1
|
Orioli L, Thissen JP. Myokines as potential mediators of changes in glucose homeostasis and muscle mass after bariatric surgery. Front Endocrinol (Lausanne) 2025; 16:1554617. [PMID: 40171198 PMCID: PMC11958187 DOI: 10.3389/fendo.2025.1554617] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Myokines are bioactive peptides released by skeletal muscle. Myokines exert auto-, para-, or endocrine effects, enabling them to regulate many aspects of metabolism in various tissues. However, the contribution of myokines to the dramatic changes in glucose homeostasis and muscle mass induced by bariatric surgery has not been established. Our review highlights that myokines such as brain-derived neurotrophic factor (BDNF), meteorin-like protein (Metrnl), secreted protein acidic and rich in cysteine (SPARC), apelin (APLN) and myostatin (MSTN) may mediate changes in glucose homeostasis and muscle mass after bariatric surgery. Our review also identifies myonectin as an interesting candidate for future studies, as this myokine may regulate lipid metabolism and muscle mass after bariatric surgery. These myokines may provide novel therapeutic targets and biomarkers for obesity, type 2 diabetes and sarcopenia.
Collapse
Affiliation(s)
- Laura Orioli
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
2
|
Pérez-Mingan GC, Sierra-Merlano RM, Yepes I, Vergara MJP, Ortiz M, Peña B, Cano-Pérez E, Gómez-Camargo D. Relationship of Interleukin 6 with Hepatic Steatosis and Liver Fibrosis in Rheumatoid Arthritis at a Rheumatology Care Center in Cartagena, Colombia. Genes (Basel) 2024; 15:1639. [PMID: 39766906 PMCID: PMC11675702 DOI: 10.3390/genes15121639] [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: 11/22/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to investigate the association of IL-6 with steatotic liver disease (SLD) and liver fibrosis (LF) in rheumatoid arthritis (RA) patients at a rheumatology center in Cartagena de Indias, Colombia. METHODS This was a cross-sectional study that included RA and non-RA cases. The level of cellular expression of interleukin 6 (IL-6) was evaluated by flow cytometry in peripheral blood leukocytes, and the presence of SLD and LF was detected by elastosonography. The main outcome was to establish the association between the levels of cellular expression of IL-6 and the development of SLD and LF. RESULTS This study included 47 cases of RA and 34 cases on-RA, with a mean age of 54 and 55 years, respectively. The frequency of SLD was 55.3% in RA and 38.2% in non-RA. The frequency of LF was 12.8% in RA and 14.7% in non-RA, with no statistical difference. The levels of cellular expression of IL-6 were significantly higher in RA compared to non-RA. Cellular expression of IL-6 was associated with the presence of SLD (54% vs. 30.3%; p = 0.002). This association was not maintained in RA cases (49.5% vs. 47.6%; p = 0.571). No association was found between cellular expression of IL-6 and LF in the total population (43.8% vs. 42.7%; p = 0.813) nor in RA cases (59.41% vs. 48.3%; p = 0.526). CONCLUSIONS IL-6 levels were related to SLD in the evaluated sample, and RA was not a risk factor for SLD or LF. The prognostic role of IL-6 for SLD in patients with RA requires further studies.
Collapse
Affiliation(s)
- Gloria Caterine Pérez-Mingan
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia; (G.C.P.-M.); (R.M.S.-M.); (I.Y.)
| | - Rita Magola Sierra-Merlano
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia; (G.C.P.-M.); (R.M.S.-M.); (I.Y.)
| | - Ismael Yepes
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia; (G.C.P.-M.); (R.M.S.-M.); (I.Y.)
| | | | - Miguel Ortiz
- Programa de Medicina, Facultad de Ciencias de la Salud, Universidad del Sinú, Cartagena 130001, Colombia;
| | - Breiner Peña
- Programa de Medicina, Facultad de Ciencias de la Salud, Universidad del Magdalena, Santa Marta 470001, Colombia;
| | - Eder Cano-Pérez
- Grupo de Investigación UNIMOL, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia;
- Doctorado en Medicina Tropical, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia
| | - Doris Gómez-Camargo
- Grupo de Investigación UNIMOL, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia;
- Doctorado en Medicina Tropical, Facultad de Medicina, Universidad de Cartagena, Cartagena 130001, Colombia
| |
Collapse
|
3
|
Wang R, Gu M, Zhang Y, Zhong Q, Chen L, Li D, Xie Z. Long-term drinking of green tea combined with exercise improves hepatic steatosis and obesity in male mice induced by high-fat diet. Food Sci Nutr 2024; 12:776-785. [PMID: 38370081 PMCID: PMC10867457 DOI: 10.1002/fsn3.3773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 02/20/2024] Open
Abstract
Dietary habits and exercise play an important role in the well-being of human health. Currently, how long of drinking tea combined with exercise could efficiently ameliorate hepatic steatosis and obesity still needs to be investigated. Here, short-term and long-term green tea drinking combined with exercise were studied to improve hepatic steatosis and obesity in high-fat diet-induced (HF) mice. Our results showed that Yunkang 10 green tea (GT) combined with exercise (Ex) exhibited synergistic prevention effects on ameliorating hepatic steatosis and obesity. Especially, 22-week intervention with GT or Ex improved all symptoms of obesity, which indicated that long-term intervention exhibited profound preventive effects than the short term. Moreover, the combined intervention of 22 weeks inhibited the activation of NF-κB pathway and the expression of proinflammatory cytokines, which suggests that tea combined exercise may improve liver steatosis mainly by inhibiting inflammation. The key molecules for regulating lipid and glucose metabolism SCD1 were obviously downregulated, and GLU2 and PPARγ were significantly upregulated by GT and exercise in the liver of high-fat diet-induced mice. This study demonstrated that long-term intervention with GT and exercise effectively relieved hepatic steatosis and obesity complications by ameliorating hepatic inflammation, reducing lipid synthesis, and accelerating glucose transport.
