyan Yu
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Xiulong Niu
- Department of Prevention and Therapy of Skin Disease in the Security Environment, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, 300162, China
| | - Yongrui Du
- Department of Reproductive Medicine, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
| | - Yan Chen
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Xiaomei Liu
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Lingling Xu
- Office of the Hospital Ethics Committee, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, 125-8585, Japan
| | - Xiaoxia Ma
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Yan Li
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Zhi Yao
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Weimin Deng
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Diseases and Microenvironment of Ministry of Education of China, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China.
| |
Collapse
|
25
|
Chittezhath M, Gunaseelan D, Zheng X, Hasan R, Tay VSY, Lim ST, Wang X, Berggren PO, Bornstein S, Boehm B, Ruedl C, Ali Y. Islet macrophages are associated with islet vascular remodeling and compensatory hyperinsulinemia during diabetes. Am J Physiol Endocrinol Metab 2019; 317:E1108-E1120. [PMID: 31573842 DOI: 10.1152/ajpendo.00248.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
β-Cells respond to peripheral insulin resistance by first increasing circulating insulin during diabetes. Islet remodeling supports this compensation, but its drivers remain poorly understood. Infiltrating macrophages have been implicated in late-stage type 2 diabetes, but relatively little is known on islet resident macrophages, especially during compensatory hyperinsulinemia. We hypothesized that islet resident macrophages would contribute to islet vascular remodeling and hyperinsulinemia during diabetes, the failure of which results in a rapid progression to frank diabetes. We used chemical (clodronate), genetics (CD169-diphtheria toxin receptor mice), or antibody-mediated (colony-stimulating factor 1 receptor α) macrophage ablation methods in diabetic (db/db) and diet-induced models of compensatory hyperinsulinemia to investigate the role of macrophages in islet remodeling. We transplanted islets devoid of macrophages into naïve diabetic mice and assessed the impact on islet vascularization. With the use of the above methods, we showed that macrophage depletion significantly and consistently compromised islet remodeling in terms of size, vascular density, and insulin secretion capacity. Depletion of islet macrophages reduced VEGF-A secretion in both human and mouse islets ex vivo, and this functionally translated to delayed revascularization upon transplantation in vivo. We revealed that islet resident macrophages were associated with islet remodeling and increased insulin secretion during diabetes. This suggests utility in harnessing islet macrophages during this phase to promote islet vascularization, remodeling, and insulin secretion.
Collapse
Affiliation(s)
- Manesh Chittezhath
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Divya Gunaseelan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Xiaofeng Zheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, Singapore
| | - Riasat Hasan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Vanessa S Y Tay
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Seok Ting Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Xiaomeng Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, Singapore
| | - Per-Olof Berggren
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, Singapore
- Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Bornstein
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- Department of Medicine, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | - Bernhard Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, Singapore
| |
Collapse
|
26
|
Abstract
Invariant natural killer T cells (iNKT cells) are an innate-like T cell subset that expresses an invariant T cell receptor (TCR) α-chain and recognizes lipids presented on CD1d. They secrete diverse cytokines and can influence many types of immune responses. Despite having highly similar TCR specificities, iNKT cells differentiate in the thymus into distinct subsets that are analogous to T helper 1 (TH1), TH2 and TH17 cell subsets. Additional iNKT cell subsets that may require peripheral activation have also been described, including one that produces IL-10. In general, iNKT cells are non-circulating, tissue-resident lymphocytes, but the prevalence of different iNKT cell subsets differs markedly between tissues. Here, we summarize the functions of iNKT cells in four tissues in which they are prevalent, namely, the liver, the lungs, adipose tissue and the intestine. Importantly, we explain how local iNKT cell responses at each site contribute to tissue homeostasis and protection from infection but can also contribute to tissue inflammation and damage.
Collapse
|
27
|
Patel R, Palit SP, Rathwa N, Ramachandran A, Begum R. Genetic variants of tumor necrosis factor-α and its levels: A correlation with dyslipidemia and type 2 diabetes susceptibility. Clin Nutr 2019; 38:1414-1422. [DOI: 10.1016/j.clnu.2018.06.962] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/04/2018] [Accepted: 06/13/2018] [Indexed: 12/26/2022]
|
28
|
Rozengurt E, Eibl G. Central role of Yes-associated protein and WW-domain-containing transcriptional co-activator with PDZ-binding motif in pancreatic cancer development. World J Gastroenterol 2019; 25:1797-1816. [PMID: 31057295 PMCID: PMC6478619 DOI: 10.3748/wjg.v25.i15.1797] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a deadly disease with no efficacious treatment options. PDAC incidence is projected to increase, which may be caused at least partially by the obesity epidemic. Significantly enhanced efforts to prevent or intercept this cancer are clearly warranted. Oncogenic KRAS mutations are recognized initiating events in PDAC development, however, they are not entirely sufficient for the development of fully invasive PDAC. Additional genetic alterations and/or environmental, nutritional, and metabolic signals, as present in obesity, type-2 diabetes mellitus, and inflammation, are required for full PDAC formation. We hypothesize that oncogenic KRAS increases the intensity and duration of the growth-promoting signaling network. Recent exciting studies from different laboratories indicate that the activity of the transcriptional co-activators Yes-associated protein (YAP) and WW-domain-containing transcriptional co-activator with PDZ-binding motif (TAZ) play a critical role in the promotion and maintenance of PDAC operating as key downstream target of KRAS signaling. While initially thought to be primarily an effector of the tumor-suppressive Hippo pathway, more recent studies revealed that YAP/TAZ subcellular localization and co-transcriptional activity is regulated by multiple upstream signals. Overall, YAP has emerged as a central node of transcriptional convergence in growth-promoting signaling in PDAC cells. Indeed, YAP expression is an independent unfavorable prognostic marker for overall survival of PDAC. In what follows, we will review studies implicating YAP/TAZ in pancreatic cancer development and consider different approaches to target these transcriptional regulators.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, United States
