1
|
Wu M, Chen K, Zheng X, Shen Y, Lo TH, Yang X. A galactoglucomannan from Termitomyces intermedius: Structural characterization, anti-obesity and anti-inflammatory activities. Pharmacol Res 2025:107768. [PMID: 40414582 DOI: 10.1016/j.phrs.2025.107768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/27/2025]
Abstract
Termitomyces mushrooms exhibit a wide range of beneficial properties, yet detailed structural characterization and exploration of their structure-activity relationships, particularly in the anti-obesity contexts, remain limited. A novel galactoglucomannan (TIP-1-1) was isolated from Termitomyces intermedius. Structural analysis showed TIP-1-1 is a 2.7kDa polysaccharide mainly composed of glucose, galactose and mannose (mass ratio: 75.7: 10.7: 13.6). The backbone contained linear 1,6-linked β-Glcp, 1,3-linked β-Glcp, 1,6-linked α-Galp, and 1,3,6-linked α-Manp contained a branched structure that substituted primarily with 1,3,6-linked α-Manp and 1,3-linked β-Glcp residues. Functionally, TIP-1-1 mitigated HFD-induced obesity in mice by reducing body weight, fat mass, and improving glucose homeostasis and lipid profiles, with effects comparable to GLP-1. It suppressed pro-inflammatory cytokines such as NO, TNF-α, IL-6, IL-1β, leptin and restored adiponectin and IL-10 levels both in vivo and in vitro, suggesting anti-inflammatory mechanisms underlying its anti-obesity effects. In lipid metabolism studies, TIP-1-1 inhibited adipocyte lipogenesis by activating AMPK, suppressing the expression of ACC and FASN, and reducing triglyceride accumulation. Mechanistically, AMPK siRNA knockdown abolished these effects, confirming its central role in TIP-1-1-mediated suppression of de novo fatty acid synthesis. These findings established TIP-1-1 as a dual-acting anti-obesity agent with therapeutic potential via anti-inflammatory and AMPK/ACC/FASN lipogenic inhibition.
Collapse
Affiliation(s)
- Mengyao Wu
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangdong, 510006, China; Department of Heath Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Kunzhui Chen
- School of Life Sciences, Guangzhou University, Guangdong, 510006, China
| | - Xiyin Zheng
- School of Life Sciences, Guangzhou University, Guangdong, 510006, China
| | - Yingbin Shen
- School of Life Sciences, Guangzhou University, Guangdong, 510006, China
| | - Tak-Ho Lo
- School of Life Sciences, Guangzhou University, Guangdong, 510006, China; Department of Heath Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xinquan Yang
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangdong, 510006, China
| |
Collapse
|
2
|
Mao X, Paerhati G, Wu Y, Cheng LF. Modulation of gut microbiota, up-regulation of ZO-1, and promotion of metabolism as therapeutic mechanisms of indole-3-carbinol against obesity in mice. Front Pharmacol 2025; 15:1499142. [PMID: 39830328 PMCID: PMC11739362 DOI: 10.3389/fphar.2024.1499142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Background Indole-3-carbinol (I3C) is a compound derived from Cruciferous vegetables. We aim to ascertain whether I3C mediates the relations between mouse gut microbiota, intestinal barrier function, and metabolism to treat obesity in mice. Methods The experimental analyses focused on the changes in lipid distribution, inflammatory cytokines, glucose tolerance, gut microbiota composition, and serum metabolomics of 60 C57BL/6N mice. Results The experimental results demonstrated that I3C reduced body weight, hepatic steatosis, and systemic inflammation and improved insulin resistance in mice on a high-fat diet (HFD). Furthermore, I3C remarkably enhanced the enrichment of probiotics Akkermansia and Ligilactobacillus as well as SCFA-producing bacteria (Eubacterium, Lactococcus, and Coprococcus), while reducing the abundance of Eisenbergiella and Rikenellaceae_RC9_gut_group. Also, I3C notably up-regulated the levels of Claudin4, Occludin, and ZO-1 proteins and modulated the metabolism of argininosuccinic acid and galactose. Conclusion The aforementioned findings suggest that I3C exerts a significant anti-obesity effect in mice by regulating abnormal gut microbiome, enhancing intestinal barrier function, and improving metabolic disorders.
Collapse
Affiliation(s)
- XuWen Mao
- College of Pharmacy, Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Xinjiang Medical University, Ürümqi, China
| | - Guliruoyi Paerhati
- College of Pharmacy, Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Xinjiang Medical University, Ürümqi, China
| | - Yuche Wu
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences (CAS), Ürümqi, China
| | - Lu Feng Cheng
- College of Pharmacy, Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Xinjiang Medical University, Ürümqi, China
| |
Collapse
|
3
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
4
|
Tallino S, Etebari R, McDonough I, Leon H, Sepulveda I, Winslow W, Bartholomew SK, Perez SE, Mufson EJ, Velazquez R. Assessing the Benefit of Dietary Choline Supplementation Throughout Adulthood in the Ts65Dn Mouse Model of Down Syndrome. Nutrients 2024; 16:4167. [PMID: 39683562 DOI: 10.3390/nu16234167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Down syndrome (DS) is the most common cause of early-onset Alzheimer's disease (AD). Dietary choline has been proposed as a modifiable factor to improve the cognitive and pathological outcomes of AD and DS, especially as many do not reach adequate daily intake levels of choline. While lower circulating choline levels correlate with worse pathological measures in AD patients, choline status and intake in DS is widely understudied. Perinatal choline supplementation (Ch+) in the Ts65Dn mouse model of DS protects offspring against AD-relevant pathology and improves cognition. Further, dietary Ch+ in adult AD models also ameliorates pathology and improves cognition. However, dietary Ch+ in adult Ts65Dn mice has not yet been explored; thus, this study aimed to supply Ch+ throughout adulthood to determine the effects on cognition and DS co-morbidities. METHODS We fed trisomic Ts65Dn mice and disomic littermate controls either a choline normal (ChN; 1.1 g/kg) or a Ch+ (5 g/kg) diet from 4.5 to 14 months of age. RESULTS We found that Ch+ in adulthood failed to improve genotype-specific deficits in spatial learning. However, in both genotypes of female mice, Ch+ significantly improved cognitive flexibility in a reverse place preference task in the IntelliCage behavioral phenotyping system. Further, Ch+ significantly reduced weight gain and peripheral inflammation in female mice of both genotypes, and significantly improved glucose metabolism in male mice of both genotypes. CONCLUSIONS Our findings suggest that adulthood choline supplementation benefits behavioral and biological factors important for general well-being in DS and related to AD risk.
Collapse
Affiliation(s)
- Savannah Tallino
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Rachel Etebari
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Ian McDonough
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Hector Leon
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Isabella Sepulveda
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Wendy Winslow
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Samantha K Bartholomew
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Sylvia E Perez
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Elliott J Mufson
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Ramon Velazquez
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| |
Collapse
|
5
|
Hofwimmer K, de Paula Souza J, Subramanian N, Vujičić M, Rachid L, Méreau H, Zhao C, Dror E, Barreby E, Björkström NK, Wernstedt Asterholm I, Böni-Schnetzler M, Meier DT, Donath MY, Laurencikiene J. IL-1β promotes adipogenesis by directly targeting adipocyte precursors. Nat Commun 2024; 15:7957. [PMID: 39261467 PMCID: PMC11390900 DOI: 10.1038/s41467-024-51938-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
Postprandial IL-1β surges are predominant in the white adipose tissue (WAT), but its consequences are unknown. Here, we investigate the role of IL-1β in WAT energy storage and show that adipocyte-specific deletion of IL-1 receptor 1 (IL1R1) has no metabolic consequences, whereas ubiquitous lack of IL1R1 reduces body weight, WAT mass, and adipocyte formation in mice. Among all major WAT-resident cell types, progenitors express the highest IL1R1 levels. In vitro, IL-1β potently promotes adipogenesis in murine and human adipose-derived stem cells. This effect is exclusive to early-differentiation-stage cells, in which the adipogenic transcription factors C/EBPδ and C/EBPβ are rapidly upregulated by IL-1β and enriched near important adipogenic genes. The pro-adipogenic, but not pro-inflammatory effect of IL-1β is potentiated by acute treatment and blocked by chronic exposure. Thus, we propose that transient postprandial IL-1β surges regulate WAT remodeling by promoting adipogenesis, whereas chronically elevated IL-1β levels in obesity blunts this physiological function.
Collapse
Affiliation(s)
- Kaisa Hofwimmer
- Lipid Laboratory, Unit of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, SE-141 52, Huddinge, Sweden
| | - Joyce de Paula Souza
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Narmadha Subramanian
- Lipid Laboratory, Unit of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, SE-141 52, Huddinge, Sweden
| | - Milica Vujičić
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Leila Rachid
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Hélène Méreau
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Cheng Zhao
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Erez Dror
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Emelie Barreby
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, SE-141 52, Huddinge, Sweden
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, SE-141 52, Huddinge, Sweden
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Marianne Böni-Schnetzler
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Daniel T Meier
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland.
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland.
| | - Marc Y Donath
- Department of Biomedicine, University of Basel and University Hospital Basel, 4031, Basel, Switzerland
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland
| | - Jurga Laurencikiene
- Lipid Laboratory, Unit of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, SE-141 52, Huddinge, Sweden.
| |
Collapse
|
6
|
Liu J, Chen Y. Cell-cell crosstalk between fat cells and immune cells. Am J Physiol Endocrinol Metab 2024; 327:E371-E383. [PMID: 39082899 DOI: 10.1152/ajpendo.00024.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/06/2024]
Abstract
Obesity is a metabolic disorder with pandemic-like implications, lacking viable pharmaceutical treatments currently. Thermogenic adipose tissues, including brown and beige adipose tissues, play an essential role in regulating systemic energy homeostasis and have emerged as appealing therapeutic targets for the treatment of obesity and obesity-related diseases. The function of adipocytes is subject to complex regulation by a cellular network of immune signaling pathways in response to environmental signals. However, the specific regulatory roles of immune cells in thermogenesis and relevant involving mechanisms are still not well understood. Here, we concentrate on our present knowledge of the interaction between thermogenic adipocytes and immune cells and present an overview of cellular and molecular mechanisms underlying immunometabolism in adipose tissues. We discuss cytokines, especially interleukins, which originate from widely variable sources, and their impacts on the development and function of thermogenic adipocytes. Moreover, we summarize the neuroimmune regulation in heat production and expand a new mode of intercellular communication mediated by mitochondrial transfer. The crosstalk between immune cells and adipocytes achieves adipose tissue homeostasis and systemic energy balance. A deep understanding of this intricate interaction would provide evidence for improving thermogenic efficiency by remodeling the immune microenvironment. Interventions based on these factors show a high potential to prevent adverse metabolic outcomes in patients with obesity.
Collapse
Affiliation(s)
- Jiadai Liu
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Laboratory of Endocrinology and Metabolism, Ministry of Education Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Laboratory of Endocrinology and Metabolism, Ministry of Education Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, People's Republic of China
| |
Collapse
|
7
|
Savulescu-Fiedler I, Mihalcea R, Dragosloveanu S, Scheau C, Baz RO, Caruntu A, Scheau AE, Caruntu C, Benea SN. The Interplay between Obesity and Inflammation. Life (Basel) 2024; 14:856. [PMID: 39063610 PMCID: PMC11277997 DOI: 10.3390/life14070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is an important condition affecting the quality of life of numerous patients and increasing their associated risk for multiple diseases, including tumors and immune-mediated disorders. Inflammation appears to play a major role in the development of obesity and represents a central point for the activity of cellular and humoral components in the adipose tissue. Macrophages play a key role as the main cellular component of the adipose tissue regulating the chronic inflammation and modulating the secretion and differentiation of various pro- and anti-inflammatory cytokines. Inflammation also involves a series of signaling pathways that might represent the focus for new therapies and interventions. Weight loss is essential in decreasing cardiometabolic risks and the degree of associated inflammation; however, the latter can persist for long after the excess weight is lost, and can involve changes in macrophage phenotypes that can ensure the metabolic adjustment. A clear understanding of the pathophysiological processes in the adipose tissue and the interplay between obesity and chronic inflammation can lead to a better understanding of the development of comorbidities and may ensure future targets for the treatment of obesity.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Razvan Mihalcea
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania (C.C.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- “Prof. Dr. Matei Balș” National Institute for Infectious Diseases, 021105 Bucharest, Romania
| |
Collapse
|
8
|
Kearns ML, Reynolds CM. Developmentally programmed obesity: Is there a role for anti-inflammatory nutritional strategies? Exp Physiol 2024; 109:633-646. [PMID: 38031876 PMCID: PMC11061634 DOI: 10.1113/ep091209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023]
Abstract
Pregnancy represents a period of immense maternal physiological adaptation, with progressive increases in lipid storage potential and insulin resistance to support fetal/placental growth. This requires significant change in the adipose tissue. Women living with obesity/overweight are more susceptible to these changes causing complications such as gestational diabetes. This is particularly worrying as up to 60% of European women are living with overweight/obesity at the onset of pregnancy. Furthermore, less than 1% meet all nutrition guidelines. There is now evidence that these deep metabolic changes can result in a predisposition to metabolic disease in both the mother and child in later life. Health and nutrition status during this period therefore represents a window to future health. This period offers a valuable opportunity for intervention to prevent the negative consequences of poor in utero environments and increases the long-term quality of life for mother and offspring. This review will examine a range of in utero factors which determine adipose tissue development, the impact of these factors on later-life obesity and metabolic health and the therapeutic value of dietary anti-inflammatory nutritional interventions during pregnancy and early life. When it comes to early life nutrition, a 'one size fits all' approach is not always appropriate. Understanding the mechanisms of adipose tissue development in response to differing nutritional strategies may be important in the context of complicated or adverse in utero environments and represents a substantial step towards a more personalised nutritional approach for the prevention of obesity, metabolic syndrome and related non-communicable diseases in future generations.