Collapse
Affiliation(s)
- Ruru Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and TechnologyAnhui Agricultural UniversityHefeiChina
| | - Mingxing Gu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and TechnologyAnhui Agricultural UniversityHefeiChina
| | - Yanzhong Zhang
- Department of Sports SciencesAnhui Agricultural UniversityHefeiChina
| | - Qinglin Zhong
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and TechnologyAnhui Agricultural UniversityHefeiChina
| | - Linbo Chen
- Tea Research InstituteYunnan Academy of Agricultural SciencesKunmingChina
| | - Daxiang Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and TechnologyAnhui Agricultural UniversityHefeiChina
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and TechnologyAnhui Agricultural UniversityHefeiChina
| |
Collapse
|
4
|
Coptis chinensis, and extracts of guava and mulberry leaves present good inhibiting potential on obesity and associated metabolic disorders in high-fat diet obesity mice model. J Tradit Complement Med 2023; 13:270-276. [PMID: 37128193 PMCID: PMC10148135 DOI: 10.1016/j.jtcme.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/03/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
This study aimed to investigate the anti-obesity effects of Coptis chinensis (CC), BALASAN (combinational guava leaf extract and mulberry leaf extract), and CC/BALASAN (CC/BAL) on high-fat diet-induced obese C57BL/6 mice and to explore possible mediating mechanisms in 3T3-L1 pre-adipocytes. Oil red-O stain was used to test the effects of CC, BALASAN, and CC/BAL on the differentiation of 3T3-L1 pre-adipocytes. Additionally, real-time PCR was used to detect the expression of genes involved in adipocyte differentiation and inflammation-related genes in adipose tissue of mice that were fed a high-fat diet. CC, BALASAN, and CC/BAL inhibited the differentiation of 3T3-L1 pre-adipocytes and exhibited excellent inhibitory ability against the expression of PPARγ and RXRα genes associated with adipocyte differentiation. Replenishing mice with a high-fat diet with CC, BALASAN, and CC/BAL reduced body weight gaining and blood glucose and plasma cholesterol levels. CC also effectively reduced liver weight, whereas BALASAN and CC/BAL had no inhibitory effect. In addition, CC effectively inhibited the expression of C/EBP-α in adipose tissue. Interestingly, BALASAN not only inhibited the expression of C/EBP-α, but also that of PPARγ, RXRα, and TNFα. Such data indicated that CC, BALASAN, and CC/BAL may have potentially beneficial effects against obesity and associated metabolic disorders by down-regulating the PPARγ/RXRα pathway.
Collapse
|
5
|
Li S, Chen L, Lv G. Interleukin-6 Receptor Blockade can Increase the Risk of Nonalcoholic Fatty Liver Disease: Indications From Mendelian Randomization. Front Pharmacol 2022; 13:905936. [PMID: 35747747 PMCID: PMC9209733 DOI: 10.3389/fphar.2022.905936] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Interleukin-6 receptor (IL-6R) blockade has been approved for inflammation-associated diseases and whether it is effective in treating non-alcoholic fatty liver disease (NAFLD) is still unknown. Methods: A target-based Mendelian randomization was performed to appraise whether inhibiting the IL-6 signaling pathway via IL-6R blockade can reduce the risk of NAFLD. The previously established genetic proxy SNP rs2228145 was mainly used to appraise the therapeutic effects and the genetic-predicted circulating IL-6 level was treated as the exposure with ∼30,000 samples. The genetic association between SNP rs2228145 (A > C) and NAFLD was obtained from non-FinnGen GWAS (1,483 cases and 17,781controls) and FinnGen GWAS (894 cases and 217,898 controls). The causal effects were estimated using a Wald ratio method and were combined using a fixed-effects meta-analysis. Furthermore, the SNP rs12048091 was employed as another proxy in the sensitivity analysis. Results: The positive control analysis suggested the SNP rs2228145 can mimic the effects of IL-6R blockade where inhibiting IL-6 signaling can reduce the risk of rheumatoid arthritis [OR = 0.68 (0.58, 0.80)] and coronary heart disease [OR = 0.75 (0.68, 0.84)]. This Mendelian randomization analysis suggested that IL-6R blockade can adversely increase the risk of NAFLD in the non-FinnGen GWAS [OR = 1.99 (1.27, 3.13)] while not significant in the FinnGen consortium. The fixed-effects meta-analysis indicated inhibiting the IL-6 signaling pathway can reduce the risk of NAFLD [OR = 1.80 (1.26, 2.57)]. When including SNP rs12048091 as the genetic instrument, the meta-analysis using two genetic variants also indicated a similar effect on NAFLD [OR = 1.83 (1.32, 2.53)]. There was no heterogeneity in the whole analysis. Conclusion: Our Mendelian randomization suggested inhibiting the IL-6 signaling pathway via IL-6R blockade might increase the risk of NAFLD, suggesting IL-6R should play a protective role in NAFLD.
Collapse
|
6
|
Antuna-Puente B, Fellahi S, McAvoy C, Fève B, Bastard JP. Interleukins in adipose tissue: Keeping the balance. Mol Cell Endocrinol 2022; 542:111531. [PMID: 34910978 DOI: 10.1016/j.mce.2021.111531] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023]
Abstract
The role of the immune system is to defend the host and preserve the functionality in response to stress. This function is not limited to infection or injury as it also plays a role in the response to overnutrition. Indeed, low-grade chronic activation of the immune system associated with overnutrition may be deleterious, contributing importantly to diabetes and long-term complications, such as cardiovascular disorders. Increasing evidence shows that adipose tissue participates in the obesity-related inflammatory response and that interleukins are one of the key players, either as a pro-inflammatory response to the metabolic dysregulation or to restore homeostasis. The crosstalk between adipocytes and immune cells through some important interleukins and their role in metabolic disruption is the topic of this review.
Collapse
Affiliation(s)
- Barbara Antuna-Puente
- Infection Disease Division, Department of Medicine, Queen's University, Kingston, ON, Canada.
| | - Soraya Fellahi
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de Biochimie-pharmacologie-biologie Moléculaire-génétique Médicale, Créteil, France; Sorbonne Université-Inserm, Centre de Recherche Saint-Antoine UMR S_938, 75012, Paris Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Paris, France
| | - Chloé McAvoy
- Unité de Recherche Clinique de L'Est Parisien (URC-Est), Hôpital Saint Antoine, Paris, France
| | - Bruno Fève
- Sorbonne Université-Inserm, Centre de Recherche Saint-Antoine UMR S_938, 75012, Paris Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Paris, France; Assistance Publique- Hôpitaux de Paris -Hôpital Saint-Antoine, Service D'Endocrinologie-Diabétologie, Centre de Référence des Maladies Rares de L'Insulino-Sécrétion et de L'Insulino-Sensibilité (PRISIS), 75012, Paris, France
| | - Jean-Philippe Bastard
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de Biochimie-pharmacologie-biologie Moléculaire-génétique Médicale, Créteil, France; FHU-SENEC, INSERM U955 and Université Paris Est (UPEC), UMR U955, Faculté de Santé, Créteil, France
| |
Collapse
|
7
|
Tetrahydrocurcumin protects against nonalcoholic fatty liver disease by improving lipid metabolism and redox homeostasis. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
8
|
Guo M, Xiang L, Yao J, Zhang J, Zhu S, Wang D, Liu C, Li G, Wang J, Gao Y, Xie C, Ma X, Xu L, Zhou J. Comprehensive Transcriptome Profiling of NAFLD- and NASH-Induced Skeletal Muscle Dysfunction. Front Endocrinol (Lausanne) 2022; 13:851520. [PMID: 35265044 PMCID: PMC8899658 DOI: 10.3389/fendo.2022.851520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), characterized by extensive triglyceride accumulation in hepatocytes, may progress to nonalcoholic steatohepatitis (NASH) with liver fibrosis and inflammation and increase the risk of cirrhosis, cancer, and death. It has been reported that physical exercise is effective in ameliorating NAFLD and NASH, while skeletal muscle dysfunctions, including lipid deposition and weakness, are accompanied with NAFLD and NASH. However, the molecular characteristics and alterations in skeletal muscle in the progress of NAFLD and NASH remain unclear. In the present study, we provide a comprehensive analysis on the similarity and heterogeneity of quadriceps muscle in NAFLD and NASH mice models by RNA sequencing. Importantly, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway functional enrichment analysis revealed that NAFLD and NASH led to impaired glucose and lipid metabolism and deteriorated functionality in skeletal muscle. Besides this, we identified that myokines possibly mediate the crosstalk between muscles and other metabolic organs in pathological conditions. Overall, our analysis revealed a comprehensive understanding of the molecular signature of skeletal muscles in NAFLD and NASH, thus providing a basis for physical exercise as an intervention against liver diseases.