- CURE: Digestive Diseases Research Center, Los Angeles, CA 90095, United States
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, United States
- CURE: Digestive Diseases Research Center, Los Angeles, CA 90095, United States
| |
Collapse
|
29
|
Xie P, Zhao C, Huang W, Yong T, Chung ACK, He K, Chen X, Cai Z. Prenatal exposure to ambient fine particulate matter induces dysregulations of lipid metabolism in adipose tissue in male offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 657:1389-1397. [PMID: 30677905 DOI: 10.1016/j.scitotenv.2018.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/29/2018] [Accepted: 12/02/2018] [Indexed: 06/09/2023]
Abstract
Prenatal exposure to ambient fine particles (diameter < 0.25 μm, PM2.5) has been found to be associated with abnormal growth and development in offspring. However, the effects of PM2.5 on the lipid metabolism of adipose tissue in offspring are unclear. In the present study, we established a mouse model of prenatal exposure to PM2.5 by intratracheal instillation to pregnant C57BL/6 female mice with PM2.5 suspension or normal saline. We found that prenatal exposure to PM2.5 of a mouse model reduced body weight in adult male offspring after 6 weeks old. Histological analysis showed that the adipocyte size was significantly reduced in epididymal adipose tissue (eWAT) in male offspring, but not in brown adipose tissue. The expression levels of genes related to fatty acid synthesis (ACC1, ACSL1) and oxidation (PPARα) in eWAT were also significantly decreased. In addition, downregulation of pro-inflammatory cytokines (TNFα, IL-1β, IL-6) was also observed. Lipidomics analysis of eWAT demonstrated that prenatal exposure of PM2.5 reduced lysophosphatidylcholines (LPC), phosphatidylcholines (PC), phosphatidylethanolamines (PE), sphingomyelins (SM), and ceramides (Cer), indicating that metabolic pathways, including SM-Cer signaling and glycerophospholipids remodeling, were disrupted. In summary, prenatal exposure to PM2.5 was associated with the dysregulations in lipid metabolism of eWAT and pro-inflammatory response in male offspring.
Collapse
Affiliation(s)
- Peisi Xie
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Chao Zhao
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Wei Huang
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Ting Yong
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Arthur C K Chung
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China; HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Kaiwu He
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xiangfeng Chen
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China; HKBU Institute for Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
30
|
Deletion of translin (Tsn) induces robust adiposity and hepatic steatosis without impairing glucose tolerance. Int J Obes (Lond) 2019; 44:254-266. [PMID: 30647452 PMCID: PMC6629527 DOI: 10.1038/s41366-018-0315-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/06/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023]
Abstract
Objective: Translin knockout (KO) mice display robust adiposity. Recent studies indicate that translin and its partner protein, trax, regulate the microRNA and ATM kinase signaling pathways, both of which have been implicated in regulating metabolism. In the course of characterizing the metabolic profile of these mice, we found that they display normal glucose tolerance despite their elevated adiposity. Accordingly, we investigated why translin KO mice display this paradoxical phenotype. Methods: To help distinguish between the metabolic effects of increased adiposity and those of translin deletion per se, we compared three groups: (1) wild-type (WT), (2) translin KO mice on a standard chow diet, and (3) adiposity-matched WT mice that were placed on a high-fat diet until they matched translin KO adiposity levels. All groups were scanned to determine their body composition and tested to evaluate their glucose and insulin tolerance. Plasma, hepatic and adipose tissue samples were collected and used for histological and molecular analyses. Results: Translin KO mice show normal glucose tolerance whereas adiposity-matched WT mice, placed on a high-fat diet, do not. In addition, translin KO mice display prominent hepatic steatosis that is more severe than that of adiposity-matched WT mice. Unlike adiposity-matched WT mice, translin KO mice display three key features that have been shown to reduce susceptibility to insulin resistance: increased accumulation of subcutaneous fat, increased levels of circulating adiponectin and decreased Tnfα expression in hepatic and adipose tissue. Conclusions: The ability of translin KO mice to retain normal glucose tolerance in the face of marked adipose tissue expansion may be due to the three protective factors noted above. Further studies aimed at defining the molecular bases for this combination of protective phenotypes may yield new approaches to limit the adverse metabolic consequences of obesity.
Collapse
|
31
|
Ayelign B, Genetu M, Wondmagegn T, Adane G, Negash M, Berhane N. TNF-α (-308) Gene Polymorphism and Type 2 Diabetes Mellitus in Ethiopian Diabetes Patients. Diabetes Metab Syndr Obes 2019; 12:2453-2459. [PMID: 31819571 PMCID: PMC6886533 DOI: 10.2147/dmso.s229987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/21/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a metabolic disorder resulting from insulin insufficiency or function. Predisposing factors for T2DM are mainly genetic and environmental. Genetic polymorphism of cytokines like tumor necrosis factor-alpha (TNF-α) is suggestive of interference with insulin-sensitive glucose uptake and induces insulin resistance that ultimately could lead to T2DM. In this study, we assessed the effect of TNF-α (-308) G/A gene polymorphism and its association with the development of T2DM in an Ethiopian population. METHODS An institutional-based cross-sectional study was conducted on study subjects with T2DM and non-diabetic healthy controls. DNA was extracted and genotyping was carried out by using amplification refractory mutation system polymerase chain reaction. A genetic-polymorphism on TNF-α (-308) G/A with T2DM was evaluated by logistic regression and Student's t-test. A P-value <0.05 was considered as statistically significant. RESULTS In the present study, we observed a significant association between T2DM and TNF-α (-308) gene polymorphism's GG genotype [χ2 test P = 0.005, OR (95% CI) =2.667 (1.309-5.45d8)]. In contrast, no statistically significant differences were observed in the frequencies of genotypes AA and AG (χ2 test P=0.132 and 0.067, respectively). Moreover, T2DM individuals had higher concentrations of lipid profiles for those carrying the TNF-α (-308) GG genotype as compared to the control group. CONCLUSION TNF-α (-308) genetic polymorphism may be implicated in the genetic susceptibility for, as well as the development of T2DM and lipid metabolism in the Ethiopian population. Therefore, a large-scale study and early screening of TNF-α (-308) genetic polymorphism may help in early management and control of diabetes and related outcomes.