Collapse
Affiliation(s)
- Michelle L. Kearns
- Conway Institute/School of Public Health Physiotherapy and Sports Science/Institute of Food and Health/Diabetes Complications Research CentreUniversity College DublinDublin 4Ireland
| | - Clare M. Reynolds
- Conway Institute/School of Public Health Physiotherapy and Sports Science/Institute of Food and Health/Diabetes Complications Research CentreUniversity College DublinDublin 4Ireland
| |
Collapse
|
9
|
Guan Y, Wei X, Li J, Zhu Y, Luo P, Luo M. Obesity-related glomerulopathy: recent advances in inflammatory mechanisms and related treatments. J Leukoc Biol 2024; 115:819-839. [PMID: 38427925 DOI: 10.1093/jleuko/qiae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 03/03/2024] Open
Abstract
Obesity-related glomerulopathy, which is an obesity-triggered kidney damage, has become a significant threat to human health. Several studies have recently highlighted the critical role of inflammation in obesity-related glomerulopathy development. Additionally, excess adipose tissue and adipocytes in patients with obesity produce various inflammatory factors that cause systemic low-grade inflammation with consequent damage to vascular endothelial cells, exacerbating glomerular injury. Therefore, we conducted a comprehensive review of obesity-related glomerulopathy and addressed the critical role of obesity-induced chronic inflammation in obesity-related glomerulopathy pathogenesis and progression, which leads to tubular damage and proteinuria, ultimately impairing renal function. The relationship between obesity and obesity-related glomerulopathy is facilitated by a network of various inflammation-associated cells (including macrophages, lymphocytes, and mast cells) and a series of inflammatory mediators (such as tumor necrosis factor α, interleukin 6, leptin, adiponectin, resistin, chemokines, adhesion molecules, and plasminogen activator inhibitor 1) and their inflammatory pathways. Furthermore, we discuss a recently discovered relationship between micronutrients and obesity-related glomerulopathy inflammation and the important role of micronutrients in the body's anti-inflammatory response. Therefore, assessing these inflammatory molecules and pathways will provide a strong theoretical basis for developing therapeutic strategies based on anti-inflammatory effects to prevent or delay the onset of kidney injury.
Collapse
Affiliation(s)
- Yucan Guan
- Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziquiang Street, Nanguan District, Changchun, Jilin 130041, China
| | - Xianping Wei
- Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziquiang Street, Nanguan District, Changchun, Jilin 130041, China
| | - Jicui Li
- Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziquiang Street, Nanguan District, Changchun, Jilin 130041, China
| | - Yuexin Zhu
- Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziquiang Street, Nanguan District, Changchun, Jilin 130041, China
| | - Ping Luo
- Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziquiang Street, Nanguan District, Changchun, Jilin 130041, China
| | - Manyu Luo
- Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziquiang Street, Nanguan District, Changchun, Jilin 130041, China
| |
Collapse
|
10
|
Mitchelson KAJ, O’Connell F, O’Sullivan J, Roche HM. Obesity, Dietary Fats, and Gastrointestinal Cancer Risk-Potential Mechanisms Relating to Lipid Metabolism and Inflammation. Metabolites 2024; 14:42. [PMID: 38248845 PMCID: PMC10821017 DOI: 10.3390/metabo14010042] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Obesity is a major driving factor in the incidence, progression, and poor treatment response in gastrointestinal cancers. Herein, we conducted a comprehensive analysis of the impact of obesity and its resulting metabolic perturbations across four gastrointestinal cancer types, namely, oesophageal, gastric, liver, and colorectal cancer. Importantly, not all obese phenotypes are equal. Obese adipose tissue heterogeneity depends on the location, structure, cellular profile (including resident immune cell populations), and dietary fatty acid intake. We discuss whether adipose heterogeneity impacts the tumorigenic environment. Dietary fat quality, in particular saturated fatty acids, promotes a hypertrophic, pro-inflammatory adipose profile, in contrast to monounsaturated fatty acids, resulting in a hyperplastic, less inflammatory adipose phenotype. The purpose of this review is to examine the impact of obesity, including dietary fat quality, on adipose tissue biology and oncogenesis, specifically focusing on lipid metabolism and inflammatory mechanisms. This is achieved with a particular focus on gastrointestinal cancers as exemplar models of obesity-associated cancers.
Collapse
Affiliation(s)
- Kathleen A. J. Mitchelson
- Nutrigenomics Research Group, UCD Conway Institute, UCD Institute of Food and Health, and School of Public Health, Physiotherapy and Sports Science, University College Dublin, D04 H1W8 Dublin, Ireland
| | - Fiona O’Connell
- Department of Surgery, Trinity St. James’s Cancer Institute and Trinity Translational Medicine Institute, St. James’s Hospital and Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Jacintha O’Sullivan
- Department of Surgery, Trinity St. James’s Cancer Institute and Trinity Translational Medicine Institute, St. James’s Hospital and Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Helen M. Roche
- Nutrigenomics Research Group, UCD Conway Institute, UCD Institute of Food and Health, and School of Public Health, Physiotherapy and Sports Science, University College Dublin, D04 H1W8 Dublin, Ireland
- Institute for Global Food Security, School of Biological Sciences, Queens University Belfast, Belfast BT9 5DL, UK
| |
Collapse
|
11
|
Cho S, Ying F, Sweeney G. Sterile inflammation and the NLRP3 inflammasome in cardiometabolic disease. Biomed J 2023; 46:100624. [PMID: 37336361 PMCID: PMC10539878 DOI: 10.1016/j.bj.2023.100624] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/21/2023] Open
Abstract
Inflammation plays an important role in the pathophysiology of cardiometabolic diseases. Sterile inflammation, a non-infectious and damage-associated molecular pattern (DAMP)-induced innate response, is now well-established to be closely associated with development and progression of cardiometabolic diseases. The NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is well-established as a major player in sterile inflammatory responses. It is a multimeric cytosolic protein complex which regulates the activation of caspase-1 and subsequently promotes cleavage and release of interleukin (IL)-1 family cytokines, which have a deleterious impact on the development of cardiometabolic diseases. Therefore, targeting NLRP3 itself or the downstream consequences of NLRP3 activation represent excellent potential therapeutic targets in inflammatory cardiometabolic diseases. Here, we review our current understanding of the role which NLRP3 inflammasome regulation plays in cardiometabolic diseases such as obesity, diabetes, non-alcoholic steatohepatitis (NASH), atherosclerosis, ischemic heart disease and cardiomyopathy. Finally, we highlight the potential of targeting NLPR3 or related signaling molecules as a therapeutic approach.
Collapse
Affiliation(s)
- Sungji Cho
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Fan Ying
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Abstract
In this review, we provide a brief synopsis of the connections between adipose tissue and metabolic health and highlight some recent developments in understanding and exploiting adipocyte biology. Adipose tissue plays critical roles in the regulation of systemic glucose and lipid metabolism and secretes bioactive molecules possessing endocrine, paracrine, and autocrine functions. Dysfunctional adipose tissue has a detrimental impact on metabolic health and is intimately involved in key aspects of metabolic diseases such as insulin resistance, lipid overload, inflammation, and organelle stress. Differences in the distribution of fat depots and adipose characteristics relate to divergent degrees of metabolic dysfunction found in metabolically healthy and unhealthy obese individuals. Thermogenic adipocytes increase energy expenditure via mitochondrial uncoupling or adenosine triphosphate-consuming futile substrate cycles, while functioning as a metabolic sink and participating in crosstalk with other metabolic organs. Manipulation of adipose tissue provides a wealth of opportunities to intervene and combat the progression of associated metabolic diseases. We discuss current treatment modalities for obesity including incretin hormone analogs and touch upon emerging strategies with therapeutic potential including exosome-based therapy, pharmacological activation of brown and beige adipocyte thermogenesis, and administration or inhibition of adipocyte-derived factors.
Collapse
Affiliation(s)
- Sung-Min An
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - Seung-Hee Cho
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - John C. Yoon
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
13
|
Li JH, Hepworth MR, O'Sullivan TE. Regulation of systemic metabolism by tissue-resident immune cell circuits. Immunity 2023; 56:1168-1186. [PMID: 37315533 PMCID: PMC10321269 DOI: 10.1016/j.immuni.2023.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023]
Abstract
Recent studies have demonstrated that tissue homeostasis and metabolic function are dependent on distinct tissue-resident immune cells that form functional cell circuits with structural cells. Within these cell circuits, immune cells integrate cues from dietary contents and commensal microbes in addition to endocrine and neuronal signals present in the tissue microenvironment to regulate structural cell metabolism. These tissue-resident immune circuits can become dysregulated during inflammation and dietary overnutrition, contributing to metabolic diseases. Here, we review the evidence describing key cellular networks within and between the liver, gastrointestinal tract, and adipose tissue that control systemic metabolism and how these cell circuits become dysregulated during certain metabolic diseases. We also identify open questions in the field that have the potential to enhance our understanding of metabolic health and disease.
Collapse
Affiliation(s)
- Joey H Li
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA; Medical Scientist Training Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| |
Collapse
|
14
|
Kiernan K, Nichols AG, Alwarawrah Y, MacIver NJ. Effects of T cell leptin signaling on systemic glucose tolerance and T cell responses in obesity. PLoS One 2023; 18:e0286470. [PMID: 37276236 PMCID: PMC10241364 DOI: 10.1371/journal.pone.0286470] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/16/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND/OBJECTIVES Leptin is an adipokine secreted in proportion to adipocyte mass and is therefore increased in obesity. Leptin signaling has been shown to directly promote inflammatory T helper 1 (Th1) and T helper 17 (Th17) cell number and function. Since T cells have a critical role in driving inflammation and systemic glucose intolerance in obesity, we sought to determine the role of leptin signaling in this context. METHODS Male and female T cell-specific leptin receptor knockout mice and littermate controls were placed on low-fat diet or high-fat diet to induce obesity for 18 weeks. Weight gain, serum glucose levels, systemic glucose tolerance, T cell metabolism, and T cell differentiation and cytokine production were examined. RESULTS In both male and female mice, T cell-specific leptin receptor deficiency did not reverse impaired glucose tolerance in obesity, although it did prevent impaired fasting glucose levels in obese mice compared to littermate controls, in a sex dependent manner. Despite these minimal effects on systemic metabolism, T cell-specific leptin signaling was required for changes in T cell metabolism, differentiation, and cytokine production observed in mice fed high-fat diet compared to low-fat diet. Specifically, we observed increased T cell oxidative metabolism, increased CD4+ T cell IFN-γ expression, and increased proportion of T regulatory (Treg) cells in control mice fed high-fat diet compared to low-fat diet, which were not observed in the leptin receptor conditional knockout mice, suggesting that leptin receptor signaling is required for some of the inflammatory changes observed in T cells in obesity. CONCLUSIONS T cell-specific deficiency of leptin signaling alters T cell metabolism and function in obesity but has minimal effects on obesity-associated systemic metabolism. These results suggest a redundancy in cytokine receptor signaling pathways in response to inflammatory signals in obesity.