Collapse
Affiliation(s)
- Mingwei Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Liping Xiang
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jing Yao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jun Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Shuangshuang Zhu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Caizhi Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Guoqiang Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiawen Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yuqing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- *Correspondence: Xinran Ma, ; Lingyan Xu, ; Jian Zhou,
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- *Correspondence: Xinran Ma, ; Lingyan Xu, ; Jian Zhou,
| | - Jian Zhou
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Xinran Ma, ; Lingyan Xu, ; Jian Zhou,
| |
Collapse
|
9
|
New insights into IL-6 family cytokines in metabolism, hepatology and gastroenterology. Nat Rev Gastroenterol Hepatol 2021; 18:787-803. [PMID: 34211157 DOI: 10.1038/s41575-021-00473-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
IL-6 family cytokines are defined by the common use of the signal-transducing receptor chain glycoprotein 130 (gp130). Increasing evidence indicates that these cytokines are essential in the regulation of metabolic homeostasis as well as in the pathophysiology of multiple gastrointestinal and liver disorders, thus making them attractive therapeutic targets. Over the past few years, therapies modulating gp130 signalling have grown exponentially in several clinical settings including obesity, cancer and inflammatory bowel disease. A newly engineered gp130 cytokine, IC7Fc, has shown promising preclinical results for the treatment of type 2 diabetes, obesity and liver steatosis. Moreover, drugs that modulate gp130 signalling have shown promise in refractory inflammatory bowel disease in clinical trials. A deeper understanding of the main roles of the IL-6 family of cytokines during homeostatic and pathological conditions, their signalling pathways, sources of production and target cells will be crucial to the development of improved treatments. Here, we review the current state of the role of these cytokines in hepatology and gastroenterology and discuss the progress achieved in translating therapeutics targeting gp130 signalling into clinical practice.
Collapse
|
10
|
Paula VG, Sinzato YK, Moraes Souza RQ, Soares TS, Souza FQG, Karki B, Andrade Paes AM, Corrente JE, Damasceno DC, Volpato GT. Metabolic changes in female rats exposed to intrauterine Hyperglycemia and post-weaning consumption of high-fat diet. Biol Reprod 2021; 106:200-212. [PMID: 34668971 DOI: 10.1093/biolre/ioab195] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/01/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
We evaluated the influence of the hyperglycemic intrauterine environment and post-weaning consumption of a high-fat diet on the glycemia, insulin, lipid and immunological profile of rat offspring in adulthood. Female rats received citrate buffer (Control - C) or Streptozotocin (a beta cell-cytotoxic drug to induce diabetes - D) on post-natal day 5. In adulthood, these rats were mated to obtain female offspring, who were fed a standard diet (SD) or high-fat diet (HFD) from weaning to adulthood (n = 10 rats/group). OC/SD and OC/HFD represent female offspring of control mothers and received SD or HFD, respectively; OD/SD and OD/HFD represent female offspring of diabetic mothers and received SD or HFD, respectively. At adulthood, the Oral Glucose Tolerance Test (OGTT) was performed and, next, the rats were anesthetized and euthanized. Pancreas was collected and analyzed, and adipose tissue was weighted. Blood samples were collected to determine biochemical and immunological profiles. The food intake was lower in HFD-fed rats and visceral fat weight was increased in the OD/HFD group. OC/HFD, OD/SD, and OD/HFD groups presented glucose intolerance and lower insulin secretion during OGTT. An impaired pancreatic beta-cell function was shown in the adult offspring of diabetic rats, regardless of diet. Interleukin (IL)-6 and IL-10 concentrations were lower in the OD/HFD group and associated to a low-grade inflammatory condition. The fetal programming was responsible for impaired beta cell function in experimental animals. The association of maternal diabetes and post-weaning high-fat diet is responsible for greater glucose intolerance, impaired insulin secretion and immunological change.
Collapse
Affiliation(s)
- Verônyca Gonçalves Paula
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil.,Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Rafaianne Queiroz Moraes Souza
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil.,Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Thaigra Souza Soares
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil.,Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Franciane Quintanilha Gallego Souza
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Barshana Karki
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Antonio Marcus Andrade Paes
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão - UFMA -Maranhão State, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Tocogynecology Postgraduate Course, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| |
Collapse
|
11
|
Aguilar-Recarte D, Palomer X, Wahli W, Vázquez-Carrera M. The PPARβ/δ-AMPK Connection in the Treatment of Insulin Resistance. Int J Mol Sci 2021; 22:8555. [PMID: 34445261 PMCID: PMC8395240 DOI: 10.3390/ijms22168555] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
The current treatment options for type 2 diabetes mellitus do not adequately control the disease in many patients. Consequently, there is a need for new drugs to prevent and treat type 2 diabetes mellitus. Among the new potential pharmacological strategies, activators of peroxisome proliferator-activated receptor (PPAR)β/δ show promise. Remarkably, most of the antidiabetic effects of PPARβ/δ agonists involve AMP-activated protein kinase (AMPK) activation. This review summarizes the recent mechanistic insights into the antidiabetic effects of the PPARβ/δ-AMPK pathway, including the upregulation of glucose uptake, muscle remodeling, enhanced fatty acid oxidation, and autophagy, as well as the inhibition of endoplasmic reticulum stress and inflammation. A better understanding of the mechanisms underlying the effects resulting from the PPARβ/δ-AMPK pathway may provide the basis for the development of new therapies in the prevention and treatment of insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- David Aguilar-Recarte
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine of the University of Barcelona (IBUB), Faculty of Pharmacy and Food Sciences, University of Barcelona, Avinguda Joan XXIII 27-31, 08028 Barcelona, Spain; (D.A.-R.); (X.P.)
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine of the University of Barcelona (IBUB), Faculty of Pharmacy and Food Sciences, University of Barcelona, Avinguda Joan XXIII 27-31, 08028 Barcelona, Spain; (D.A.-R.); (X.P.)
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland;
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- ToxAlim (Research Center in Food Toxicology), INRAE, UMR1331, CEDEX, 31300 Toulouse, France
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine of the University of Barcelona (IBUB), Faculty of Pharmacy and Food Sciences, University of Barcelona, Avinguda Joan XXIII 27-31, 08028 Barcelona, Spain; (D.A.-R.); (X.P.)