Collapse
Affiliation(s)
- Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopa
- Correspondence: Birhanu Ayelign Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopa Email
| | - Meaza Genetu
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopa
| | - Tadelo Wondmagegn
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopa
| | - Gashaw Adane
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopa
| | - Markos Negash
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopa
| | - Nega Berhane
- Institute of Biotechnology, College of Computational and Natural Science, University of Gondar, Gondar, Ethiopa
| |
Collapse
|
32
|
Burhans MS, Hagman DK, Kuzma JN, Schmidt KA, Kratz M. Contribution of Adipose Tissue Inflammation to the Development of Type 2 Diabetes Mellitus. Compr Physiol 2018; 9:1-58. [PMID: 30549014 DOI: 10.1002/cphy.c170040] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The objective of this comprehensive review is to summarize and discuss the available evidence of how adipose tissue inflammation affects insulin sensitivity and glucose tolerance. Low-grade, chronic adipose tissue inflammation is characterized by infiltration of macrophages and other immune cell populations into adipose tissue, and a shift toward more proinflammatory subtypes of leukocytes. The infiltration of proinflammatory cells in adipose tissue is associated with an increased production of key chemokines such as C-C motif chemokine ligand 2, proinflammatory cytokines including tumor necrosis factor α and interleukins 1β and 6 as well as reduced expression of the key insulin-sensitizing adipokine, adiponectin. In both rodent models and humans, adipose tissue inflammation is consistently associated with excess fat mass and insulin resistance. In humans, associations with insulin resistance are stronger and more consistent for inflammation in visceral as opposed to subcutaneous fat. Further, genetic alterations in mouse models of obesity that reduce adipose tissue inflammation are-almost without exception-associated with improved insulin sensitivity. However, a dissociation between adipose tissue inflammation and insulin resistance can be observed in very few rodent models of obesity as well as in humans following bariatric surgery- or low-calorie-diet-induced weight loss, illustrating that the etiology of insulin resistance is multifactorial. Taken together, adipose tissue inflammation is a key factor in the development of insulin resistance and type 2 diabetes in obesity, along with other factors that likely include inflammation and fat accumulation in other metabolically active tissues. © 2019 American Physiological Society. Compr Physiol 9:1-58, 2019.
Collapse
Affiliation(s)
- Maggie S Burhans
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Derek K Hagman
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jessica N Kuzma
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kelsey A Schmidt
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Mario Kratz
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
33
|
Tans R, Verschuren L, Wessels HJCT, Bakker SJL, Tack CJ, Gloerich J, van Gool AJ. The future of protein biomarker research in type 2 diabetes mellitus. Expert Rev Proteomics 2018; 16:105-115. [DOI: 10.1080/14789450.2018.1551134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Roel Tans
- Translational Metabolic Laboratory, Department of Laboratory Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, TNO, Zeist, The Netherlands
| | - Hans J. C. T. Wessels
- Translational Metabolic Laboratory, Department of Laboratory Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stephan J. L. Bakker
- Department of Internal Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Cees J. Tack
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jolein Gloerich
- Translational Metabolic Laboratory, Department of Laboratory Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alain J. van Gool
- Translational Metabolic Laboratory, Department of Laboratory Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
34
|
LaMarche NM, Kohlgruber AC, Brenner MB. Innate T Cells Govern Adipose Tissue Biology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:1827-1834. [PMID: 30224362 PMCID: PMC6201318 DOI: 10.4049/jimmunol.1800556] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023]
Abstract
During the past 25 y, the immune system has appeared as a key regulator of adipose tissue biology and metabolic homeostasis. In lean animals, adipose-resident leukocytes maintain an anti-inflammatory microenvironment that preserves the proper functioning of the tissue. In this review, we describe two populations of innate T cells enriched in adipose tissue, invariant NKT and γδ T cells, and how they serve overlapping and nonredundant roles in controlling adipose tissue functions. These cells interact with and expand anti-inflammatory regulatory T cells and M2 macrophages, thereby driving a metabolically beneficial tissue milieu. Surprisingly, we have found that adipose invariant NKT and γδ T cells also promote weight loss and heat production in a process called "nonshivering thermogenesis." The data surrounding these two cell types highlight their powerful ability to regulate not only other leukocytes, but also tissue-wide processes that affect an entire organism.
Collapse
Affiliation(s)
- Nelson M LaMarche
- Division of Rheumatology, Immunology, and Allergy, Harvard Medical School, Boston, MA 02115; and
| | - Ayano C Kohlgruber
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Michael B Brenner
- Division of Rheumatology, Immunology, and Allergy, Harvard Medical School, Boston, MA 02115; and
| |
Collapse
|
35
|
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
36
|
Luteolin-Enriched Artichoke Leaf Extract Alleviates the Metabolic Syndrome in Mice with High-Fat Diet-Induced Obesity. Nutrients 2018; 10:nu10080979. [PMID: 30060507 PMCID: PMC6115887 DOI: 10.3390/nu10080979] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 12/19/2022] Open
Abstract
This current study aimed to elucidate the effects and possible underlying mechanisms of long-term supplementation with dietary luteolin (LU)-enriched artichoke leaf (AR) in high-fat diet (HFD)-induced obesity and its complications (e.g., dyslipidemia, insulin resistance, and non-alcoholic fatty liver disease) in C57BL/6N mice. The mice were fed a normal diet, an HFD, or an HFD plus AR or LU for 16 weeks. In the HFD-fed mice, AR decreased the adiposity and dyslipidemia by decreasing lipogenesis while increasing fatty acid oxidation, which contributed to better hepatic steatosis. LU also prevented adiposity and hepatic steatosis by suppressing lipogenesis while increasing biliary sterol excretion. Moreover, AR and LU prevented insulin sensitivity by decreasing the level of plasma gastric inhibitory polypeptide and activity of hepatic glucogenic enzymes, which may be linked to the lowering of inflammation as evidenced by the reduced plasma interleukin (IL)-6, IL-1β, and plasminogen activator inhibitor-1 levels. Although the anti-metabolic syndrome effects of AR and LU were similar, the anti-adiposity and anti-dyslipidemic effects of AR were more pronounced. These results in mice with diet-induced obesity suggest that long-term supplementation with AR can prevent adiposity and related metabolic disorders such as dyslipidemia, hepatic steatosis, insulin resistance, and inflammation.