Collapse
Affiliation(s)
- Kaitlin Kiernan
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Amanda G. Nichols
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Yazan Alwarawrah
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Nancie J. MacIver
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
- Department of Nutrition, University of North Carolina School of Medicine and Gillings School of Global Public Health, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
15
|
Bauer S, Hezinger L, Rexhepi F, Ramanathan S, Kufer TA. NOD-like Receptors-Emerging Links to Obesity and Associated Morbidities. Int J Mol Sci 2023; 24:ijms24108595. [PMID: 37239938 DOI: 10.3390/ijms24108595] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity and its associated metabolic morbidities have been and still are on the rise, posing a major challenge to health care systems worldwide. It has become evident over the last decades that a low-grade inflammatory response, primarily proceeding from the adipose tissue (AT), essentially contributes to adiposity-associated comorbidities, most prominently insulin resistance (IR), atherosclerosis and liver diseases. In mouse models, the release of pro-inflammatory cytokines such as TNF-alpha (TNF-α) and interleukin (IL)-1β and the imprinting of immune cells to a pro-inflammatory phenotype in AT play an important role. However, the underlying genetic and molecular determinants are not yet understood in detail. Recent evidence demonstrates that nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family proteins, a group of cytosolic pattern recognition receptors (PRR), contribute to the development and control of obesity and obesity-associated inflammatory responses. In this article, we review the current state of research on the role of NLR proteins in obesity and discuss the possible mechanisms leading to and the outcomes of NLR activation in the obesity-associated morbidities IR, type 2 diabetes mellitus (T2DM), atherosclerosis and non-alcoholic fatty liver disease (NAFLD) and discuss emerging ideas about possibilities for NLR-based therapeutic interventions of metabolic diseases.
Collapse
Affiliation(s)
- Sarah Bauer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| | - Lucy Hezinger
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| | - Fjolla Rexhepi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Thomas A Kufer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, 70593 Stuttgart, Germany
| |
Collapse
|
16
|
Mitchelson KAJ, Tran TTT, Dillon ET, Vlckova K, Harrison SM, Ntemiri A, Cunningham K, Gibson I, Finucane FM, O'Connor EM, Roche HM, O'Toole PW. Yeast β-Glucan Improves Insulin Sensitivity and Hepatic Lipid Metabolism in Mice Humanized with Obese Type 2 Diabetic Gut Microbiota. Mol Nutr Food Res 2022; 66:e2100819. [PMID: 36038526 PMCID: PMC9787509 DOI: 10.1002/mnfr.202100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Indexed: 12/30/2022]
Abstract
SCOPE Gut microbiota alterations are associated with obesity and type 2 diabetes. Yeast β-glucans are potential modulators of the innate immune-metabolic response, by impacting glucose, lipid, and cholesterol homeostasis. The study examines whether yeast β-glucan interacts differentially with either an obese healthy or obese diabetic gut microbiome, to impact metabolic health through hepatic effects under high-fat dietary challenge. METHODS AND RESULTS Male C57BL/6J mice are pre-inoculated with gut microbiota from obese healthy (OBH) or obese type 2 diabetic (OBD) subjects, in conjunction with a high-fat diet (HFD) with/without yeast β-glucan. OBD microbiome colonization adversely impacts metabolic health compared to OBH microbiome engraftment. OBD mice are more insulin resistant and display hepatic lipotoxicity compared to weight matched OBH mice. Yeast β-glucan supplementation resolves this adverse metabolic phenotype, coincident with increasing the abundance of health-related bacterial taxa. Hepatic proteomics demonstrates that OBD microbiome transplantation increases HFD-induced hepatic mitochondrial dysfunction, disrupts oxidative phosphorylation, and reduces protein synthesis, which are partly reverted by yeast β-glucan supplementation. CONCLUSIONS Hepatic metabolism is adversely affected by OBD microbiome colonization with high-fat feeding, but partially resolved by yeast β-glucan. More targeted dietary interventions that encompass the interactions between diet, gut microbiota, and host metabolism may have greater treatment efficacy.
Collapse
Affiliation(s)
- Kathleen A. J. Mitchelson
- Nutrigenomics Research Group and Institute of Food and HealthUniversity College DublinDublinDublin 4Republic of Ireland
| | - Tam T. T. Tran
- APC Microbiome IrelandUniversity College CorkCorkT12 K8AFRepublic of Ireland
- School of MicrobiologyUniversity College CorkCorkT12 K8AFRepublic of Ireland
- Present address:
Vietnam Academy of Science and TechnologyUniversity of Science and Technology of HanoiHanoiVietnam
| | - Eugene T. Dillon
- Mass Spectrometry ResourceConway Institute of Biomolecular & Biomedical ResearchUniversity College DublinDublinDublin 4Republic of Ireland
| | - Klara Vlckova
- APC Microbiome IrelandUniversity College CorkCorkT12 K8AFRepublic of Ireland
- School of MicrobiologyUniversity College CorkCorkT12 K8AFRepublic of Ireland
| | - Sabine M. Harrison
- UCD School of Agriculture & Food ScienceUniversity College DublinDublinDublin 4Republic of Ireland
| | - Alexandra Ntemiri
- APC Microbiome IrelandUniversity College CorkCorkT12 K8AFRepublic of Ireland
- School of MicrobiologyUniversity College CorkCorkT12 K8AFRepublic of Ireland
| | - Katie Cunningham
- Bariatric Medicine ServiceCentre for Diabetes, Endocrinology and MetabolismGalway University HospitalsGalwayH91 YR71Republic of Ireland
- Heart and Stroke CentreCroiThe West of Ireland Cardiac FoundationMoyola Lane, NewcastleGalwayGalwayH91 FF68Republic of Ireland
| | - Irene Gibson
- Heart and Stroke CentreCroiThe West of Ireland Cardiac FoundationMoyola Lane, NewcastleGalwayGalwayH91 FF68Republic of Ireland
| | - Francis M. Finucane
- Bariatric Medicine ServiceCentre for Diabetes, Endocrinology and MetabolismGalway University HospitalsGalwayH91 YR71Republic of Ireland
- HRB Clinical Research FacilityNational University of IrelandGalwayH91 TK33Republic of Ireland
| | - Eibhlís M. O'Connor
- APC Microbiome IrelandUniversity College CorkCorkT12 K8AFRepublic of Ireland
- Department of Biological SciencesSchool of Natural SciencesUniversity of LimerickLimerickV94 T9PXRepublic of Ireland
- Health Research InstituteUniversity of LimerickLimerickV94 T9PXRepublic of Ireland
| | - Helen M. Roche
- Nutrigenomics Research Group and Institute of Food and HealthUniversity College DublinDublinDublin 4Republic of Ireland
- Diabetes Complications Research CentreUniversity College DublinDublinDublin 4Republic of Ireland
- The Institute for Global Food SecuritySchool of Biological SciencesQueen's University BelfastBelfastBT9 5DLUK
| | - Paul W. O'Toole
- APC Microbiome IrelandUniversity College CorkCorkT12 K8AFRepublic of Ireland
- School of MicrobiologyUniversity College CorkCorkT12 K8AFRepublic of Ireland
| |
Collapse
|
17
|
Gehrke N, Hofmann LJ, Straub BK, Rühle F, Waisman A, Galle PR, Schattenberg JM. Hepatic interleukin-1 receptor type 1 signalling regulates insulin sensitivity in the early phases of nonalcoholic fatty liver disease. Clin Transl Med 2022; 12:e1048. [PMID: 36101976 PMCID: PMC9471277 DOI: 10.1002/ctm2.1048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is associated with hepatic as well as systemic insulin resistance even in the absence of type 2 diabetes. The extent and pathways through which hepatic inflammation modulates insulin sensitivity in NAFLD are only partially understood. We explored the contribution of hepatic interleukin (IL)-1 signalling in a novel conditional knockout mouse model and expand the knowledge on this signalling pathway with regard to its liver-specific functions. METHODS A high-fat, high-carbohydrate diet (HFD) over 12 weeks was used in male hepatocyte-specific IL-1 receptor type 1 (IL-1R1) knockout mice (Il1r1Hep-/- ) and wild-type (WT) littermates. RESULTS Both genotypes developed an obese phenotype and accompanying macrovesicular hepatic steatosis. In contrast to WT mice, microvesicular steatosis and ballooning injury was less pronounced in HFD-fed Il1r1Hep-/- mice, and alanine aminotransferase remained in the normal range. This was paralleled by the suppression of injurious and proinflammatory hepatic c-Jun N-terminal kinases and extracellular signal-regulated kinases signalling, stable peroxisome proliferator activated receptor gamma coactivator-1alpha and farnesoid X receptor-alpha expression and preservation of mitochondrial function. Strikingly, despite HFD-feeding Il1r1Hep-/- mice remained highly insulin sensitive as indicated by lower insulin levels, homeostatic model assessment for insulin resistance, higher glucose tolerance, more stable hepatic insulin signalling cascade, and less adipose tissue inflammation compared to the WT. CONCLUSIONS The current data highlights that hepatocyte IL-1R1 contributes to hepatic and extrahepatic insulin resistance. Future liver-directed therapies in NAFLD could have effects on insulin sensitivity when improving hepatic inflammation and IL-1R1 signalling.
Collapse
Affiliation(s)
- Nadine Gehrke
- I. Department of MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainz55131Germany
| | - Lea J. Hofmann
- I. Department of MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainz55131Germany
| | - Beate K. Straub
- Institute of PathologyUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Frank Rühle
- Bioinformatics Core FacilityInstitute of Molecular Biology (IMB)MainzGermany
| | - Ari Waisman
- Institute for Molecular MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Research Center for ImmunotherapyUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Peter R. Galle
- I. Department of MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainz55131Germany
- Research Center for ImmunotherapyUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Jörn M. Schattenberg
- I. Department of MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainz55131Germany
| |
Collapse
|
18
|
Gene expression pattern in severely progressing covid-19 patients is related to diabetes mellitus type 1: A functional annotation analysis. HUMAN GENE 2022. [PMID: 37520164 PMCID: PMC9217787 DOI: 10.1016/j.humgen.2022.201039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Aims The aim of this study was to extract the signaling mediators or biological pathways that link covid-19 to other diseases such as type 1 diabetes mellitus (T1DM). Methods Microarray data of covid-19 (GSE164805) was extracted from Gene Expression Omnibus (GEO) and analyses were performed by R package and GEO2R. Functional enrichment analysis was done to extract enriched molecular pathways (MP), biological process (BP) and molecular function (MF). Then commonly up- and down-regulated genes in covid-19 and T1DM were extracted by comparing deferentially expressed genes (DEGs) of GSE164805 and GSE9006. Results Down-regulated DEGs in the severely progressing covid-19 patients (SPCP) had a link to T1DM. Major histocompatibility system (MHC) class II, gamma interferon (IFNγ), and IL-1B were enriched in extracted pathway that leads to T1DM. In addition, comparing extracted DEGs from GSE164805 and GSE9006 indicated that MTUS1, EGR1 and EGR3 are the genes that are up-regulated in both SPCP and T1DM. Conclusion The findings of this study indicate that coincidence of SARS-COV-2 infection and T1DM may increase the severity of both diseases. Although covid-19 reduced the T cell mediated immune response, but increased mediators of T-cell signaling pathway such as IL-1 in both diseases. This could potentiate the inflammation response and worsens the severity of covid-19 cytokine storm or increase the resistance to insulin.
Collapse
|
19
|
Macrophages, Low-Grade Inflammation, Insulin Resistance and Hyperinsulinemia: A Mutual Ambiguous Relationship in the Development of Metabolic Diseases. J Clin Med 2022; 11:jcm11154358. [PMID: 35955975 PMCID: PMC9369133 DOI: 10.3390/jcm11154358] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic derangement with poor glycemic control accompanying overweight and obesity is associated with chronic low-grade inflammation and hyperinsulinemia. Macrophages, which present a very heterogeneous population of cells, play a key role in the maintenance of normal tissue homeostasis, but functional alterations in the resident macrophage pool as well as newly recruited monocyte-derived macrophages are important drivers in the development of low-grade inflammation. While metabolic dysfunction, insulin resistance and tissue damage may trigger or advance pro-inflammatory responses in macrophages, the inflammation itself contributes to the development of insulin resistance and the resulting hyperinsulinemia. Macrophages express insulin receptors whose downstream signaling networks share a number of knots with the signaling pathways of pattern recognition and cytokine receptors, which shape macrophage polarity. The shared knots allow insulin to enhance or attenuate both pro-inflammatory and anti-inflammatory macrophage responses. This supposedly physiological function may be impaired by hyperinsulinemia or insulin resistance in macrophages. This review discusses the mutual ambiguous relationship of low-grade inflammation, insulin resistance, hyperinsulinemia and the insulin-dependent modulation of macrophage activity with a focus on adipose tissue and liver.