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
12
|
Pillai VB, Gupta MP. Is nuclear sirtuin SIRT6 a master regulator of immune function? Am J Physiol Endocrinol Metab 2021; 320:E399-E414. [PMID: 33308014 PMCID: PMC7988780 DOI: 10.1152/ajpendo.00483.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022]
Abstract
The ability to ward off pathogens with minimal damage to the host determines the immune system's robustness. Multiple factors, including pathogen processing, identification, secretion of mediator and effector molecules, and immune cell proliferation and differentiation into various subsets, constitute the success of mounting an effective immune response. Cellular metabolism controls all of these intricate processes. Cells utilize diverse fuel sources and switch back and forth between different metabolic pathways depending on their energy needs. The three most critical metabolic pathways on which immune cells depend to meet their energy needs are oxidative metabolism, glycolysis, and glutaminolysis. Dynamic switching between these metabolic pathways is needed for optimal function of the immune cells. Moreover, switching between these metabolic pathways needs to be tightly regulated to achieve the best results. Immune cells depend on the Warburg effect for their growth, proliferation, secretory, and effector functions. Here, we hypothesize that the sirtuin, SIRT6, could be a negative regulator of the Warburg effect. We also postulate that SIRT6 could act as a master regulator of immune cell metabolism and function by regulating critical signaling pathways.
Collapse
Affiliation(s)
- Vinodkumar B Pillai
- Department of Surgery (Division of Cardiothoracic Surgery), Pritzker School of Medicine, Basic Science Division, University of Chicago, Chicago, Illinois
| | - Mahesh P Gupta
- Department of Surgery (Division of Cardiothoracic Surgery), Pritzker School of Medicine, Basic Science Division, University of Chicago, Chicago, Illinois
| |
Collapse
|
13
|
Cao B, Liu C, Zhang Q, Dong Y. Maternal High-Fat Diet Leads to Non-alcoholic Fatty Liver Disease Through Upregulating Hepatic SCD1 Expression in Neonate Rats. Front Nutr 2020; 7:581723. [PMID: 33282902 PMCID: PMC7705221 DOI: 10.3389/fnut.2020.581723] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the leading cause of liver disease in children, with evidence that the maternal diet and the early life nutritional environment are potential risk for such disease. This study was aimed to investigate the effects of maternal high-fat diet (HFD) on the occurrence of NAFLD in offspring rats and the underlying mechanisms. In this study, the incidence of NAFLD was compared in F1 offspring rats between the maternal HFD group and standard chow (SC) group. In addition, the expression levels of inflammatory cytokines in the placenta, in the umbilical cord blood, and in the livers of neonate offsprings were compared between two groups. HepG2 cells were treated with recombinant IL6 (rIL6) to assess stearoyl-CoA desaturase 1 (SCD1) expression and lipid synthesis in an inflammatory condition. Lipid accumulation was assayed in both SCD1 overexpression and interference HepG2 cells as well as in neonatal rats. Our results showed that HFD exposure before and throughout the pregnancy induced the elevated hepatic TG content of F1 neonates. The levels of inflammatory cytokines in the placenta, umbilical cord blood, and the livers of HFD F1 neonates were significantly higher than those of the SC group. In addition, rIL6 treatment led to TG accumulation accompanied by the upregulation of SCD1 in HepG2 cell lines. Overexpression of SCD1 led to the accumulation of TG contents in HepG2 cells, whereas Scd1 knockdown attenuated the effects of rIL6 treatment. Overexpression of SCD1 in F1 neonatal rats led to hepatic lipid accumulation. Our study indicated that maternal HFD led to intrauterine inflammation, which subsequently caused transgenerationally abnormal hepatic lipid metabolism of F1 neonates. This modulation might be mediated by upregulating SCD1 expression in hepatic cells.
Collapse
Affiliation(s)
- Baige Cao
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongxiao Liu
- Department of Endocrinology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qianren Zhang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Dong
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Gong H, Gao J, Wang Y, Luo Q, Guo K, Ren F, Mao X. Identification of novel peptides from goat milk casein that ameliorate high-glucose-induced insulin resistance in HepG2 cells. J Dairy Sci 2020; 103:4907-4918. [DOI: 10.3168/jds.2019-17513] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/04/2020] [Indexed: 12/21/2022]
|
15
|
Wu L, Sun J, Liu L, Du X, Liu Y, Yan X, Kombo Osoro E, Zhang F, Feng L, Liang D, Li Y, Chen Q, Sun S, Zhang L, Lan X, Li D, Lu S. Anti-toll-like receptor 2 antibody ameliorates hepatic injury, inflammation, fibrosis and steatosis in obesity-related metabolic disorder rats via regulating MAPK and NF-κB pathways. Int Immunopharmacol 2020; 82:106368. [PMID: 32151955 DOI: 10.1016/j.intimp.2020.106368] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide, which includes a spectrum of histological liver changes. Non-alcoholic steatohepatitis (NASH) is considered to be the progressive subtype of NAFLD, which is characterized by lobular inflammation and cellular ballooning on the basis of steatosis. There is a critical need to develop novel and effective therapeutic approaches for NAFLD/NASH. The activation of toll-like receptor 2 (TLR2) signaling pathway plays a key role in high-fat-related inflammation, triggering the occurrence and development of NASH. Herein, the anti-TLR2 monoclonal antibody (TLR2 mAb) was prepared and investigated for its ability to ameliorate the inflammatory response in vivo and in vitro. The anti-inflammatory role of TLR2 mAb in vitro was examined in NR8383 macrophage cells and THP-1 derived macrophage cells. For confirmation in vivo, three groups of SD rats were treated for 20 weeks: rats in the control were fed with a standard diet; rates in the IgG and TLR2 mAb groups were fed with a high-fat diet and with IgG or TLR2 mAb, respectively. Liver tissue and serum were collected for further analysis. Results showed that after 4-week treatment with TLR2 mAb, metabolic parameters in rats were improved markedly (body weight, fasting blood glucose level, liver steatosis, inflammatory response and fibrosis). Moreover, western blotting demonstrated that the TLR2 mAb blocked MAPKs and NF-κB activation, and inhibited the expression of inflammatory factors in rat liver tissue. These effects suggested that TLR2 mAb could improve HFD-induced hepatic injury, inflammation, fibrosis and steatosis by suppressing inflammatory response and regulating the hepatic MAPKs and NF-κB signaling pathways. This suggests that TLR2 may be a novel therapeutic target for metabolic diseases especially NASH.
Collapse
Affiliation(s)
- Litao Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Juan Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Li Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China; Department of Pathology, Sunshine Union Hospital, Weifang, Shandong Province 261061, PR China
| | - Xiaojuan Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Yan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Xiaofei Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Ezra Kombo Osoro
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Lina Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Dong Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Yazhao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Qian Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Sha Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Science, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, PR China
| | - Xi Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China.
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China.