Collapse
|
37
|
Chen M, Liang S, Qin X, Zhang L, Qiu L, Chen S, Hu Z, Xu Y, Wang W, Zhang Y, Cao Q, Ying Z. Prenatal exposure to diesel exhaust PM 2.5 causes offspring β cell dysfunction in adulthood. Am J Physiol Endocrinol Metab 2018; 315:E72-E80. [PMID: 29351483 PMCID: PMC6087722 DOI: 10.1152/ajpendo.00336.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Environmental stressors that encounter in early-life and cause abnormal fetal and/or neonatal development may increase susceptibility to non-communicable diseases such as diabetes. Maternal exposure to ambient fine particulate matter (PM2.5) is associated with various fetal abnormalities, suggesting that it may program offspring's susceptibility to diabetes. In the present study, we therefore examined whether maternal exposure to diesel exhaust PM2.5 (DEP), one of the major sources of ambient PM2.5 in urban areas, programs adult offspring's glucose metabolism. Female C57Bl/6J mice were intratracheally instilled with DEP or vehicle throughout a 7-wk preconceptional period, gestation, and lactation, and the glucose homeostasis of their adult male offspring was assessed. Intraperitoneal glucose tolerance test (IPGTT) revealed that the maternal exposure to DEP significantly impaired adult male offspring's glucose tolerance. Unexpectedly, it did not influence their insulin sensitivity, whereas it significantly decreased their glucose-induced insulin secretion (GIIS). This deficit in insulin secretion was corroborated by their significant decrease in arginine-induced insulin secretion. Histological analysis demonstrated that the deficit in insulin secretion was accompanied by the decrease in pancreatic islet and β cell sizes. To differentiate the effects of maternal exposure to DEP before birth and during lactation, some offspring were cross-fostered once born. We did not observe any significant effect of cross-fostering on the glucose homeostasis of adult male offspring and the function and morphology of their β cells. Prenatal exposure to DEP programs the morphology and function of β cells and thus homeostatic regulation of glucose metabolism in adult male offspring.
Collapse
Affiliation(s)
- Minjie Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine , Baltimore, Maryland
- Department of Environmental Health, School of Public Health, Fudan University , Shanghai , China
| | - Shuai Liang
- Department of Medicine Cardiology Division, University of Maryland School of Medicine , Baltimore, Maryland
- Department of Bile Pancreatic Surgery, Xiangya Hospital, Central South University , Changsha, Hunan , China
| | - Xiaobo Qin
- Department of Medicine Cardiology Division, University of Maryland School of Medicine , Baltimore, Maryland
- Department of Electrocardiography, the People's Hospital of Guangxi Zhuang Autonomous Region , Nanning , China
| | - Li Zhang
- Department of Medicine Cardiology Division, University of Maryland School of Medicine , Baltimore, Maryland
- Department of Pharmacology, Medical College of Wuhan University , Wuhan, Hubei , China
| | - Lianglin Qiu
- Department of Medicine Cardiology Division, University of Maryland School of Medicine , Baltimore, Maryland
- Department of Occupational and Environmental Health, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Sufang Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine , Baltimore, Maryland
- Department of Geriatric Endocrinology, the First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan , China
| | - Ziying Hu
- Department of Medicine Cardiology Division, University of Maryland School of Medicine , Baltimore, Maryland
- Department of Endocrinology, the People's Hospital of Zhengzhou University (Henan Provincial People's Hospital) , Zhengzhou, Henan , China
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University , Shanghai , China
| | - Wanjun Wang
- Department of Environmental Health, School of Public Health, Fudan University , Shanghai , China
| | - Yuhao Zhang
- Department of Neurology, Zhongshan Hospital, Fudan University , Shanghai , China
| | - Qi Cao
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| | - Zhekang Ying
- Department of Medicine Cardiology Division, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
38
|
Macrophage Polarization in Chronic Inflammatory Diseases: Killers or Builders? J Immunol Res 2018. [PMID: 29507865 DOI: 10.1155/2018/8917804]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Macrophages are key cellular components of the innate immunity, acting as the main player in the first-line defence against the pathogens and modulating homeostatic and inflammatory responses. Plasticity is a major feature of macrophages resulting in extreme heterogeneity both in normal and in pathological conditions. Macrophages are not homogenous, and they are generally categorized into two broad but distinct subsets as either classically activated (M1) or alternatively activated (M2). However, macrophages represent a continuum of highly plastic effector cells, resembling a spectrum of diverse phenotype states. Induction of specific macrophage functions is closely related to the surrounding environment that acts as a relevant orchestrator of macrophage functions. This phenomenon, termed polarization, results from cell/cell, cell/molecule interaction, governing macrophage functionality within the hosting tissues. Here, we summarized relevant cellular and molecular mechanisms driving macrophage polarization in "distant" pathological conditions, such as cancer, type 2 diabetes, atherosclerosis, and periodontitis that share macrophage-driven inflammation as a key feature, playing their dual role as killers (M1-like) and/or builders (M2-like). We also dissect the physio/pathological consequences related to macrophage polarization within selected chronic inflammatory diseases, placing polarized macrophages as a relevant hallmark, putative biomarkers, and possible target for prevention/therapy.