Collapse
|
20
|
Qi B, Ren D, Li T, Niu P, Zhang X, Yang X, Xiao J. Fu Brick Tea Manages HFD/STZ-Induced Type 2 Diabetes by Regulating the Gut Microbiota and Activating the IRS1/PI3K/Akt Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:8274-8287. [PMID: 35767631 DOI: 10.1021/acs.jafc.2c02400] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The antidiabetic effects of Fu brick tea aqueous extract (FTE) and its underlying molecular mechanism in type 2 diabetes mellitus (T2DM) mice were investigated. FTE treatment significantly relieved dyslipidemia, insulin resistance (IR), and hepatic oxidative stress caused by T2DM. FTE also ameliorated the T2DM-induced gut dysbiosis by decreasing the Firmicutes/Bacteroidota (F/B) ratio at the phylum level and promoting the proliferation of Bifidobacterium, Parabacteroides, and Roseburia at the genus level. Besides, FTE significantly improved colonic short-chain fatty acid levels of T2DM mice. Furthermore, the antidiabetic effects of FTE were proved to be mediated by the IRS1/PI3K/Akt and AMPK-mediated gluconeogenesis signaling pathways. Metabolomics analysis illustrated that FTE recovered the levels of 28 metabolites associated with T2DM to the levels of normal mice. Taken together, these findings suggest that FTE can alleviate T2DM by reshaping the gut microbiota, activating the IRS1/PI3K/Akt pathway, and regulating intestinal metabolites.
Collapse
Affiliation(s)
- Bangran Qi
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Ting Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Pengfei Niu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xiangnan Zhang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain
| |
Collapse
|
21
|
Diacerein ameliorates letrozole-induced polycystic ovarian syndrome in rats. Biomed Pharmacother 2022; 149:112870. [PMID: 35367769 DOI: 10.1016/j.biopha.2022.112870] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/02/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common gynaecological endocrine disease that causes anovulatory infertility. The current study aimed to explore the possible role of diacerein (DIA), an IL-1β inhibitor, in treating letrozole-induced PCOS in rats that exhibit the metabolic and endocrinal criteria of PCOS patients. PCOS was induced in female Wistar rats by the oral administration of letrozole (1 mg/kg, per orally, p.o.) for 21 days. Rats were then treated with DIA (25 mg/kg/day, p.o.), DIA (50 mg/kg/day, p.o.), or metformin (2 mg/100 g/day, p.o.) for 14 days after the PCOS induction. PCOS resulted in a significantly higher body weight, ovarian weight, ovarian size, and cysts, as well as an elevation in serum testosterone, LH, insulin, glycemia, and lipid profile levels. All of these effects were significantly reduced by the DIA administration. Additionally, DIA remarkably inhibited the letrozole-induced oxidative stress in the ovaries, muscles, and liver by reducing the upraised levels of malondialdehyde and total nitrite and increasing the suppressed levels of superoxide dismutase and catalase. DIA enhanced the protective proteins Keap-1, Nrf2, and OH-1 levels. Finally, DIA inhibited the elevated mRNA levels of NLRP3 and caspase-1, the up-regulated inflammatory cytokines IL-6, TNF-α, and the IL-1β/NFκB signaling pathway. Our results proved that DIA ameliorates letrozole-induced PCOS through its antioxidant and anti-inflammatory properties.
Collapse
|
22
|
Dai M, Yang X, Yu Y, Pan W. Helminth and Host Crosstalk: New Insight Into Treatment of Obesity and Its Associated Metabolic Syndromes. Front Immunol 2022; 13:827486. [PMID: 35281054 PMCID: PMC8913526 DOI: 10.3389/fimmu.2022.827486] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity and its associated Metabolic Syndromes (Mets) represent a global epidemic health problem. Metabolic inflammation, lipid accumulation and insulin resistance contribute to the progression of these diseases, thereby becoming targets for drug development. Epidemiological data have showed that the rate of helminth infection negatively correlates with the incidence of obesity and Mets. Correspondingly, numerous animal experiments and a few of clinic trials in human demonstrate that helminth infection or its derived molecules can mitigate obesity and Mets via induction of macrophage M2 polarization, inhibition of adipogenesis, promotion of fat browning, and improvement of glucose tolerance, insulin resistance and metabolic inflammation. Interestingly, sporadic studies also uncover that several helminth infections can reshape gut microbiota of hosts, which is intimately implicated in the pathogenesis of obesity and Mets. Overall, these findings indicate that the crosstalk between helminth and hosts may be a novel direction for obesity and Mets therapy. The present article reviews the molecular mechanism of how helminth masters immunity and metabolism in obesity.
Collapse
Affiliation(s)
- Mengyu Dai
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The Second Clinical Medicine, Xuzhou Medical University, Xuzhou, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Wei Pan, ; Yinghua Yu,
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Wei Pan, ; Yinghua Yu,
| |
Collapse
|
23
|
Modulatory Properties of Food and Nutraceutical Components Targeting NLRP3 Inflammasome Activation. Nutrients 2022; 14:nu14030490. [PMID: 35276849 PMCID: PMC8840562 DOI: 10.3390/nu14030490] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 12/27/2022] Open
Abstract
Inflammasomes are key intracellular multimeric proteins able to initiate the cellular inflammatory signaling pathway. NLRP3 inflammasome represents one of the main protein complexes involved in the development of inflammatory events, and its activity has been largely demonstrated to be connected with inflammatory or autoinflammatory disorders, including diabetes, gouty arthritis, liver fibrosis, Alzheimer’s disease, respiratory syndromes, atherosclerosis, and cancer initiation. In recent years, it has been demonstrated how dietary intake and nutritional status represent important environmental elements that can modulate metabolic inflammation, since food matrices are an important source of several bioactive compounds. In this review, an updated status of knowledge regarding food bioactive compounds as NLRP3 inflammasome modulators is discussed. Several chemical classes, namely polyphenols, organosulfurs, terpenes, fatty acids, proteins, amino acids, saponins, sterols, polysaccharides, carotenoids, vitamins, and probiotics, have been shown to possess NLRP3 inflammasome-modulating activity through in vitro and in vivo assays, mainly demonstrating an anti-NLRP3 inflammasome activity. Plant foods are particularly rich in important bioactive compounds, each of them can have different effects on the pathway of inflammatory response, confirming the importance of the nutritional pattern (food model) as a whole rather than any single nutrient or functional compound.
Collapse
|
24
|
Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity 2022; 55:31-55. [PMID: 35021057 PMCID: PMC8773457 DOI: 10.1016/j.immuni.2021.12.013] [Citation(s) in RCA: 912] [Impact Index Per Article: 304.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 01/13/2023]
Abstract
Obesity leads to chronic, systemic inflammation and can lead to insulin resistance (IR), β-cell dysfunction, and ultimately type 2 diabetes (T2D). This chronic inflammatory state contributes to long-term complications of diabetes, including non-alcoholic fatty liver disease (NAFLD), retinopathy, cardiovascular disease, and nephropathy, and may underlie the association of type 2 diabetes with other conditions such as Alzheimer's disease, polycystic ovarian syndrome, gout, and rheumatoid arthritis. Here, we review the current understanding of the mechanisms underlying inflammation in obesity, T2D, and related disorders. We discuss how chronic tissue inflammation results in IR, impaired insulin secretion, glucose intolerance, and T2D and review the effect of inflammation on diabetic complications and on the relationship between T2D and other pathologies. In this context, we discuss current therapeutic options for the treatment of metabolic disease, advances in the clinic and the potential of immune-modulatory approaches.
Collapse
Affiliation(s)
- Theresa V. Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel T. Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Jerrold M. Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland.,Correspondence:
| |
Collapse
|
25
|
Wang X, Yang Y, Zhao D, Zhang S, Chen Y, Chen Y, Feng K, Li X, Han J, Iwakiri Y, Duan Y, Yang X. Inhibition of high-fat diet-induced obesity via reduction of ER-resident protein Nogo occurs through multiple mechanisms. J Biol Chem 2022; 298:101561. [PMID: 34998825 PMCID: PMC8814669 DOI: 10.1016/j.jbc.2022.101561] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a risk factor for insulin resistance, type 2 diabetes, and cardiovascular diseases. Reticulon-4 (Nogo) is an endoplasmic reticulum–resident protein with unclear functions in obesity. Herein, we investigated the effect of Nogo on obesity and associated metabolic disorders. Human serum samples were collected to explore the relationship between circulating Nogo-B and body mass index value. Nogo-deficient and WT littermate control mice were fed normal chow or high-fat diet (HFD) for 14 weeks, and HFD-induced obese C57BL/6J mice were injected scrambled or Nogo siRNA for 2 weeks. We found that in human and mouse serum, Nogo-B was positively correlated to body mass index/bodyweight and lipid profiles. Reduced Nogo (by genetic deletion or siRNA transfection) protected mice against HFD-induced obesity and related metabolic disorders. We demonstrate that Nogo deficiency reversed HFD-induced whitening of brown adipose tissue, thereby increasing thermogenesis. It also ameliorated lipid accumulation in tissues by activating the adiponectin–adiponectin receptor 1–AMP-activated kinase α signaling axis. Finally, Nogo deficiency potently reduced HFD-induced serum proinflammatory cytokines and infiltration of macrophages into metabolic organs, which is related to enhanced NF-κB p65 degradation via the lysosome pathway. Collectively, our study suggests that reduced levels of Nogo protect mice against HFD-induced obesity by increasing thermogenesis and energy metabolism while inhibiting NF-κB-mediated inflammation. Our results indicate that inhibition of Nogo may be a potential strategy for obesity treatment.
Collapse
Affiliation(s)
- Xiaolin Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Yanfang Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Dan Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yi Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Feng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoju Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Jihong Han
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
26
|
Kajani S, Curley S, O'Reilly ME, Yin X, Dillon ET, Guo W, Nilaweera KN, Brennan L, Roche HM, McGillicuddy FC. Sodium salicylate rewires hepatic metabolic pathways in obesity and attenuates IL-1β secretion from adipose tissue - implications for obesity-impaired reverse cholesterol transport. Mol Metab 2021; 56:101425. [PMID: 34954383 PMCID: PMC8762459 DOI: 10.1016/j.molmet.2021.101425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/13/2021] [Accepted: 12/21/2021] [Indexed: 11/26/2022] Open
Abstract
Introduction High-fat diet (HFD)-induced obesity impairs clearance of cholesterol through the Reverse Cholesterol Transport (RCT) pathway, with downregulation in hepatic expression of cholesterol and bile acid transporters, namely ABCG5/8 and ABCB11, and reduced high-density lipoprotein (HDL) cholesterol efflux capacity (CEC). In the current study, we hypothesized that the development of hepatosteatosis, secondary to adipose-tissue dysfunction, contributes to obesity-impaired RCT and that such effects could be mitigated using the anti-inflammatory drug sodium salicylate (NaS). Materials and methods C57BL/6J mice, fed HFD ± NaS or low-fat diet (LFD) for 24 weeks, underwent glucose and insulin tolerance testing. The 3H-cholesterol movement from macrophage-to-feces was assessed in vivo. HDL-CEC was determined ex vivo. Cytokine secretion from adipose-derived stromal vascular fraction (SVF) cells was measured ex vivo. Liver and HDL proteins were determined by mass spectrometry and analyzed using Ingenuity Pathway Analysis. Results NaS delayed HFD-induced weight gain, abrogated priming of pro-IL-1β in SVFs, attenuated insulin resistance, and prevented steatohepatitis (ectopic fat accumulation in the liver). Prevention of hepatosteatosis coincided with increased expression of PPAR-alpha/beta-oxidation proteins with NaS and reduced expression of LXR/RXR-induced proteins including apolipoproteins. The latter effects were mirrored within the HDL proteome in circulation. Despite remarkable protection shown against steatosis, HFD-induced hypercholesterolemia and repression of the liver-to-bile cholesterol transporter, ABCG5/8, could not be rescued with NaS. Discussions and conclusions The cardiometabolic health benefits of NaS may be attributed to the reprogramming of hepatic metabolic pathways to increase fatty acid utilization in the settings of nutritional overabundance. Reduced hepatic cholesterol levels, coupled with reduced LXR/RXR-induced proteins, may underlie the lack of rescue of ABCG5/8 expression with NaS. This remarkable protection against HFD-induced hepatosteatosis did not translate to improvements in cholesterol homeostasis. Sodium salicylate (NaS) initially delays weight-gain in mice fed high-fat diet (HFD) - catch-up evident in weeks 12–24. NaS prevents HFD-induced insulin resistance, hepatosteatosis and pro-IL-1β priming in adipose tissue even upon weight-gain. Hepatic expression of proteins involved in beta oxidation, oxidative phosphorylation and TCA cycle upregulated with NaS. Hepatic expression of LXR/RXR proteins eg. apolipoproteins reduced with NaS; these effects were mirrored in HDL proteome. NaS failed to improve HFD-impaired Reverse Cholesterol Transport or hypercholesterolemia despite preventing hepatosteatosis.