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an, Shaanxi 710061, PR China
| |
Collapse
|
16
|
Inhibition of Serine Protease Activity Protects Against High Fat Diet-Induced Inflammation and Insulin Resistance. Sci Rep 2020; 10:1725. [PMID: 32015418 PMCID: PMC6997356 DOI: 10.1038/s41598-020-58361-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 01/13/2020] [Indexed: 12/30/2022] Open
Abstract
Recent evidence suggests that enhanced protease-mediated inflammation may promote insulin resistance and result in diabetes. This study tested the hypothesis that serine protease plays a pivotal role in type 2 diabetes, and inhibition of serine protease activity prevents hyperglycemia in diabetic animals by modulating insulin signaling pathway. We conducted a single-center, cross-sectional study with 30 healthy controls and 57 patients with type 2 diabetes to compare plasma protease activities and inflammation marker between groups. Correlations of plasma total and serine protease activities with variables were calculated. In an in-vivo study, LDLR−/− mice were divided into normal chow diet, high-fat diet (HFD), and HFD with selective serine protease inhibition groups to examine the differences of obesity, blood glucose level, insulin resistance and serine protease activity among groups. Compared with controls, diabetic patients had significantly increased plasma total protease, serine protease activities, and also elevated inflammatory cytokines. Plasma serine protease activity was positively correlated with body mass index, hemoglobin A1c, homeostasis model assessment-insulin resistance index (HOMA-IR), tumor necrosis factor-α, and negatively with adiponectin concentration. In the animal study, administration of HFD progressively increased body weight, fasting glucose level, HOMA-IR, and upregulated serine protease activity. Furthermore, in-vivo serine protease inhibition significantly suppressed systemic inflammation, reduced fasting glucose level, and improved insulin resistance, and these effects probably mediated by modulating insulin receptor and cytokine expression in visceral adipose tissue. Our findings support the serine protease may play an important role in type 2 diabetes and suggest a rationale for a therapeutic strategy targeting serine protease for clinical prevention of type 2 diabetes.
Collapse
|
17
|
Inhibiting Extracellular Cathepsin D Reduces Hepatic Steatosis in Sprague⁻Dawley Rats †. Biomolecules 2019; 9:biom9050171. [PMID: 31060228 PMCID: PMC6571693 DOI: 10.3390/biom9050171] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/28/2019] [Accepted: 05/02/2019] [Indexed: 12/30/2022] Open
Abstract
Dietary and lifestyle changes are leading to an increased occurrence of non-alcoholic fatty liver disease (NAFLD). Using a hyperlipidemic murine model for non-alcoholic steatohepatitis (NASH), we have previously demonstrated that the lysosomal protease cathepsin D (CTSD) is involved with lipid dysregulation and inflammation. However, despite identifying CTSD as a major player in NAFLD pathogenesis, the specific role of extracellular CTSD in NAFLD has not yet been investigated. Given that inhibition of intracellular CTSD is highly unfavorable due to its fundamental physiological function, we here investigated the impact of a highly specific and potent small-molecule inhibitor of extracellular CTSD (CTD-002) in the context of NAFLD. Treatment of bone marrow-derived macrophages with CTD-002, and incubation of hepatic HepG2 cells with a conditioned medium derived from CTD-002-treated macrophages, resulted in reduced levels of inflammation and improved cholesterol metabolism. Treatment with CTD-002 improved hepatic steatosis in high fat diet-fed rats. Additionally, plasma levels of insulin and hepatic transaminases were significantly reduced upon CTD-002 administration. Collectively, our findings demonstrate for the first time that modulation of extracellular CTSD can serve as a novel therapeutic modality for NAFLD.
Collapse
|
18
|
Casagrande BP, Gomes MFP, Moura EOC, Santos ACC, Kubota MC, Ribeiro DA, Pisani LP, Medeiros A, Estadella D. Age-dependent hepatic alterations induced by a high-fat high-fructose diet. Inflamm Res 2019; 68:359-368. [PMID: 30874869 DOI: 10.1007/s00011-019-01223-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE The present study aimed to evaluate and clarify how the age at which the intake of a high-fat and high-fructose diet begins can affect animals' livers. METHODS Thirty-eight male wistar rats aged 6 and 12 weeks were fed a high-fat and high-fructose diet for 13 weeks. Inflammatory cytokines, hepatic glycogen, serum and hepatic triacylglycerol and pAkt protein content in the liver were assessed. Percentage of weight gained, and visceral adiposity were also evaluated. RESULTS Young animal presented increased hepatic triacylglycerol and decreased glycogen, while adult animals had no significant alterations regarding its contents. IL6 and IL10 to IL6 ratio were also altered in young animals exposed to HFHF, while adult animals fed with HFHF had only increases in TNF-α. Both groups which received HFHF had increased serum triacylglycerol and visceral adiposity. However, only young animals gained more relative weight and had greater final body weight, gains which were related to alterations found in hepatic triacylglycerol and glycogen. CONCLUSION Age of which consumption begins interferes in how the liver deals with an excess of nutrient and subsequent proinflammatory stimulation, leading to different phenotypes.
Collapse
Affiliation(s)
- B P Casagrande
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - M F P Gomes
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - E O C Moura
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - A C C Santos
- Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - M C Kubota
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - D A Ribeiro
- Departamento de Biociências, Instituto de Saúde e Sociedade, Federal University of São Paulo (UNIFESP), 11015-020, Santos, SP, Brazil
| | - L P Pisani
- Departamento de Biociências, Instituto de Saúde e Sociedade, Federal University of São Paulo (UNIFESP), 11015-020, Santos, SP, Brazil
| | - A Medeiros
- Departamento de Biociências, Instituto de Saúde e Sociedade, Federal University of São Paulo (UNIFESP), 11015-020, Santos, SP, Brazil
| | - D Estadella
- Departamento de Biociências, Instituto de Saúde e Sociedade, Federal University of São Paulo (UNIFESP), 11015-020, Santos, SP, Brazil.
| |
Collapse
|
19
|
Abstract
In this issue of Immunity, Šestan et al. (2018) show that viral-induced inflammation leads to insulin resistance in skeletal muscle, followed by compensatory hyperinsulinemia, which promotes the anti-viral effector response of CD8+ T cells. Interestingly, this leads to persistent glucose intolerance and the progression of type 2 diabetes in pre-diabetic animals.
Collapse
Affiliation(s)
| | - Nancie J MacIver
- Department of Immunology, Duke University, Durham, NC, USA; Department of Pediatrics, Duke University, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
20
|
Zarzour RHA, Alshawsh MA, Asif M, Al-Mansoub MA, Mohamed Z, Ahmad M, Majid AMSA, Asmawi MZ, Kaur G, Al-Dualimi DW, Yam MF. Adipocytokine Regulation and Antiangiogenic Activity Underlie the Molecular Mechanisms of Therapeutic Effects of Phyllanthus niruri against Non-Alcoholic Fatty Liver Disease. Nutrients 2018; 10:E1057. [PMID: 30096951 PMCID: PMC6115813 DOI: 10.3390/nu10081057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 12/23/2022] Open
Abstract
The growth of adipose tissues is considered angiogenesis-dependent during non-alcoholic fatty liver disease (NAFLD). We have recently reported that our standardized 50% methanolic extract (ME) of Phyllanthus niruri (50% ME of P. niruri) has alleviated NAFLD in Sprague⁻Dawley rats. This study aimed to assess the molecular mechanisms of action, and to further evaluate the antiangiogenic effect of this extract. NAFLD was induced by eight weeks of high-fat diet, and treatment was applied for four weeks. Antiangiogenic activity was assessed by aortic ring assay and by in vitro tests. Our findings demonstrated that the therapeutic effects of 50% ME among NAFLD rats, were associated with a significant increase in serum adiponectin, reduction in the serum levels of RBP4, vaspin, progranulin, TNF-α, IL-6, and significant downregulation of the hepatic gene expression of PPARγ, SLC10A2, and Collα1. Concomitantly, 50% ME of P. niruri has exhibited a potent antiangiogenic activity on ring assay, cell migration, vascular endothelial growth factor (VEGF), and tube formation, without any cytotoxic effect. Together, our findings revealed that the protective effects of P. niruri against NAFLD might be attributed to its antiangiogenic effect, as well as to the regulation of adipocytokines and reducing the expression of adipogenic genes.