Collapse
|
39
|
Macrophage Polarization in Chronic Inflammatory Diseases: Killers or Builders? J Immunol Res 2018; 2018:8917804. [PMID: 29507865 PMCID: PMC5821995 DOI: 10.1155/2018/8917804] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 11/01/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022] Open
Abstract
Macrophages are key cellular components of the innate immunity, acting as the main player in the first-line defence against the pathogens and modulating homeostatic and inflammatory responses. Plasticity is a major feature of macrophages resulting in extreme heterogeneity both in normal and in pathological conditions. Macrophages are not homogenous, and they are generally categorized into two broad but distinct subsets as either classically activated (M1) or alternatively activated (M2). However, macrophages represent a continuum of highly plastic effector cells, resembling a spectrum of diverse phenotype states. Induction of specific macrophage functions is closely related to the surrounding environment that acts as a relevant orchestrator of macrophage functions. This phenomenon, termed polarization, results from cell/cell, cell/molecule interaction, governing macrophage functionality within the hosting tissues. Here, we summarized relevant cellular and molecular mechanisms driving macrophage polarization in “distant” pathological conditions, such as cancer, type 2 diabetes, atherosclerosis, and periodontitis that share macrophage-driven inflammation as a key feature, playing their dual role as killers (M1-like) and/or builders (M2-like). We also dissect the physio/pathological consequences related to macrophage polarization within selected chronic inflammatory diseases, placing polarized macrophages as a relevant hallmark, putative biomarkers, and possible target for prevention/therapy.
Collapse
|
40
|
Human adipocyte differentiation and characterization in a perfusion-based cell culture device. Biomed Microdevices 2018; 19:18. [PMID: 28357654 DOI: 10.1007/s10544-017-0164-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adipocytes have gained significant attention recently, because they are not only functioning as energy storage but also as endocrine cells. Adipocytes secret various signaling molecules, including adiponectin, MCP-1, and IL-6, termed collectively as "adipokines". Adipokines regulate glucose metabolism, thereby play an important role in obesity, diabetes type 2, and other metabolic disorders. Conventionally, to study the secretory function, adipocytes are cultured in vitro in static conditions. However, static culturing condition falls short of mimicking the interstitial fluid flows in living systems. Here, we developed a perfusion device which allows dynamic culture of adipocytes under constant and mild flow using a double-layered fluidic structure. Adipocytes were cultured in the bottom layer while the culture media were constantly flown in the upper layer and perfused through a porous membrane that separate the two chambers. The porous membrane between the two chambers physically separates the cells from the flow stream while maintain a fluidic connection by diffusion. This setting not only provides continuous nutrient supply to adipocytes but also maintains a steady and mild shear stress on the cell membrane. It was found the perfusion-based culture conditions promoted faster growth of primary preadipocytes and stimulated greater adipogenesis compared to static culture condition. Adipocytes cultured under perfusion systems produced more MCP-1 and IL-6, but less adiponectin. When stimulated with TNF-α, adipocytes expressed higher level of MCP-1 and IL-6, but lower level of adiponectin. No significant glucose uptake regulation was observed after treating the adipocytes with insulin in both static and perfusion-based culture. Our results demonstrate that perfusion-base culture has played a role in the adipocyte function particularly the secretion of adipokines. More future studies are required to unveil the mechanisms behind perfusion's impact on adipocytes.
Collapse
|
41
|
Zhang J, Zhang L, Zhang S, Yu Q, Xiong F, Huang K, Wang CY, Yang P. HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose tissue in obesity. Mol Cell Endocrinol 2017; 454:103-111. [PMID: 28619625 DOI: 10.1016/j.mce.2017.06.012] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/17/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022]
Abstract
Obesity has emerged as an imminent global public health concern over the past several decades. It has now become evident that obesity is characterized by the persistent and low-grade inflammation in the adipose tissue, and serves as an independent risk factor for many metabolic disorders such as diabetes and cardiovascular disease. Particularly, adipocytes originated from obese mice and humans likely predominate necrosis upon stressful insults, leading to passive release of cellular contents including the high mobility group box 1 (HMGB1) into the extracellular milieu. Extracellular HMGB1 acts as an innate alarmin to stimulate the activation of resident immune cells in the adipose tissue. Upon activation, those resident immune cells actively secrete additional HMGB1, which in turn activates/recruits additional immune cells, and induces adipocyte death. This review summarizes those novel discoveries in terms of HMGB1 in the initiation and maintenance of chronic inflammatory state in adipose tissue in obesity, and discusses its potential application in clinical settings.
Collapse
Affiliation(s)
- Jing Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Lei Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Shu Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Qilin Yu
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Fei Xiong
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
| | - Ping Yang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
| |
Collapse
|
42
|
Suk S, Kwon GT, Lee E, Jang WJ, Yang H, Kim JH, Thimmegowda NR, Chung MY, Kwon JY, Yang S, Kim JK, Park JHY, Lee KW. Gingerenone A, a polyphenol present in ginger, suppresses obesity and adipose tissue inflammation in high-fat diet-fed mice. Mol Nutr Food Res 2017; 61:10.1002/mnfr.201700139. [PMID: 28556482 PMCID: PMC5947313 DOI: 10.1002/mnfr.201700139] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 12/12/2022]
Abstract
SCOPE Ginger exerts protective effects on obesity and its complications. Our objectives here are to identify bioactive compounds that inhibit adipogenesis and lipid accumulation in vitro, elucidate the anti-obesity effect of gingerenone A (GA) in diet-induced obesity (DIO), and investigate whether GA affects adipose tissue inflammation (ATI). METHODS AND RESULTS Oil red O staining showed that GA had the most potent inhibitory effect on adipogenesis and lipid accumulation in 3T3-L1 cells among ginger components tested at a single concentration (40 μM). Consistent with in vitro data, GA attenuates DIO by reducing fat mass in mice. This was accompanied by a modulation of fatty acid metabolism via activation of AMP-activated protein kinase (AMPK) in vitro and in vivo. Additionally, GA suppressed ATI by inhibiting macrophage recruitment and downregulating pro-inflammatory cytokines. CONCLUSION These results suggest that GA may be used as a potential therapeutic candidate for the treatment of obesity and its complications by suppressing adipose expansion and inflammation.