Collapse
Affiliation(s)
- Sarina Kajani
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health
| | - Sean Curley
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health
| | - Marcella E O'Reilly
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health
| | - Xiaofei Yin
- UCD Conway Institute; UCD Institute of Food and Health; School of Agriculture and Food Science, University College Dublin, Dublin 4, Ireland
| | | | - Weili Guo
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health
| | - Kanishka N Nilaweera
- Teagasc Food Research Centre; VistaMilk Research Centre, Moorepark, Fermoy, Ireland
| | - Lorraine Brennan
- UCD Conway Institute; UCD Institute of Food and Health; School of Agriculture and Food Science, University College Dublin, Dublin 4, Ireland
| | - Helen M Roche
- Diabetes Complications Research Centre; UCD Conway Institute; Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science; UCD Institute of Food and Health
| | - Fiona C McGillicuddy
- Diabetes Complications Research Centre; UCD School of Medicine; UCD Conway Institute; UCD Institute of Food and Health.
| |
Collapse
|
27
|
Karampatsi D, Zabala A, Wilhelmsson U, Dekens D, Vercalsteren E, Larsson M, Nyström T, Pekny M, Patrone C, Darsalia V. Diet-induced weight loss in obese/diabetic mice normalizes glucose metabolism and promotes functional recovery after stroke. Cardiovasc Diabetol 2021; 20:240. [PMID: 34937562 PMCID: PMC8697500 DOI: 10.1186/s12933-021-01426-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Post-stroke functional recovery is severely impaired by type 2 diabetes (T2D). This is an important clinical problem since T2D is one of the most common diseases. Because weight loss-based strategies have been shown to decrease stroke risk in people with T2D, we aimed to investigate whether diet-induced weight loss can also improve post-stroke functional recovery and identify some of the underlying mechanisms. METHODS T2D/obesity was induced by 6 months of high-fat diet (HFD). Weight loss was achieved by a short- or long-term dietary change, replacing HFD with standard diet for 2 or 4 months, respectively. Stroke was induced by middle cerebral artery occlusion and post-stroke recovery was assessed by sensorimotor tests. Mechanisms involved in neurovascular damage in the post-stroke recovery phase, i.e. neuroinflammation, impaired angiogenesis and cellular atrophy of GABAergic parvalbumin (PV)+ interneurons were assessed by immunohistochemistry/quantitative microscopy. RESULTS Both short- and long-term dietary change led to similar weight loss. However, only the latter enhanced functional recovery after stroke. This effect was associated with pre-stroke normalization of fasting glucose and insulin resistance, and with the reduction of T2D-induced cellular atrophy of PV+ interneurons. Moreover, stroke recovery was associated with decreased T2D-induced neuroinflammation and reduced astrocyte reactivity in the contralateral striatum. CONCLUSION The global diabetes epidemic will dramatically increase the number of people in need of post-stroke treatment and care. Our results suggest that diet-induced weight loss leading to pre-stroke normalization of glucose metabolism has great potential to reduce the sequelae of stroke in the diabetic population.
Collapse
MESH Headings
- Animals
- Behavior, Animal
- Biomarkers/blood
- Blood Glucose/metabolism
- Brain/metabolism
- Brain/pathology
- Brain/physiopathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/diet therapy
- Diabetes Mellitus, Type 2/physiopathology
- Diet, High-Fat
- Disease Models, Animal
- Glycemic Control
- Infarction, Middle Cerebral Artery/blood
- Infarction, Middle Cerebral Artery/diet therapy
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Male
- Mice, Inbred C57BL
- Obesity/blood
- Obesity/diet therapy
- Obesity/physiopathology
- Recovery of Function
- Stroke/blood
- Stroke/diet therapy
- Stroke/pathology
- Stroke/physiopathology
- Time Factors
- Weight Loss
- Mice
Collapse
Affiliation(s)
- Dimitra Karampatsi
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Alexander Zabala
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Ulrika Wilhelmsson
- Laboratory of Astrocyte Biology and CNS Regeneration, Department of Clinical Neuroscience and Rehabilitation, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Doortje Dekens
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Ellen Vercalsteren
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Martin Larsson
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Thomas Nyström
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Department of Clinical Neuroscience and Rehabilitation, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cesare Patrone
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden.
| | - Vladimer Darsalia
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, 118 83, Stockholm, Sweden.
| |
Collapse
|
28
|
Kovačević S, Brkljačić J, Vojnović Milutinović D, Gligorovska L, Bursać B, Elaković I, Djordjevic A. Fructose Induces Visceral Adipose Tissue Inflammation and Insulin Resistance Even Without Development of Obesity in Adult Female but Not in Male Rats. Front Nutr 2021; 8:749328. [PMID: 34869524 PMCID: PMC8632624 DOI: 10.3389/fnut.2021.749328] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Introduction: Obesity and related metabolic disturbances are frequently related to modern lifestyle and are characterized by excessive fructose intake. Visceral adipose tissue (VAT) inflammation has a central role in the development of insulin resistance, type 2 diabetes (T2D), and metabolic syndrome. Since sex-related differences in susceptibility and progression of metabolic disorders are not yet fully understood, our aim was to examine inflammation and insulin signaling in VAT of fructose-fed female and male adult rats. Methods: We analyzed effects of 9-week 10% fructose-enriched diet on energy intake, VAT mass and histology, and systemic insulin sensitivity. VAT insulin signaling and markers of VAT inflammation, and antioxidative defense status were also evaluated. Results: The fructose diet had no effect on VAT mass and systemic insulin signaling in the female and male rats, while it raised plasma uric acid, increased PPARγ level in the VAT, and initiated the development of a distinctive population of small adipocytes in the females. Also, adipose tissue insulin resistance, evidenced by increased PTP1B and insulin receptor substrate 1 (IRS1) inhibitory phosphorylation and decreased Akt activity, was detected. In addition, fructose stimulated the nuclear accumulation of NFκB, increased expression of proinflammatory cytokines (IL-1β, IL-6, and TNFα), and protein level of macrophage marker F4/80, superoxide dismutase 1, and glutathione reductase. In contrast to the females, the fructose diet had no effect on plasma uric acid and VAT inflammation in the male rats, but less prominent alterations in VAT insulin signaling were observed. Conclusion: Even though dietary fructose did not elicit changes in energy intake and led to obesity in the females, it initiated the proliferation of small-sized adipocytes capable of storing fats further. In contrast to the males, this state of VAT was accompanied with enhanced inflammation, which most likely contributed to the development of insulin resistance. The observed distinction could possibly originate from sex-related differences in uric acid metabolism. Our results suggest that VAT inflammation could precede obesity and start even before the measurable increase in VAT mass, making it a silent risk factor for the development of T2D. Our results emphasize that adipose tissue dysfunction, rather than its simple enlargement, could significantly contribute to the onset and development of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Sanja Kovačević
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Brkljačić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Vojnović Milutinović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ljupka Gligorovska
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Biljana Bursać
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivana Elaković
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
29
|
Innate-Immunity Genes in Obesity. J Pers Med 2021; 11:jpm11111201. [PMID: 34834553 PMCID: PMC8623883 DOI: 10.3390/jpm11111201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 01/07/2023] Open
Abstract
The main functions of adipose tissue are thought to be storage and mobilization of the body’s energy reserves, active and passive thermoregulation, participation in the spatial organization of internal organs, protection of the body from lipotoxicity, and ectopic lipid deposition. After the discovery of adipokines, the endocrine function was added to the above list, and after the identification of crosstalk between adipocytes and immune cells, an immune function was suggested. Nonetheless, it turned out that the mechanisms underlying mutual regulatory relations of adipocytes, preadipocytes, immune cells, and their microenvironment are complex and redundant at many levels. One possible way to elucidate the picture of adipose-tissue regulation is to determine genetic variants correlating with obesity. In this review, we examine various aspects of adipose-tissue involvement in innate immune responses as well as variants of immune-response genes associated with obesity.
Collapse
|
30
|
Postler TS, Peng V, Bhatt DM, Ghosh S. Metformin selectively dampens the acute inflammatory response through an AMPK-dependent mechanism. Sci Rep 2021; 11:18721. [PMID: 34548527 PMCID: PMC8455559 DOI: 10.1038/s41598-021-97441-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/25/2021] [Indexed: 12/30/2022] Open
Abstract
Metformin is a first-line drug in the treatment of type-2 diabetes mellitus (T2DM). In addition to its antigluconeogenic and insulin-sensitizing properties, metformin has emerged as a potent inhibitor of the chronic inflammatory response of macrophages. In particular, metformin treatment has been shown to reduce expression of interleukin (IL-) 1β during long-term exposure to the pro-inflammatory stimulus lipopolysaccharide (LPS) through a reduction in reactive oxygen species (ROS), which decreases the levels of the hypoxia-inducible factor (HIF) 1-α, and through enhanced expression of IL-10. However, the effect of metformin on the acute inflammatory response, before significant levels of ROS accumulate in the cell, has not been explored. Here, we show that metformin alters the acute inflammatory response through its activation of AMP-activated protein kinase (AMPK), but independently of HIF1-α and IL-10, in primary macrophages and two macrophage-like cell lines. Thus, metformin changes the acute and the chronic inflammatory response through fundamentally distinct mechanisms. Furthermore, RNA-seq analysis reveals that metformin pretreatment affects the levels of a large yet selective subset of inflammatory genes, dampening the response to short-term LPS exposure and affecting a wide range of pathways and biological functions. Taken together, these findings reveal an unexpected complexity in the anti-inflammatory properties of this widely used drug.
Collapse
Affiliation(s)
- Thomas S Postler
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Vincent Peng
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Dev M Bhatt
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Amgen Research Oncology and Inflammation, South San Francisco, CA, 94080, USA
| | - Sankar Ghosh
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
31
|
Della Guardia L, Codella R. Exercise tolls the bell for key mediators of low-grade inflammation in dysmetabolic conditions. Cytokine Growth Factor Rev 2021; 62:83-93. [PMID: 34620559 DOI: 10.1016/j.cytogfr.2021.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/30/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022]
Abstract
Metabolic conditions share a common low-grade inflammatory milieu, which represents a key-factor for their ignition and maintenance. Exercise is instrumental for warranting systemic cardio-metabolic balance, owing to its regulatory effect on inflammation. This review explores the effect of physical activity in the modulation of sub-inflammatory framework characterizing dysmetabolic conditions. Regular exercise suppresses plasma levels of TNFα, IL-1β, FFAs and MCP-1, in dysmetabolic subjects. In addition, a single session of training increases the anti-inflammatory IL-10, IL-1 receptor antagonist (IL-1ra), and muscle-derived IL-6, mitigating low-grade inflammation. Resting IL-6 levels are decreased in trained-dysmetabolic subjects, compared to sedentary. On the other hand, the acute release of muscle-IL-6, after exercise, seems to exert a regulatory effect on the metabolic and inflammatory balance. In fact, muscle-released IL-6 is presumably implicated in fat loss and boosts plasma levels of IL-10 and IL-1ra. The improvement of adipose tissue functionality, following regular exercise, is also critical for the mitigation of sub-inflammation. This effect is likely mediated by muscle-released IL-15 and IL-6 and partly relies on the brown-shifting of white adipocytes, induced by exercise. In obese-dysmetabolic subjects, moderate training is shown to restore gut-microbiota health, and this mitigates the translocation of bacterial-LPS into bloodstream. Finally, regular exercise can lower plasma advanced glycated endproducts. The articulated physiology of circulating mediators and the modulating effect of the pathophysiological background, render the comprehension of the exercise-regulatory effect on sub-inflammation a key issue, in dysmetabolism.
Collapse
Affiliation(s)
- Lucio Della Guardia
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Fratelli Cervi 93, Segrate, 20090 Milano, Italy
| | - Roberto Codella
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Fratelli Cervi 93, Segrate, 20090 Milano, Italy; Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milano, Italy.
| |
Collapse
|
32
|
Chen X, Zhang D, Li Y, Wang W, Bei W, Guo J. NLRP3 inflammasome and IL-1β pathway in type 2 diabetes and atherosclerosis: Friend or foe? Pharmacol Res 2021; 173:105885. [PMID: 34536551 DOI: 10.1016/j.phrs.2021.105885] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/23/2021] [Accepted: 09/09/2021] [Indexed: 12/28/2022]
Abstract
Type 2 diabetes and atherosclerosis have gradually garnered great attention as inflammatory diseases. Previously, the fact that Interleukin-1β (IL-1β) accelerates the development of type 2 diabetes and atherosclerosis has been proved in animal experiments and clinical trials. However, the continued studies found that the effect of IL-1β on type 2 diabetes and atherosclerosis is much more complicated than the negative impact. Nucleotide-binding oligomerization domain and leucine-rich repeat pyrin 3 domain (NLRP3) inflammasome, whose activation and assembly significantly affect the release of IL-1β, is a crucial effector activated by a variety of metabolites. The diversity of NLRP3 activation mode is one of the fundamental reasons for the intricate effects on the progression of type 2 diabetes and atherosclerosis, providing many new insights for us to intervene in metabolic diseases. This review focuses on how NLRP3 inflammasome affects the progression of type 2 diabetes and atherosclerosis and what opportunities and challenges it can bring us.