Collapse
Affiliation(s)
- Raghdaa Hamdan Al Zarzour
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - Mohammed A Alshawsh
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Muhammad Asif
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Majed Ahmed Al-Mansoub
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Mariam Ahmad
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - Amin Malik Shah Abdul Majid
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - Mohd Zaini Asmawi
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - Dhamraa Waleed Al-Dualimi
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| | - Mun Fei Yam
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Penang, Malaysia.
| |
Collapse
|
21
|
Ghanemi A, St-Amand J. Interleukin-6 as a "metabolic hormone". Cytokine 2018; 112:132-136. [PMID: 29983356 DOI: 10.1016/j.cyto.2018.06.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 06/22/2018] [Accepted: 06/30/2018] [Indexed: 12/11/2022]
Abstract
Interleukin 6 (IL-6) is a cytokine that is involved in divers immune responses and implicated in a number of diseases. However, this cytokine has other non-immune functions. Within this review, we highlight selected effects on metabolic pathways, which are mediated, controlled or modified by the IL-6. Importantly, putting spotlight on such concepts could allow us to classify IL-6 among the metabolic hormones and further study it to both deepen our knowledge on disorders involving metabolic or energy imbalances such as obesity and develop novel therapeutic strategies. Furthermore, potential explanations related to IL-6 roles in both physiology and pathology as well as relevant implications and applications on both research and therapeutic fields are also pointed as consequences of the involvement of IL-6 in the energy and metabolic homeostasis via its "endocrine" roles.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec G1V 0A6, Canada; Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Québec, Québec G1V 4G2, Canada
| | - Jonny St-Amand
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec G1V 0A6, Canada; Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Québec, Québec G1V 4G2, Canada.
| |
Collapse
|
22
|
Blossom SJ, Fernandes L, Bai S, Khare S, Gokulan K, Yuan Y, Dewall M, Simmen FA, Gilbert KM. Opposing Actions of Developmental Trichloroethylene and High-Fat Diet Coexposure on Markers of Lipogenesis and Inflammation in Autoimmune-Prone Mice. Toxicol Sci 2018; 164:313-327. [PMID: 29669109 PMCID: PMC6016708 DOI: 10.1093/toxsci/kfy091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Trichloroethylene (TCE) is a widespread environmental pollutant associated with immunotoxicity and autoimmune disease. Previous studies showed that mice exposed from gestation through early life demonstrated CD4+ T cell alterations and autoimmune hepatitis. Determining the role of one environmental risk factor for any disease is complicated by the presence of other stressors. Based on its known effects, we hypothesized that developmental overnutrition in the form of a moderately high-fat diet (HFD) consisting of 40% kcal fat would exacerbate the immunotoxicity and autoimmune-promoting effects of low-level (<10 μg/kg/day) TCE in autoimmune-prone MRL+/+ mice over either stressor alone. When female offspring were evaluated at 27 weeks of age we found that a continuous exposure beginning at 4 weeks preconception in the dams until 10 weeks of age in offspring that TCE and HFD promoted unique effects that were often antagonistic. For a number of adiposity endpoints, TCE significantly reversed the expected effects of HFD on expression of genes involved in fatty acid synthesis/insulin resistance, as well as mean pathology scores of steatosis. Although none of the animals developed pathological signs of autoimmune hepatitis, the mice generated unique patterns of antiliver antibodies detected by western blotting attributable to TCE exposure. A majority of cytokines in liver, gut, and splenic CD4+ T cells were significantly altered by TCE, but not HFD. Levels of bacterial populations in the intestinal ileum were also altered by TCE exposure rather than HFD. Thus, in contrast to our expectations this coexposure did not promote synergistic effects.
Collapse
Affiliation(s)
- Sarah J Blossom
- Department of Pediatrics, Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72202
| | - Lorenzo Fernandes
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Shasha Bai
- Department of Pediatrics, Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72202
| | - Sangeeta Khare
- Division of Microbiology, National Center for Toxicological Research, U.S. FDA, Jefferson, Arkansas 72079
| | - Kuppan Gokulan
- Division of Microbiology, National Center for Toxicological Research, U.S. FDA, Jefferson, Arkansas 72079
| | | | | | - Frank A Simmen
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Kathleen M Gilbert
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| |
Collapse
|
23
|
Wu R, Liu X, Yin J, Wu H, Cai X, Wang N, Qian Y, Wang F. IL-6 receptor blockade ameliorates diabetic nephropathy via inhibiting inflammasome in mice. Metabolism 2018; 83:18-24. [PMID: 29336982 DOI: 10.1016/j.metabol.2018.01.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 12/24/2017] [Accepted: 01/08/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Interleukin 6 (IL-6) has been identified as a key mediator in inflammation, immune responses and glucose metabolism. In this study, we assessed the effects of an IL-6 receptor antibody on diabetic nephropathy in a mouse model of type 2 diabetes mellitus. METHODS Twelve week old male db/db mice were treated with Tocilizumab (an IL-6 receptor antibody), normal IgG1 control antibody, insulin or normal saline for 12 weeks. Renal injury, inflammation and insulin resistance were assessed. RESULTS Db/db mice treated with Tocilizumab exhibited reduced proteinuria and glomerular mesangial matrix accumulation compared to db/db + IgG controls. Additionally, Tocilizumab suppressed inflammatory response, oxidative stress and the IL-6 signaling pathway in the diabetic kidneys. It is noteworthy that blockade of IL-6 receptor blunted the activation of NLRP3 inflammasome partly through inhibition of IL-17A. Furthermore, insulin resistance assessed by glucose tolerance test, was ameliorated by Tocilizumab treatment. CONCLUSIONS The protective effects of an IL-6 receptor blockade against diabetic renal injury may be due to decreased insulin resistance and inhibition of the inflammasome.