Collapse
Affiliation(s)
- Sujin Suk
- Major in Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Gyoo Taik Kwon
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea
| | - Eunjung Lee
- Major in Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Woo Jung Jang
- Major in Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Hee Yang
- Major in Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Jong Hun Kim
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - N. R. Thimmegowda
- Chemical Biology Research Center and World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Republic of Korea
| | - Min-Yu Chung
- Major in Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Jung Yeon Kwon
- Program in Molecular Medicine and Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Massachusetts Medical School, Worcester, MA, USA
| | - Seunghee Yang
- Major in Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Jason K. Kim
- Major in Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
- Program in Molecular Medicine and Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jung Han Yoon Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ki Won Lee
- Major in Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Cimini FA, Barchetta I, Porzia A, Mainiero F, Costantino C, Bertoccini L, Ceccarelli V, Morini S, Baroni MG, Lenzi A, Cavallo MG. Circulating IL-8 levels are increased in patients with type 2 diabetes and associated with worse inflammatory and cardiometabolic profile. Acta Diabetol 2017; 54:961-967. [PMID: 28836077 DOI: 10.1007/s00592-017-1039-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/04/2017] [Indexed: 01/20/2023]
Abstract
AIMS Interleukin-8 (IL-8) is a chemokine involved in systemic immunity, macrophages infiltration and activation in adipose tissue and may play a significant role in the pathogenesis of type 2 diabetes (T2D) and atherosclerosis. Aims of this study were to evaluate circulating IL-8 levels in adult patients with T2D in comparison with non-diabetic subjects and to describe clinical and biochemical correlates of IL-8 concentration. METHODS For this cross-sectional study, we enrolled 79 consecutive T2D individuals referring to our diabetes outpatient clinics at Sapienza University of Rome, and 37 sex, age and BMI comparable non-diabetic subjects as a control group. Clinical parameters and medical history were recorded; fasting blood sampling was performed for biochemistry and for measuring serum IL-8, IL-6, TNF-α, CRP, adiponectin and 25(OH)vitamin D [25(OH)D] levels. RESULTS Patients with T2D exhibited significantly higher serum IL-8 levels than non-diabetic subjects (69.27 ± 112.83 vs. 16.03 ± 24.27 pg/mL, p < 0.001). In diabetic patients, increased IL-8 concentration correlated with higher IL-6 (p < 0.001), TNF-α (p = 0.02), FBG (p = 0.035), HbA1c (p = 0.04) and LDL-C (p = 0.04) and with lower adiponectin (p = 0.02) and 25(OH)D (p = 0.003) concentrations. CONCLUSIONS Patients with T2D display a marked elevation of circulating IL-8 levels which identify subjects with worse inflammatory, glycometabolic and lipid profile and lower vitamin D levels. Further studies are warranted for evaluating a possible role of IL-8 as a novel marker for risk stratification in T2D patients.
Collapse
Affiliation(s)
- F A Cimini
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - I Barchetta
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - A Porzia
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - F Mainiero
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - C Costantino
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - L Bertoccini
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - V Ceccarelli
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - S Morini
- Microscopic and Ultrastructural Anatomy (CIR), University Campus Bio-Medico, Rome, Italy
| | - M G Baroni
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - M G Cavallo
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy.
| |
Collapse
|
44
|
Iacomino G, Siani A. Role of microRNAs in obesity and obesity-related diseases. GENES AND NUTRITION 2017; 12:23. [PMID: 28974990 PMCID: PMC5613467 DOI: 10.1186/s12263-017-0577-z] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/12/2017] [Indexed: 12/15/2022]
Abstract
In recent years, the link between regulatory microRNAs (miRNAs) and diseases has been the object of intensive research. miRNAs have emerged as key mediators of metabolic processes, playing crucial roles in maintaining/altering physiological processes, including energy balance and metabolic homeostasis. Altered miRNAs expression has been reported in association with obesity, both in animal and human studies. Dysregulation of miRNAs may affect the status and functions of different tissues and organs, including the adipose tissue, pancreas, liver, and muscle, possibly contributing to metabolic abnormalities associated with obesity and obesity-related diseases. More recently, the discovery of circulating miRNAs easily detectable in plasma and other body fluids has emphasized their potential as both endocrine signaling molecules and disease indicators. In this review, the status of current research on the role of miRNAs in obesity and related metabolic abnormalities is summarized and discussed.
Collapse
Affiliation(s)
- Giuseppe Iacomino
- Institute of Food Sciences, CNR, Via Roma, 64, 83100 Avellino, Italy
| | - Alfonso Siani
- Institute of Food Sciences, CNR, Via Roma, 64, 83100 Avellino, Italy
| |
Collapse
|
45
|
Kammoun HL, Allen TL, Henstridge DC, Kraakman MJ, Peijs L, Rose-John S, Febbraio MA. Over-expressing the soluble gp130-Fc does not ameliorate methionine and choline deficient diet-induced non alcoholic steatohepatitis in mice. PLoS One 2017. [PMID: 28632778 PMCID: PMC5478123 DOI: 10.1371/journal.pone.0179099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a liver disease with the potential to lead to cirrhosis and hepatocellular carcinoma. Interleukin-6 (IL-6) has been implicated in the pathogenesis of NASH, with the so-called IL-6 ‘trans-signaling’ cascade being responsible for the pro-inflammatory actions of this cytokine. We aimed to block IL-6 ‘trans-signaling’, using a transgenic mouse that overexpresses human soluble glycoprotein130 (sgp130Fc Tg mice) fed a commonly used dietary model of inducing NASH (methionine and choline deficient-diet; MCD diet) and hypothesized that markers of NASH would be ameliorated in such mice. Sgp130Fc Tg and littermate control mice were fed a MCD or control diet for 4 weeks. The MCD diet induced many hallmarks of NASH including hepatomegaly, steatosis, and liver inflammation. However, in contrast with other mouse models and, indeed, human NASH, the MCD diet model did not increase the mRNA or protein expression of IL-6. Not surprisingly, therefore, markers of MCD diet-induced NASH were unaffected by sgp130Fc transgenic expression. While the MCD diet model induces many pathophysiological markers of NASH, it does not induce increased IL-6 expression in the liver, a key hallmark of human NASH. We, therefore, caution the use of the MCD diet as a viable mouse model of NASH.