Collapse
Affiliation(s)
- Xu Chen
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangzhou, China
| | - Dongxing Zhang
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangzhou, China
| | - Yuping Li
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangzhou, China
| | - Weixuan Wang
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangzhou, China
| | - Weijian Bei
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangzhou, China.
| | - Jiao Guo
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangzhou, China.
| |
Collapse
|
33
|
Abstract
In this review, Lee and Olefsky discuss the characteristics of chronic inflammation in the major metabolic tissues and how obesity triggers these events, including a focus on the role of adipose tissue hypoxia and macrophage-derived exosomes. Obesity is the most common cause of insulin resistance, and the current obesity epidemic is driving a parallel rise in the incidence of T2DM. It is now widely recognized that chronic, subacute tissue inflammation is a major etiologic component of the pathogenesis of insulin resistance and metabolic dysfunction in obesity. Here, we summarize recent advances in our understanding of immunometabolism. We discuss the characteristics of chronic inflammation in the major metabolic tissues and how obesity triggers these events, including a focus on the role of adipose tissue hypoxia and macrophage-derived exosomes. Last, we also review current and potential new therapeutic strategies based on immunomodulation.
Collapse
Affiliation(s)
- Yun Sok Lee
- Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Diego, La Jolla, California 92093, USA
| | - Jerrold Olefsky
- Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
34
|
Martínez-Chacón G, Yatkin E, Polari L, Deniz Dinç D, Peuhu E, Hartiala P, Saarinen N, Mäkelä S. CC chemokine ligand 2 (CCL2) stimulates aromatase gene expression in mammary adipose tissue. FASEB J 2021; 35:e21536. [PMID: 33913559 DOI: 10.1096/fj.201902485rrr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/10/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Obesity is a risk factor for postmenopausal breast cancer. Obesity-related inflammation upregulates aromatase expression, the rate-limiting enzyme for estrogen synthesis, in breast adipose tissue (BAT), increasing estrogen production and cancer risk. The regulation of aromatase gene (CYP19A1) in BAT is complex, and the mechanisms linking obesity and aromatase dysregulation are not fully understood. An obesity-associated factor that could regulate aromatase is the CC chemokine ligand (CCL) 2, a pro-inflammatory factor that also activates signaling pathways implicated in CYP19A1 transcription. By using human primary breast adipose stromal cells (ASCs) and aromatase reporter (hARO-Luc) mouse mammary adipose explants, we demonstrated that CCL2 enhances the glucocorticoid-mediated CYP19A1 transcription. The potential mechanism involves the activation of PI.4 via ERK1/2 pathway. We also showed that CCL2 contributes to the pro-inflammatory milieu and aromatase expression in obesity, evidenced by increased expression of CCL2 and CYP19A1 in mammary tissues from obese hARO-Luc mice, and subcutaneous adipose tissue from obese women. In summary, our results indicate that postmenopausal obesity may promote CCL2 production in BAT, leading to exacerbation of the menopause-related inflammatory state and further stimulation of local aromatase and estrogens. These results provide new insights into the regulation of aromatase and may aid in finding approaches to prevent breast cancer.
Collapse
Affiliation(s)
- Gabriela Martínez-Chacón
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Emrah Yatkin
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,Central Animal Laboratory, University of Turku, Turku, Finland
| | - Lauri Polari
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Defne Deniz Dinç
- Institute of Biomedicine, University of Turku, Turku, Finland.,FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Emilia Peuhu
- Institute of Biomedicine, University of Turku, Turku, Finland.,FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Pauliina Hartiala
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Plastic and General Surgery, Turku University Hospital (TYS), Turku, Finland
| | - Niina Saarinen
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Sari Mäkelä
- Functional Foods Forum, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| |
Collapse
|
35
|
Sultan S. Aberrant expression of proatherogenic cytokines and growth factors in human umbilical vein endothelial cells from newborns of type 2 diabetic women. SAGE Open Med 2021; 9:20503121211026832. [PMID: 34211712 PMCID: PMC8216400 DOI: 10.1177/20503121211026832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/02/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives: This study reports the levels of cytokines, chemokines, and growth factors previously identified as taking part in the pathology of atherosclerosis in human umbilical vein endothelial cells derived from mothers with type 2 diabetes and compares them with those in human umbilical vein endothelial cells derived from healthy mothers under normal glucose conditions. Methods: Cytokine analysis measures of human umbilical vein endothelial cell lysates were obtained using a multiple analyte profiling (xMAP) assay based on magnetic bead-based technology, using the MAGPIX instrument. The correlation between cytokines, chemokines, and growth factors was examined statistically in human umbilical vein endothelial cells derived from mothers with type 2 diabetes. Results: This study showed that the expression of proinflammatory cytokine interleukin-1 alpha was significantly greater in human umbilical vein endothelial cells derived from mothers with type 2 diabetes than those derived from healthy mothers. The protein level of granulocyte colony-stimulating factor was higher in human umbilical vein endothelial cells derived from mothers with type 2 diabetes than those derived from healthy mothers. A significant positive correlation was demonstrated between the protein expression of granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor in human umbilical vein endothelial cells derived from mothers with type 2 diabetes. Conclusion: Diabetes evokes a persistent inflammatory phenotype in human umbilical vein endothelial cells, as indicated by the enhanced production of cytokines and growth factors under normal glucose conditions.
Collapse
Affiliation(s)
- Samar Sultan
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
36
|
Meliț LE, Mărginean CO, Mărginean CD, Săsăran MO. The Peculiar Trialogue between Pediatric Obesity, Systemic Inflammatory Status, and Immunity. BIOLOGY 2021; 10:biology10060512. [PMID: 34207683 PMCID: PMC8229553 DOI: 10.3390/biology10060512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022]
Abstract
Pediatric obesity is not only an energetic imbalance, but also a chronic complex multisystem disorder that might impair both the life length and quality. Its pandemic status should increase worldwide awareness regarding the long-term life-threatening associated complications. Obesity related complications, such as cardiovascular, metabolic, or hepatic ones, affect both short and long-term wellbeing, and they do not spare pediatric subjects, defined as life-threatening consequences of the systemic inflammatory status triggered by the adipose tissue. The energetic imbalance of obesity clearly results in adipocytes hypertrophy and hyperplasia expressing different degrees of chronic inflammation. Adipose tissue might be considered an immune organ due to its rich content in a complex array of immune cells, among which the formerly mentioned macrophages, neutrophils, mast cells, but also eosinophils along with T and B cells, acting together to maintain the tissue homeostasis in normal weight individuals. Adipokines belong to the class of innate immunity humoral effectors, and they play a crucial role in amplifying the immune responses with a subsequent trigger effect on leukocyte activation. The usefulness of complete cellular blood count parameters, such as leukocytes, lymphocytes, neutrophils, erythrocytes, and platelets as predictors of obesity-triggered inflammation, was also proved in pediatric patients with overweight or obesity. The dogma that adipose tissue is a simple energy storage tissue is no longer accepted since it has been proved that it also has an incontestable multifunctional role acting like a true standalone organ resembling to endocrine or immune organs.
Collapse
Affiliation(s)
- Lorena Elena Meliț
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania; (L.E.M.); (C.D.M.)
| | - Cristina Oana Mărginean
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania; (L.E.M.); (C.D.M.)
- Correspondence: ; Tel.: +40-723-278543
| | - Cristian Dan Mărginean
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania; (L.E.M.); (C.D.M.)
| | - Maria Oana Săsăran
- Department of Pediatrics III, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
| |
Collapse
|
37
|
Amaral GA, Alves JD, Honorio-França AC, Fagundes DL, Araujo GG, Lobato NS, Lima VV, Giachini FR. Interleukin 1-beta is Linked to Chronic Low-Grade Inflammation and Cardiovascular Risk Factors in Overweight Adolescents. Endocr Metab Immune Disord Drug Targets 2021; 20:887-894. [PMID: 31738140 DOI: 10.2174/1871530319666191116141159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/12/2019] [Accepted: 10/19/2019] [Indexed: 11/22/2022]
Abstract
AIM We hypothesized that IL-1β concentrations are augmented in overweight adolescents, who do not display metabolic syndrome. Additionally, we aimed to correlate the IL-1β concentrations with several established risk factors for CVD. METHODS Overweight or control subjects, aging from 14-18 years, were classified according to their adjusted body mass index and evaluated for biochemical and anthropometric parameters. The proinflammatory cytokine IL-1β was assessed in the serum. RESULTS Increased body fat percentage, waist circumference, triglycerides, total cholesterol, Very Low-Density Lipoprotein (VLDL) cholesterol, Low-Density Lipoprotein (LDL) cholesterol, Castelli I index, IL-1β, and IL-8 levels, were observed in overweight adolescents. No differences were observed in systolic blood pressure, diastolic blood pressure, glucose or High-Density Lipoprotein (HDL) cholesterol. Positive correlations between IL-1β with anthropometric and or biochemical parameters were found. CONCLUSION In conclusion, increased IL-1β levels correlate to dyslipidemic factors and may further support low-grade inflammation. IL-1β may further predict the early onset of cardiovascular disease in this population, taking into consideration its important regulatory role.
Collapse
Affiliation(s)
- Gisele A Amaral
- Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garcas, MT, Brazil.,UNIVAR – Araguaia Valley University Center, Barra do Garças, MT, Brazil
| | - Josilene D Alves
- Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garcas, MT, Brazil
| | - Adenilda C Honorio-França
- Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garcas, MT, Brazil
| | - Danny L Fagundes
- Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garcas, MT, Brazil
| | - Gabriel Gomes Araujo
- Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garcas, MT, Brazil
| | - Nubia S Lobato
- Department of Biological Sciences, Division of Cardiovascular Physiology, Federal University of Goias, Jatai, Brazil
| | - Victor V Lima
- Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garcas, MT, Brazil
| | - Fernanda R Giachini
- Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garcas, MT, Brazil
| |
Collapse
|
38
|
Sharma BR, Kanneganti TD. NLRP3 inflammasome in cancer and metabolic diseases. Nat Immunol 2021; 22:550-559. [PMID: 33707781 PMCID: PMC8132572 DOI: 10.1038/s41590-021-00886-5] [Citation(s) in RCA: 662] [Impact Index Per Article: 165.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/25/2021] [Indexed: 01/31/2023]
Abstract
The NLRP3 inflammasome is a multimeric cytosolic protein complex that assembles in response to cellular perturbations. This assembly leads to the activation of caspase-1, which promotes maturation and release of the inflammatory cytokines interleukin-1β (IL-1β) and IL-18, as well as inflammatory cell death (pyroptosis). The inflammatory cytokines contribute to the development of systemic low-grade inflammation, and aberrant NLRP3 activation can drive a chronic inflammatory state in the body to modulate the pathogenesis of inflammation-associated diseases. Therefore, targeting NLRP3 or other signaling molecules downstream, such as caspase-1, IL-1β or IL-18, has the potential for great therapeutic benefit. However, NLRP3 inflammasome-mediated inflammatory cytokines play dual roles in mediating human disease. While they are detrimental in the pathogenesis of inflammatory and metabolic diseases, they have a beneficial role in numerous infectious diseases and some cancers. Therefore, fine tuning of NLRP3 inflammasome activity is essential for maintaining proper cellular homeostasis and health. In this Review, we will cover the mechanisms of NLRP3 inflammasome activation and its divergent roles in the pathogenesis of inflammation-associated diseases such as cancer, atherosclerosis, diabetes and obesity, highlighting the therapeutic potential of targeting this pathway.