Collapse
Affiliation(s)
- Rui Wu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xuanchen Liu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai 200233, China
| | - Jianyong Yin
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Huijuan Wu
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiulei Cai
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Youcun Qian
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Feng Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Jiangsu University Affiliated Shanghai Eighth People's Hospital, Shanghai 200233, China.
| |
Collapse
|
24
|
Alwarawrah Y, Kiernan K, MacIver NJ. Changes in Nutritional Status Impact Immune Cell Metabolism and Function. Front Immunol 2018; 9:1055. [PMID: 29868016 PMCID: PMC5968375 DOI: 10.3389/fimmu.2018.01055] [Citation(s) in RCA: 326] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
Immune cell function and metabolism are closely linked. Many studies have now clearly demonstrated that alterations in cellular metabolism influence immune cell function and that, conversely, immune cell function determines the cellular metabolic state. Less well understood, however, are the effects of systemic metabolism or whole organism nutritional status on immune cell function and metabolism. Several studies have demonstrated that undernutrition is associated with immunosuppression, which leads to both increased susceptibility to infection and protection against several types of autoimmune disease, whereas overnutrition is associated with low-grade, chronic inflammation that increases the risk of metabolic and cardiovascular disease, promotes autoreactivity, and disrupts protective immunity. Here, we review the effects of nutritional status on immunity and highlight the effects of nutrition on circulating cytokines and immune cell populations in both human studies and mouse models. As T cells are critical members of the immune system, which direct overall immune response, we will focus this review on the influence of systemic nutritional status on T cell metabolism and function. Several cytokines and hormones have been identified which mediate the effects of nutrition on T cell metabolism and function through the expression and action of key regulatory signaling proteins. Understanding how T cells are sensitive to both inadequate and overabundant nutrients may enhance our ability to target immune cell metabolism and alter immunity in both malnutrition and obesity.
Collapse
Affiliation(s)
- Yazan Alwarawrah
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Kaitlin Kiernan
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Nancie J MacIver
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States.,Department of Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
25
|
Wueest S, Laesser CI, Böni-Schnetzler M, Item F, Lucchini FC, Borsigova M, Müller W, Donath MY, Konrad D. IL-6-Type Cytokine Signaling in Adipocytes Induces Intestinal GLP-1 Secretion. Diabetes 2018; 67:36-45. [PMID: 29066599 DOI: 10.2337/db17-0637] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/15/2017] [Indexed: 11/13/2022]
Abstract
We recently showed that interleukin (IL)-6-type cytokine signaling in adipocytes induces free fatty acid release from visceral adipocytes, thereby promoting obesity-induced hepatic insulin resistance and steatosis. In addition, IL-6-type cytokines may increase the release of leptin from adipocytes and by those means induce glucagon-like peptide 1 (GLP-1) secretion. We thus hypothesized that IL-6-type cytokine signaling in adipocytes may regulate insulin secretion. To this end, mice with adipocyte-specific knockout of gp130, the signal transducer protein of IL-6, were fed a high-fat diet for 12 weeks. Compared with control littermates, knockout mice showed impaired glucose tolerance and circulating leptin, GLP-1, and insulin levels were reduced. In line, leptin release from isolated adipocytes was reduced, and intestinal proprotein convertase subtilisin/kexin type 1 (Pcsk1) expression, the gene encoding PC1/3, which controls GLP-1 production, was decreased in knockout mice. Importantly, treatment with the GLP-1 receptor antagonist exendin 9-39 abolished the observed difference in glucose tolerance between control and knockout mice. Ex vivo, supernatant collected from isolated adipocytes of gp130 knockout mice blunted Pcsk1 expression and GLP-1 release from GLUTag cells. In contrast, glucose- and GLP-1-stimulated insulin secretion was not affected in islets of knockout mice. In conclusion, adipocyte-specific IL-6 signaling induces intestinal GLP-1 release to enhance insulin secretion, thereby counteracting insulin resistance in obesity.
Collapse
Affiliation(s)
- Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Céline I Laesser
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Marianne Böni-Schnetzler
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department Biomedicine, University of Basel, Basel, Switzerland
| | - Flurin Item
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Fabrizio C Lucchini
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Marcela Borsigova
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Werner Müller
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department Biomedicine, University of Basel, Basel, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Interleukin-6 deficiency facilitates myocardial dysfunction during high fat diet-induced obesity by promoting lipotoxicity and inflammation. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3128-3141. [DOI: 10.1016/j.bbadis.2017.08.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/10/2017] [Accepted: 08/22/2017] [Indexed: 12/28/2022]
|
27
|
von Loeffelholz C, Lieske S, Neuschäfer-Rube F, Willmes DM, Raschzok N, Sauer IM, König J, Fromm M, Horn P, Chatzigeorgiou A, Pathe-Neuschäfer-Rube A, Jordan J, Pfeiffer AFH, Mingrone G, Bornstein SR, Stroehle P, Harms C, Wunderlich FT, Helfand SL, Bernier M, de Cabo R, Shulman GI, Chavakis T, Püschel GP, Birkenfeld AL. The human longevity gene homolog INDY and interleukin-6 interact in hepatic lipid metabolism. Hepatology 2017; 66:616-630. [PMID: 28133767 PMCID: PMC5519435 DOI: 10.1002/hep.29089] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 12/15/2016] [Accepted: 01/19/2017] [Indexed: 12/17/2022]
Abstract
UNLABELLED Reduced expression of the Indy ("I am Not Dead, Yet") gene in lower organisms promotes longevity in a manner akin to caloric restriction. Deletion of the mammalian homolog of Indy (mIndy, Slc13a5) encoding for a plasma membrane-associated citrate transporter expressed highly in the liver, protects mice from high-fat diet-induced and aging-induced obesity and hepatic fat accumulation through a mechanism resembling caloric restriction. We studied a possible role of mIndy in human hepatic fat metabolism. In obese, insulin-resistant patients with nonalcoholic fatty liver disease, hepatic mIndy expression was increased and mIndy expression was also independently associated with hepatic steatosis. In nonhuman primates, a 2-year high-fat, high-sucrose diet increased hepatic mIndy expression. Liver microarray analysis showed that high mIndy expression was associated with pathways involved in hepatic lipid metabolism and immunological processes. Interleukin-6 (IL-6) was identified as a regulator of mIndy by binding to its cognate receptor. Studies in human primary hepatocytes confirmed that IL-6 markedly induced mIndy transcription through the IL-6 receptor and activation of the transcription factor signal transducer and activator of transcription 3, and a putative start site of the human mIndy promoter was determined. Activation of the IL-6-signal transducer and activator of transcription 3 pathway stimulated mIndy expression, enhanced cytoplasmic citrate influx, and augmented hepatic lipogenesis in vivo. In contrast, deletion of mIndy completely prevented the stimulating effect of IL-6 on citrate uptake and reduced hepatic lipogenesis. These data show that mIndy is increased in liver of obese humans and nonhuman primates with NALFD. Moreover, our data identify mIndy as a target gene of IL-6 and determine novel functions of IL-6 through mINDY. CONCLUSION Targeting human mINDY may have therapeutic potential in obese patients with nonalcoholic fatty liver disease. German Clinical Trials Register: DRKS00005450. (Hepatology 2017;66:616-630).