Collapse
Affiliation(s)
- Helene L. Kammoun
- Cellular and Molecular Metabolism Laboratory, Baker Heart & Diabetes Institute, Melbourne, Australia
- Immunology department, Monash University, Melbourne, Australia
- * E-mail:
| | - Tamara Louise Allen
- Cellular and Molecular Metabolism Laboratory, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Darren Colin Henstridge
- Cellular and Molecular Metabolism Laboratory, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Michael James Kraakman
- Cellular and Molecular Metabolism Laboratory, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Lone Peijs
- Cellular and Molecular Metabolism Laboratory, Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Stefan Rose-John
- Department of Biochemistry, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Mark Anthony Febbraio
- Cellular and Molecular Metabolism Laboratory, Baker Heart & Diabetes Institute, Melbourne, Australia
- Cellular and Molecular Metabolism, Garvan Institute, Sydney, Australia
| |
Collapse
|
46
|
M2-specific reduction of CD1d switches NKT cell-mediated immune responses and triggers metaflammation in adipose tissue. Cell Mol Immunol 2017; 15:506-517. [PMID: 28392574 DOI: 10.1038/cmi.2017.11] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/21/2017] [Accepted: 01/21/2017] [Indexed: 01/21/2023] Open
Abstract
Metaflammation is responsible for several metabolic syndromes, such as type 2 diabetes. However, the mechanisms by which metabolic disorders trigger metaflammation remain unclear. We identified a cell type-specific downregulation of CD1d expression in M2 macrophages during the progression of obesity prior to the onset of inflammation in visceral adipose tissues. A reduction in CD1d expression influenced the ability of M2 macrophages to present antigens and caused a change in antigen-presenting cells from M2 macrophages to M1 macrophages. With CD1d conditional knockout (KO) mice, we further demonstrated that natural killer T (NKT) cell activation by M2 macrophages inhibited metaflammation and insulin resistance by promoting Th2 responses and M2 polarization in visceral adipose tissues of obese mice, whereas NKT cell activation by M1 macrophages exacerbated metaflammation and insulin resistance by promoting Th1 responses and inhibiting M2 polarization. Our results suggest that an M2-specific reduction of CD1d is an initiating event that switches NKT cell-mediated immune responses and disrupts the immune balance in visceral adipose tissues in obese mice.
Collapse
|
47
|
Glöde A, Naumann J, Gnad T, Cannone V, Kilic A, Burnett JC, Pfeifer A. Divergent effects of a designer natriuretic peptide CD-NP in the regulation of adipose tissue and metabolism. Mol Metab 2017; 6:276-287. [PMID: 28271034 PMCID: PMC5323888 DOI: 10.1016/j.molmet.2016.12.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/20/2016] [Accepted: 12/29/2016] [Indexed: 01/06/2023] Open
Abstract
Objective Obesity is defined as an abnormal increase in white adipose tissue (WAT) and is a major risk factor for type 2 diabetes and cardiovascular disease. Brown adipose tissue (BAT) dissipates energy and correlates with leanness. Natriuretic peptides have been shown to be beneficial for brown adipocyte differentiation and browning of WAT. Methods Here, we investigated the effects of an optimized designer natriuretic peptide (CD-NP) on murine adipose tissues in vitro and in vivo. Results In murine brown and white adipocytes, CD-NP activated cGMP production, promoted adipogenesis, and increased thermogenic markers. Consequently, mice treated for 10 days with CD-NP exhibited increased “browning” of WAT. To study CD-NP effects on diet-induced obesity (DIO), we delivered CD-NP for 12 weeks. Although CD-NP reduced inflammation in WAT, CD-NP treated DIO mice exhibited a significant increase in body mass, worsened glucose tolerance, and hepatic steatosis. Long-term CD-NP treatment resulted in an increased expression of the NP scavenging receptor (NPR-C) and decreased lipolytic activity. Conclusions NP effects differed depending on the duration of treatment raising questions about the rational of natriuretic peptide treatment in obese patients. The optimized designer natriuretic peptide CD-NP promotes adipogenesis. Duration of treatment is decisive: short-term promotes browning whereas long-term treatment exacerbates obesity and diabetes. Long-term CD-NP treatment reduces WAT inflammation and increases adiponectin expression.
Collapse
Affiliation(s)
- Anja Glöde
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany; Bonn International Graduate School of Drug Sciences BIGSDrugS, University of Bonn, Bonn, Germany
| | - Jennifer Naumann
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Valentina Cannone
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, USA
| | - Ana Kilic
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, USA
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany; Bonn International Graduate School of Drug Sciences BIGSDrugS, University of Bonn, Bonn, Germany.