Collapse
Affiliation(s)
- Bhesh Raj Sharma
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA,Correspondence to: Thirumala-Devi Kanneganti, Department of Immunology, St. Jude Children’s Research Hospital, MS #351, 262 Danny Thomas Place, Memphis TN 38105-3678, Tel: (901) 595-3634; Fax. (901) 595-5766.,
| |
Collapse
|
39
|
Sen ZD, Danyeli LV, Woelfer M, Lamers F, Wagner G, Sobanski T, Walter M. Linking atypical depression and insulin resistance-related disorders via low-grade chronic inflammation: Integrating the phenotypic, molecular and neuroanatomical dimensions. Brain Behav Immun 2021; 93:335-352. [PMID: 33359233 DOI: 10.1016/j.bbi.2020.12.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Insulin resistance (IR) and related disorders, such as T2DM, increase the risk of major depressive disorder (MDD) and vice versa. Current evidence indicates that psychological stress and overeating can induce chronic low-grade inflammation that can interfere with glutamate metabolism in MDD as well as insulin signaling, particularly in the atypical subtype. Here we first review the interactive role of inflammatory processes in the development of MDD, IR and related metabolic disorders. Next, we describe the role of the anterior cingulate cortex in the pathophysiology of MDD and IR-related disorders. Furthermore, we outline how specific clinical features of atypical depression, such as hyperphagia, are more associated with inflammation and IR-related disorders. Finally, we examine the regional specificity of the effects of inflammation on the brain that show an overlap with the functional and morphometric brain patterns activated in MDD and IR-related disorders.
Collapse
Affiliation(s)
- Zümrüt Duygu Sen
- Department of Psychiatry and Psychotherapy, University Tuebingen, Calwerstraße 14, 72076 Tuebingen, Germany; Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany
| | - Lena Vera Danyeli
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Femke Lamers
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Oldenaller 1, 1081 HJ Amsterdam, the Netherlands
| | - Gerd Wagner
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany
| | - Thomas Sobanski
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, Thueringen-Kliniken "Georgius Agricola" GmbH, Rainweg 68, 07318 Saalfeld, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, University Tuebingen, Calwerstraße 14, 72076 Tuebingen, Germany; Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany; Clinical Affective Neuroimaging Laboratory (CANLAB), Leipziger Str. 44, Building 65, 39120 Magdeburg, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany.
| |
Collapse
|
40
|
Gora IM, Ciechanowska A, Ladyzynski P. NLRP3 Inflammasome at the Interface of Inflammation, Endothelial Dysfunction, and Type 2 Diabetes. Cells 2021; 10:314. [PMID: 33546399 PMCID: PMC7913585 DOI: 10.3390/cells10020314] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/24/2021] [Accepted: 01/30/2021] [Indexed: 01/08/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), accounting for 90-95% cases of diabetes, is characterized by chronic inflammation. The mechanisms that control inflammation activation in T2DM are largely unexplored. Inflammasomes represent significant sensors mediating innate immune responses. The aim of this work is to present a review of links between the NLRP3 inflammasome, endothelial dysfunction, and T2DM. The NLRP3 inflammasome activates caspase-1, which leads to the maturation of pro-inflammatory cytokines interleukin 1β and interleukin 18. In this review, we characterize the structure and functions of NLRP3 inflammasome as well as the most important mechanisms and molecules engaged in its activation. We present evidence of the importance of the endothelial dysfunction as the first key step to activating the inflammasome, which suggests that suppressing the NLRP3 inflammasome could be a new approach in depletion hyperglycemic toxicity and in averting the onset of vascular complications in T2DM. We also demonstrate reports showing that the expression of a few microRNAs that are also known to be involved in either NLRP3 inflammasome activation or endothelial dysfunction is deregulated in T2DM. Collectively, this evidence suggests that T2DM is an inflammatory disease stimulated by pro-inflammatory cytokines. Finally, studies revealing the role of glucose concentration in the activation of NLRP3 inflammasome are analyzed. The more that is known about inflammasomes, the higher the chances to create new, effective therapies for patients suffering from inflammatory diseases. This may offer potential novel therapeutic perspectives in T2DM prevention and treatment.
Collapse
Affiliation(s)
- Ilona M. Gora
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4, 02-109 Warsaw, Poland; (A.C.); (P.L.)
| | | | | |
Collapse
|
41
|
Plows JF, Vickers MH, Ganapathy TP, Bridge-Comer PE, Stanley JL, Reynolds CM. Interleukin-1 Receptor-1 Deficiency Impairs Metabolic Function in Pregnant and Non-Pregnant Female Mice. Mol Nutr Food Res 2021; 65:e1900770. [PMID: 31738006 DOI: 10.1002/mnfr.201900770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/03/2019] [Indexed: 01/02/2023]
Abstract
SCOPE Glucose intolerance during pregnancy is associated with short- and long-term maternal and offspring health consequences. In young male mice, knockout of the major pro-inflammatory mediator interleukin-1-receptor-1 (IL1R1) protects against high-fat diet (HFD)-induced glucose intolerance and metabolic dysfunction. This phenotype has not been examined during pregnancy. The hypothesis that IL1R1 depletion will protect females against HFD-induced glucose intolerance and metabolic dysfunction before, during, and post pregnancy is tested. METHODS AND RESULTS C57BL/6J control and IL1R1 knockout (IL1R1-/- ) mice are randomized to either a control diet (10% kcal from fat) or HFD (45% kcal from fat), and three distinct cohorts are established: nulliparous, pregnant, and postpartum females. Contrary to the authors' hypothesis, it is found that IL1R1-/- does not protect against glucose intolerance in nulliparous or pregnant females, and while control HFD animals see a resolution of glucose tolerance postpartum, IL-1R1-/- mice remain impaired. These effects are accompanied by adipocyte hypertrophy, hyperleptinemia, and increased adipose tissue inflammatory gene expression. Maternal genotype differentially affects fetal growth in male and female fetuses, demonstrating sexual dimorphism in this genotype prior to birth. CONCLUSIONS These findings suggest that IL1R1 signaling is important for normal metabolic functioning in females, during and outside of pregnancy.
Collapse
Affiliation(s)
- Jasmine F Plows
- The Liggins Institute, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
- Children's Hospital Los Angeles, Saban Research Institute, 4641 Sunset Blvd, Los Angeles, CA, 90027, USA
| | - Mark H Vickers
- The Liggins Institute, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Thashma P Ganapathy
- The Liggins Institute, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Pania E Bridge-Comer
- The Liggins Institute, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Joanna L Stanley
- The Liggins Institute, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Clare M Reynolds
- The Liggins Institute, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| |
Collapse
|
42
|
Jeevanandam J, Chan YS, Danquah MK. Cytotoxicity and insulin resistance reversal ability of biofunctional phytosynthesized MgO nanoparticles. 3 Biotech 2020; 10:489. [PMID: 33123456 DOI: 10.1007/s13205-020-02480-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/12/2020] [Indexed: 12/22/2022] Open
Abstract
The present study investigates the cytotoxicity of hexagonal MgO nanoparticles synthesized via Amaranthus tricolor leaf extract and spherical MgO nanoparticles synthesized via Amaranthus blitum and Andrographis paniculata leaf extracts. In vitro cytotoxicity analysis showed that the hexagonal MgO nanoparticles synthesized from A. tricolor extract demonstrated the least toxicity to both diabetic and non-diabetic cells at 600 μl/ml dosage. The viability of the diabetic cells (3T3-L1) after incubation with varying dosages of MgO nanoparticles was observed to be 55.3%. The viability of normal VERO cells was 86.6% and this stabilized to about 75% even after exposure to MgO nanoparticles dosage of up to 1000 μl/ml. Colorimetric glucose assay revealed that the A. tricolor extract synthesized MgO nanoparticles resulted in ~ 28% insulin resistance reversal. A reduction in the expression of GLUT4 protein at 54 KDa after MgO nanopaSrticles incubation with diabetic cells was observed via western blot analysis to confirm insulin reversal ability. Fluorescence microscopic analysis with propidium iodide and acridine orange dyes showed the release of reactive oxygen species as a possible mechanism of the cytotoxic effect of MgO nanoparticles. It was inferred that the synergistic effect of the phytochemicals and MgO nanoparticles played a significant role in delivering enhanced insulin resistance reversal capability in adipose cells.
Collapse
Affiliation(s)
- Jaison Jeevanandam
- CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
| | - Yen San Chan
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, CDT 250, 98009 Miri, Sarawak Malaysia
| | - Michael K Danquah
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN 37403 USA
| |
Collapse
|
43
|
Scheithauer TPM, Rampanelli E, Nieuwdorp M, Vallance BA, Verchere CB, van Raalte DH, Herrema H. Gut Microbiota as a Trigger for Metabolic Inflammation in Obesity and Type 2 Diabetes. Front Immunol 2020; 11:571731. [PMID: 33178196 PMCID: PMC7596417 DOI: 10.3389/fimmu.2020.571731] [Citation(s) in RCA: 364] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota has been linked to the development of obesity and type 2 diabetes (T2D). The underlying mechanisms as to how intestinal microbiota may contribute to T2D are only partly understood. It becomes progressively clear that T2D is characterized by a chronic state of low-grade inflammation, which has been linked to the development of insulin resistance. Here, we review the current evidence that intestinal microbiota, and the metabolites they produce, could drive the development of insulin resistance in obesity and T2D, possibly by initiating an inflammatory response. First, we will summarize major findings about immunological and gut microbial changes in these metabolic diseases. Next, we will give a detailed view on how gut microbial changes have been implicated in low-grade inflammation. Lastly, we will critically discuss clinical studies that focus on the interaction between gut microbiota and the immune system in metabolic disease. Overall, there is strong evidence that the tripartite interaction between gut microbiota, host immune system and metabolism is a critical partaker in the pathophysiology of obesity and T2D.
Collapse
Affiliation(s)
- Torsten P M Scheithauer
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, Amsterdam, Netherlands
| | - Elena Rampanelli
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, Amsterdam, Netherlands
| | - Max Nieuwdorp
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, Amsterdam, Netherlands
| | - Bruce A Vallance
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, Vancouver, BC, Canada
| | - C Bruce Verchere
- Department of Surgery, University of British Columbia and BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, Amsterdam, Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
44
|
Sulen A, Islam S, Wolff ASB, Oftedal BE. The prospects of single-cell analysis in autoimmunity. Scand J Immunol 2020; 92:e12964. [PMID: 32869859 DOI: 10.1111/sji.12964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/18/2020] [Accepted: 08/21/2020] [Indexed: 12/29/2022]
Abstract
In the last decade, there has been a tremendous development of technologies focused on analysing various molecular attributes in single cells, with an ever-increasing number of parameters becoming available at the DNA, RNA and protein levels. Much of this progress has involved cells in suspension, but also in situ analysis of tissues has taken great leaps. Paralleling the development in the laboratory, and because of increasing complexity, the analysis of single-cell data is also constantly being updated with new algorithms and analysis platforms. Our immune system shares this complexity, and immunologists have therefore been in the forefront of this technological development. These technologies clearly open new avenues for immunology research, maybe particularly within autoimmunity where the interaction between the faulty immune system and the thymus or the target organ is important. However, the technologies currently available can seem overwhelming and daunting. The aim of this review is to remedy this by giving a balanced overview of the prospects of using single-cell analysis in basal and clinical autoimmunity research, with an emphasis on endocrine autoimmunity.
Collapse
Affiliation(s)
- André Sulen
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Shahinul Islam
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S B Wolff
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Bergithe E Oftedal
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
45
|
Curley S, Gall J, Byrne R, Yvan‐Charvet L, McGillicuddy FC. Metabolic Inflammation in Obesity—At the Crossroads between Fatty Acid and Cholesterol Metabolism. Mol Nutr Food Res 2020; 65:e1900482. [DOI: 10.1002/mnfr.201900482] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/16/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Sean Curley
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| | - Julie Gall
- University of Nice Unité Mixte de Recherce (UMR) Institut National de la Santé et de la Recherche Médicale U1065 062104 Nice Cedex 3 France
| | - Rachel Byrne
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| | - Laurent Yvan‐Charvet
- University of Nice Unité Mixte de Recherce (UMR) Institut National de la Santé et de la Recherche Médicale U1065 062104 Nice Cedex 3 France
| | - Fiona C. McGillicuddy
- Cardiometabolic Research Group UCD Diabetes Complications Research Centre UCD Conway Institute UCD School of Medicine University College Dublin Dublin 4 Ireland
| |
Collapse
|
46
|
Bridge-Comer PE, Plows JF, Ramzan F, Patel R, Ganapathy TP, Stanley JL, Vickers MH, Reynolds CM. Interleukin 1 Receptor 1 Knockout and Maternal High Fat Diet Exposure Induces Sex-Specific Effects on Adipose Tissue Adipogenic and Inflammatory Gene Expression in Adult Mouse Offspring. Front Physiol 2020; 11:601. [PMID: 32655404 PMCID: PMC7324782 DOI: 10.3389/fphys.2020.00601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/14/2020] [Indexed: 12/17/2022] Open
Abstract
Background: The global incidence of obesity continues to rise, increasing the prevalence of metabolic diseases such as insulin resistance, dyslipidemia, and type 2 diabetes mellitus. Low-grade chronic inflammation, associated with the obese state, also contributes to the development of these metabolic comorbidities. Interleukin-1-receptor-1 (IL-1R1), a pro-inflammatory mediator, bridges the metabolic and inflammatory systems. In young male mice, deficiency of IL-1R1 (IL-1R1-/-) paired with a high-fat diet (HFD) offered beneficial metabolic effects, however in female mice, the same pairing led to metabolic dysfunction. Therefore, we examined the contribution of maternal HFD in combination with IL1R1-/- to metabolic health in adult offspring. Methods: Female C57BL/6 and IL-1R1-/- mice were randomly assigned to a control diet (10% kcal from fat) or HFD (45% kcal from fat) 10 days prior to mating and throughout gestation and lactation. Male and female offspring were housed in same-sex pairs post-weaning and maintained on control diets until 16 weeks old. At 15 weeks, an oral glucose tolerance test (OGTT) was performed to assess glucose tolerance. Histological analysis was carried out to assess adipocyte size and gene expression of adipogenic and inflammatory markers were examined. Results: IL-1R1-/- contributed to increased body weight in male and female adult offspring, irrespective of maternal diet. IL-1R1-/- and maternal HFD increased adipocyte size in the gonadal fat depot of female, but not male offspring. In female offspring, there was reduced expression of genes involved in adipogenesis and lipid metabolism in response to IL1R1-/- and maternal HFD. While there was an increase in inflammatory gene expression in response to maternal HFD, this appeared to be reversed in IL1R1-/- female offspring. In male offspring, there was no significant impact on adipogenic or lipid metabolism pathways. There was an increase in inflammatory gene expression in IL1R1-/- male offspring from HFD-fed mothers. Conclusion: This study suggests that IL-1R1 plays a complex and important role in the metabolic health of offspring, impacting adipogenesis, lipogenesis, and inflammation in a sex-specific manner.
Collapse
Affiliation(s)
- Pania E Bridge-Comer
- Developmental Programming Research Group, The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Jasmine F Plows
- Developmental Programming Research Group, The Liggins Institute, The University of Auckland, Auckland, New Zealand.,Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Farha Ramzan
- Developmental Programming Research Group, The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Rachna Patel
- Developmental Programming Research Group, The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Thashma P Ganapathy
- Developmental Programming Research Group, The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Joanna L Stanley
- Developmental Programming Research Group, The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Mark H Vickers
- Developmental Programming Research Group, The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Clare M Reynolds
- Developmental Programming Research Group, The Liggins Institute, The University of Auckland, Auckland, New Zealand.,UCD School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland.,Conway Institute/Institute of Food and Health, University College Dublin, Dublin, Ireland
| |
Collapse
|
47
|
Ralston JC, Mitchelson KAJ, Lynch GM, Tran TTT, Wang H, Strain CR, Lenighan YM, Kennedy EB, Stanton C, McGillicuddy FC, Su Q, O'Toole PW, Roche HM. Microbiome Transfer Partly Overrides Lack of IL-1RI Signaling to Alter Hepatic but not Adipose Tissue Phenotype and Lipid Handling following a High-Fat Diet Challenge. Mol Nutr Food Res 2020; 65:e2000202. [PMID: 32558187 DOI: 10.1002/mnfr.202000202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/27/2020] [Indexed: 01/05/2023]
Abstract
SCOPE IL-1RI-mediated inflammatory signaling alters metabolic tissue responses to dietary challenges (e.g., high-fat diet [HFD]). Recent work suggests that metabolic phenotype is transferrable between mice in a shared living environment (i.e., co-housing) due to gut microbiome exchange. The authors examine whether the metabolic phenotype of IL-1RI-/- mice fed HFD or low-fat diet (LFD) could be transferred to wild-type (WT) mice through gut microbiome exchange facilitated by co-housing. METHODS AND RESULTS Male WT (C57BL/J6) and IL-1RI-/- mice are fed HFD (45% kcal) or LFD (10% kcal) for 24 weeks and housed i) by genotype (single-housed) or ii) with members of the other genotype in a shared microbial environment (co-housed). The IL-1RI-/- gut microbiome is dominant to WT, meaning that co-housed WT mice adopted the IL-1RI-/- microbiota profile. This is concomitant with greater body weight, hepatic lipid accumulation, adipocyte hypertrophy, and hyperinsulinemia in co-housed WT mice, compared to single-housed counterparts. These effects are most evident following HFD. Primary features of microbiome differences are Lachnospiraceae and Ruminococcaceae (known producers of SCFA). CONCLUSION Transfer of SCFA-producing microbiota from IL-1RI-/- mice highlights a new connection between diet, inflammatory signaling, and the gut microbiome, an association that is dependent on the nature of the dietary fat challenge.
Collapse
Affiliation(s)
- Jessica C Ralston
- Nutrigenomics Research Group and Institute of Food and Health, University College Dublin, Dublin, D04 V1W8, Republic of Ireland
| | - Kathleen A J Mitchelson
- Nutrigenomics Research Group and Institute of Food and Health, University College Dublin, Dublin, D04 V1W8, Republic of Ireland
| | - Gina M Lynch
- Nutrigenomics Research Group and Institute of Food and Health, University College Dublin, Dublin, D04 V1W8, Republic of Ireland
| | - Tam T T Tran
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Republic of Ireland.,School of Microbiology, University College Cork, Cork, T12 T656, Republic of Ireland
| | - Hao Wang
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, United Kingdom
| | - Conall R Strain
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Republic of Ireland.,Teagasc Food Research Centre, Moorepark, Cork, P61 C996, Republic of Ireland
| | - Yvonne M Lenighan
- Nutrigenomics Research Group and Institute of Food and Health, University College Dublin, Dublin, D04 V1W8, Republic of Ireland
| | - Elaine B Kennedy
- Nutrigenomics Research Group and Institute of Food and Health, University College Dublin, Dublin, D04 V1W8, Republic of Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Republic of Ireland.,Teagasc Food Research Centre, Moorepark, Cork, P61 C996, Republic of Ireland
| | - Fiona C McGillicuddy
- Nutrigenomics Research Group and Institute of Food and Health, University College Dublin, Dublin, D04 V1W8, Republic of Ireland.,Diabetes Complications Research Centre, University College Dublin, Dublin, D04 V1W8, Republic of Ireland
| | - Qiaozhu Su
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, United Kingdom
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Republic of Ireland.,School of Microbiology, University College Cork, Cork, T12 T656, Republic of Ireland
| | - Helen M Roche
- Nutrigenomics Research Group and Institute of Food and Health, University College Dublin, Dublin, D04 V1W8, Republic of Ireland.,Diabetes Complications Research Centre, University College Dublin, Dublin, D04 V1W8, Republic of Ireland.,The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, United Kingdom
| |
Collapse
|
48
|
Ying W, Fu W, Lee YS, Olefsky JM. The role of macrophages in obesity-associated islet inflammation and β-cell abnormalities. Nat Rev Endocrinol 2020; 16:81-90. [PMID: 31836875 PMCID: PMC8315273 DOI: 10.1038/s41574-019-0286-3] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/16/2022]
Abstract
Chronic, unresolved tissue inflammation is a well-described feature of obesity, type 2 diabetes mellitus (T2DM) and other insulin-resistant states. In this context, adipose tissue and liver inflammation have been particularly well studied; however, abundant evidence demonstrates that inflammatory processes are also activated in pancreatic islets from obese animals and humans with obesity and/or T2DM. In this Review, we focus on the characteristics of immune cell-mediated inflammation in islets and the consequences of this with respect to β-cell function. In contrast to type 1 diabetes mellitus, the dominant immune cell type causing inflammation in obese and T2DM islets is the macrophage. The increased macrophage accumulation in T2DM islets primarily arises through local proliferation of resident macrophages, which then provide signals (such as platelet-derived growth factor) that drive β-cell hyperplasia (a classic feature of obesity). In addition, islet macrophages also impair the insulin secretory capacity of β-cells. Through these mechanisms, islet-resident macrophages underlie the inflammatory response in obesity and mechanistically participate in the β-cell hyperplasia and dysfunction that characterizes this insulin-resistant state. These findings point to the possibility of therapeutics that target islet inflammation to elicit beneficial effects on β-cell function and glycaemia.
Collapse
Affiliation(s)
- Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Wenxian Fu
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Yun Sok Lee
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jerrold M Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
49
|
Pirzada RH, Javaid N, Choi S. The Roles of the NLRP3 Inflammasome in Neurodegenerative and Metabolic Diseases and in Relevant Advanced Therapeutic Interventions. Genes (Basel) 2020; 11:E131. [PMID: 32012695 PMCID: PMC7074480 DOI: 10.3390/genes11020131] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
Inflammasomes are intracellular multiprotein complexes in the cytoplasm that regulate inflammation activation in the innate immune system in response to pathogens and to host self-derived molecules. Recent advances greatly improved our understanding of the activation of nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasomes at the molecular level. The NLRP3 belongs to the subfamily of NLRP which activates caspase 1, thus causing the production of proinflammatory cytokines (interleukin 1β and interleukin 18) and pyroptosis. This inflammasome is involved in multiple neurodegenerative and metabolic disorders including Alzheimer's disease, multiple sclerosis, type 2 diabetes mellitus, and gout. Therefore, therapeutic targeting to the NLRP3 inflammasome complex is a promising way to treat these diseases. Recent research advances paved the way toward drug research and development using a variety of machine learning-based and artificial intelligence-based approaches. These state-of-the-art approaches will lead to the discovery of better drugs after the training of such a system.
Collapse
Affiliation(s)
| | | | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (R.H.P.); (N.J.)
| |
Collapse
|
50
|
Microbiome transfer between IL-1RI-/- and wild-type mice during high or low-fat feeding alters metabolic tissue functionality but not glucose homeostasis. Proc Nutr Soc 2020. [DOI: 10.1017/s0029665120000403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractReduced inflammatory signaling (IL-1RI-/-) alters metabolic responses to dietary challenges (1). Inflammasome deficiency (e.g. IL-18-/-, Asc-/-) can modify gut microbiota concomitant with hepatosteatosis; an effect that was transferable to wild-type (WT) mice by co-housing (2). Taken together, this evidence suggests that links between diet, microbiota and IL-1RI-signaling can influence metabolic health. Our aim was to determine whether IL-1RI-mediated signaling interacted with the gut microbiome to impact metabolic tissue functionality in a diet-specific fashion. Male WT (C57BL/J6) and IL-1RI-/- mice were fed either high-fat diet (HFD; 45% kcal) or low-fat diet (LFD; 10% kcal) for 24 weeks and were housed i) separately by genotype or ii) with genotypes co-housed together (i.e. isolated vs shared microbial environment; n = 8–10 mice per group). Glucose tolerance and insulin secretion response (1.5 g/kg i.p.), gut microbiota composition and caecal short-chain fatty acids (SCFA) were assessed. Liver and adipose tissue were harvested and examined for triacylglycerol (TAG) formation, cholesterol and metabolic markers (Fasn, Cpt1α, Pparg, Scd1, Dgat1/2), using histology, gas-chromatography and RT-PCR, respectively. Statistical analysis included 1-way or 2-way ANOVA, where appropriate, with Bonferroni post-hoc correction. Co-housing significantly affected gut microbiota composition, illustrated by clustering in PCoA (unweighted UniFrac distance) of co-housed mice but not their single-housed counterparts, on both HFD and LFD. The taxa driving these differences were primarily from Lachnospiraceae and Ruminococcaceae families. Single-housed WT had lower hepatic weight, TAG, cholesterol levels and Fasn despite HFD, an effect lost in their co-housed counterparts, who aligned more to IL-1RI-/- hepatic lipid status. Hepatic Cpt1α was lowest in co-housed WT. Adipose from IL-1RI-/- groups on HFD displayed increased adipocyte size and reduced adipocyte number compared to WT groups, but greater lipogenic potential (Pparg, Scd1, Dgat2) alongside a blunted IL-6 response to pro-inflammatory stimuli (~32%, P = 0.025). Whilst caecal SCFA concentrations were not different between groups, single-housed IL-1RI-/- adipocytes showed greatest sensitivity to SCFA-induced lipogenesis. Interestingly, differences in tissue functionality and gut microbiome occurred despite unaltered glucose tolerance; although there was a trend for phenotypic transfer of body weight via co-housing. For all endpoints examined, similar genotype/co-housing effects were observed for both HFD and LFD with the greatest impacts seen in HFD-fed mice. In conclusion, while the gut microbiome may be an important consideration in dietary interventions, these results question the magnitude of its impact in relation to the IL-1RI-dependent immunometabolism-glucose homeostasis axis.
Collapse
|