| |
Collapse
|
48
|
Boersema GSA, Utomo L, Bayon Y, Kops N, van der Harst E, Lange JF, Bastiaansen-Jenniskens YM. Monocyte subsets in blood correlate with obesity related response of macrophages to biomaterials in vitro. Biomaterials 2016; 109:32-39. [PMID: 27662579 DOI: 10.1016/j.biomaterials.2016.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/31/2016] [Accepted: 09/11/2016] [Indexed: 02/07/2023]
Abstract
Macrophages play a key role in the foreign body response. In this study it was investigated whether obesity affects the acute response of macrophages to biomaterials in vitro and whether this response is associated with biomarkers in blood. CD14 + monocytes were isolated from blood from obese and age and gender matched lean persons. Monocyte subsets were determined based on CD14 and CD16 on their surface. C-reactive protein (CRP) was measured in peripheral blood. The response of monocyte-derived macrophages to polypropylene (PP), polylactic acid (PLA), polyethylene terephthalate (PET) monofilament, and PET-multifilament (mPET) in culture was based on cytokine production. More IL-6 (for PET), less CCL18 (all materials) and IL-1ra (for PLA) was produced by macrophages from obese patients than lean subjects. Body mass index, serum CRP and to a lesser extend percentages of monocyte subtypes correlated with IL-6, TNFα, CCL18, and IL-1ra production. Taken together, monocyte-derived macrophages of obese patients respond more pro-inflammatory and less anti-inflammatory to biomaterials than macrophages from lean subjects, depending on the material. These results are a step towards personalized medicine for the development of a model or even a blood test to decide which biomaterial might be suitable for each patient.
Collapse
Affiliation(s)
- G S A Boersema
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, The Netherlands; Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - L Utomo
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Y Bayon
- Medtronic-Sofradim Production, 116 Avenue Formans, 01600 Trévoux, France
| | - N Kops
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - E van der Harst
- Department of Surgery, Maasstad Hospital, Rotterdam, The Netherlands
| | - J F Lange
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | | |
Collapse
|
49
|
Abstract
The skin is considered the mirror of the soul and is affected by neurohormonal triggers, especially stress. Hair follicles, keratinocytes, mast cells, melanocytes, and sebocytes all express sex and stress hormones implicating them in a local "hypothalamic-pituitary-adrenal axis." In particular, the peptides corticotropin-releasing hormone (CRH) and neurotensin (NT) have synergistic action stimulating mast cells and are uniquely elevated in the serum of patients with skin diseases exacerbated by stress. Addressing the neurohormonal regulation of skin function could lead to new targets for effective treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Department of Integrative Physiology and Pathobiology, Molecular Immunopharmacology and Drug Discovery Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Suite J304, Boston, MA, 02111, USA.
- Sackler School of Graduate Biomedical Sciences, Program in Pharmacology and Experimental Therapeutics, Tufts University, Boston, MA, USA.
- Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, MA, USA.
| | - Julia M Stewart
- Department of Integrative Physiology and Pathobiology, Molecular Immunopharmacology and Drug Discovery Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Suite J304, Boston, MA, 02111, USA
| | - Alexandra Taracanova
- Department of Integrative Physiology and Pathobiology, Molecular Immunopharmacology and Drug Discovery Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Suite J304, Boston, MA, 02111, USA
- Sackler School of Graduate Biomedical Sciences, Program in Pharmacology and Experimental Therapeutics, Tufts University, Boston, MA, USA
| | - Pio Conti
- Department of Graduate Medical Sciences, University of Chieti, Chieti, Italy
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Dessau, Germany
| |
Collapse
|
50
|
Kang MG, Lee HJ, Cho JY, Kim K, Yang SJ, Kim D. Anti-inflammatory effects of sucrose-derived oligosaccharides produced by a constitutive mutant L. mesenteroides B-512FMCM dextransucrase in high fat diet-fed mice. Biochem Biophys Res Commun 2016; 477:350-5. [PMID: 27342664 DOI: 10.1016/j.bbrc.2016.06.102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 01/07/2023]
Abstract
Oligosaccharide (OS) is used as a sugar replacement as well as an ingredient in functional foods because of its beneficial effects, mainly on reducing calorie content and promoting intestinal health. By contrast, the effects of OS on inflammation are less well investigated. The purpose of this study was to investigate the effects of sucrose-derived OS on glucose control and inflammation in high fat (HF) diet-fed mice. Male C57BL6 mice were randomly assigned to six treatment groups (n = 10-14 mice per group): 1) lean control (CON), 2) HF control, 3) HF-low sucrose (LS, 100 mg/kg/day), 4) HF-high sucrose (HS, 1000 mg/kg/day), 5) HF-low OS (LOS, 100 mg/kg/day), and 6) HF-high OS (HOS, 1000 mg/kg/day). PBS (vehicle), sucrose, and OS were administered by stomach gavage. Body weight, food intake, and markers of liver function (activities of aspartate aminotransferase and alanine aminotransferase) were not affected by the treatments. HOS treatment decreased levels of serum glucose, insulin, and homeostasis model assessment-insulin resistance compared with sucrose treatment. However, serum adiponectin levels of the HOS group were higher than those of the sucrose groups. Serum levels of the pro-inflammatory cytokines interleukin-6 (IL-6) and fetuin-A were lower in the HOS group than in the sucrose groups. Hepatic gene expression levels of pro-inflammatory cytokines and related factors (fetuin-A, NF-κB, TLR4, TNF-alpha, and IL-6) were decreased and the levels of insulin signaling-related molecules (sirtuin 1, insulin receptor, and Akt) were increased in HOS-treated mice as compared with sucrose-treated mice. These results demonstrate that OS treatment is effective in improving glucose control and inflammation in high fat diet-fed mice.
Collapse
Affiliation(s)
- Min-Gyung Kang
- Division of Food and Nutrition, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hee Jae Lee
- Department of Food and Nutrition, Seoul Women's University, Seoul, 01797, Republic of Korea
| | - Jae-Young Cho
- Graduate School of International Agricultural Technology and Institute of Food Industrialization, Institutes of Green Bio Science & Technology, Seoul National University, Gangwon-do, 25354, Republic of Korea
| | - Kanghwa Kim
- Division of Food and Nutrition, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Soo Jin Yang
- Department of Food and Nutrition, Seoul Women's University, Seoul, 01797, Republic of Korea.
| | - Doman Kim
- Graduate School of International Agricultural Technology and Institute of Food Industrialization, Institutes of Green Bio Science & Technology, Seoul National University, Gangwon-do, 25354, Republic of Korea.
| |
Collapse
|