Collapse
Affiliation(s)
- Christian von Loeffelholz
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Friedrich Schiller University, and Department of Anesthesiology and Intensive Care, Jena University Hospital, Jena, 01774, Germany
| | - Stefanie Lieske
- Section of Metabolic Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Paul Langerhans Institute Dresden (PLID), TU Dresden,01307 Germany
- Lehrstuhl für Biochemie der Ernährung, Universität Potsdam, Potsdam, 14558, Germany
| | | | - Diana M. Willmes
- Section of Metabolic Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Paul Langerhans Institute Dresden (PLID), TU Dresden,01307 Germany
| | - Nathanael Raschzok
- General, Visceral, and Transplantation Surgery, Charité – University School of Medicine, Berlin, 10117, Germany
| | - Igor M. Sauer
- General, Visceral, and Transplantation Surgery, Charité – University School of Medicine, Berlin, 10117, Germany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität, Erlangen-Nürnberg, 91054, Germany
| | - Martin Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität, Erlangen-Nürnberg, 91054, Germany
| | - Paul Horn
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Friedrich Schiller University, and Department of Anesthesiology and Intensive Care, Jena University Hospital, Jena, 01774, Germany
| | - Antonis Chatzigeorgiou
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Dresden, TUD, Germany
| | | | - Jens Jordan
- Institute for Clinical Pharmacology, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité – University School of Medicine, Berlin, 10117, Germany
- German Centre for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Geltrude Mingrone
- Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 8WA, UK
- Catholic University of Rome, Department of Internal Medicine, Rome, Italy
| | - Stefan R. Bornstein
- Section of Metabolic Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Paul Langerhans Institute Dresden (PLID), TU Dresden,01307 Germany
- Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 8WA, UK
- German Centre for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Peter Stroehle
- Max Planck Institute for Metabolism Research, Excellence cluster on cellular stress responses in aging associated diseases (CECAD), Cologne, 5093, Germany
| | - Christoph Harms
- Charité-Universitätsmedizin Berlin, Center for Stroke Research, Department of Experimental Neurology, Charitéplatz 1, 10117 Berlin, Germany
| | - F. Thomas Wunderlich
- Max Planck Institute for Metabolism Research, Excellence cluster on cellular stress responses in aging associated diseases (CECAD), Cologne, 5093, Germany
| | - Stephen. L. Helfand
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Gerald I. Shulman
- Department of Internal Medicine, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Dresden, TUD, Germany
| | - Gerhard. P. Püschel
- Lehrstuhl für Biochemie der Ernährung, Universität Potsdam, Potsdam, 14558, Germany
| | - Andreas. L. Birkenfeld
- Section of Metabolic Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Paul Langerhans Institute Dresden (PLID), TU Dresden,01307 Germany
- Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 8WA, UK
- German Centre for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Competence Center for Metabolic Vascular Medicine, GWT-TU Dresden, Germany
| |
Collapse
|
28
|
Lopez-Pascual A, Lasa A, Portillo MP, Arós F, Mansego ML, González-Muniesa P, Martinez JA. Low Oxygen Consumption is Related to a Hypomethylation and an Increased Secretion of IL-6 in Obese Subjects with Sleep Apnea-Hypopnea Syndrome. ANNALS OF NUTRITION AND METABOLISM 2017; 71:16-25. [DOI: 10.1159/000478276] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/07/2017] [Indexed: 12/17/2022]
Abstract
Background: Deoxyribonucleic acid (DNA) methylation is an epigenetic modification involved in gene expression regulation, usually via gene silencing, which contributes to the risks of many multifactorial diseases. The aim of the present study was to analyze the influence of resting oxygen consumption on global and gene DNA methylation as well as protein secretion of inflammatory markers in blood cells from obese subjects with sleep apnea-hypopnea syndrome (SAHS). Methods: A total of 44 obese participants with SAHS were categorized in 2 groups according to their resting oxygen consumption. DNA methylation levels were evaluated using a methylation-sensitive high resolution melting approach. Results: The analyzed interleukin 6 (IL6) gene cytosine phosphate guanine (CpG) islands showed a hypomethylation, while serum IL-6 was higher in the low compared to the high oxygen consumption group (p < 0.05). Moreover, an age-related loss of DNA methylation of tumor necrosis factor (B = -0.82, 95% CI -1.33 to -0.30) and long interspersed nucleotide element 1 (B = -0.46; 95% CI -0.87 to -0.04) gene CpGs were found. Finally, studied CpG methylation levels of serpin peptidase inhibitor, clade E member 1 (r = 0.43; p = 0.01), and IL6 (r = 0.41; p = 0.02) were positively associated with fat-free mass. Conclusions: These findings suggest a potential role of oxygen in the regulation of inflammatory genes. Oxygen consumption measurement at rest could be proposed as a clinical biomarker of metabolic health.
Collapse
|
29
|
The Role of Lipid and Lipoprotein Metabolism in Non-Alcoholic Fatty Liver Disease. CHILDREN-BASEL 2017; 4:children4060046. [PMID: 28587303 PMCID: PMC5483621 DOI: 10.3390/children4060046] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 12/14/2022]
Abstract
Due to the epidemic of obesity across the world, nonalcoholic fatty liver disease (NAFLD) has become one of the most prevalent chronic liver disorders in children and adolescents. NAFLD comprises a spectrum of fat-associated liver conditions that can result in end-stage liver disease and the need for liver transplantation. Simple steatosis, or fatty liver, occurs early in NAFLD and may progress to nonalcoholic steatohepatitis, fibrosis and cirrhosis with increased risk of hepatocellular carcinoma. The mechanism of the liver injury in NAFLD is currently thought to be a “multiple-hit process” where the first “hit” is an increase in liver fat, followed by multiple additional factors that trigger the inflammatory activity. At the onset of disease, NAFLD is characterized by hepatic triglyceride accumulation and insulin resistance. Liver fat accumulation is associated with increased lipotoxicity from high levels of free fatty acids, free cholesterol and other lipid metabolites. As a consequence, mitochondrial dysfunction with oxidative stress and production of reactive oxygen species and endoplasmic reticulum stress-associated mechanisms, are activated. The present review focuses on the relationship between intra-cellular lipid accumulation and insulin resistance, as well as on lipid and lipoprotein metabolism in NAFLD.
Collapse
|
30
|
Jurado-Ruiz E, Varela LM, Luque A, Berná G, Cahuana G, Martinez-Force E, Gallego-Durán R, Soria B, de Roos B, Romero Gómez M, Martín F. An extra virgin olive oil rich diet intervention ameliorates the nonalcoholic steatohepatitis induced by a high-fat “Western-type” diet in mice. Mol Nutr Food Res 2016; 61. [DOI: 10.1002/mnfr.201600549] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/07/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Enrique Jurado-Ruiz
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER); Universidad Pablo Olavide; Seville Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Lourdes M. Varela
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER); Universidad Pablo Olavide; Seville Spain
| | - Amparo Luque
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER); Universidad Pablo Olavide; Seville Spain
| | - Genoveva Berná
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER); Universidad Pablo Olavide; Seville Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Gladys Cahuana
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER); Universidad Pablo Olavide; Seville Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | | | - Rocío Gallego-Durán
- Unidad de Enfermedades Digestivas Hospitales Virgen Macarena-Virgen del Rocío; Instituto de Biomedicina de Sevilla, Universidad de Sevilla; Seville Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd); Madrid Spain
| | - Bernat Soria
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER); Universidad Pablo Olavide; Seville Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| | - Baukje de Roos
- Rowett Institute of Nutrition and Health; University of Aberdeen; Aberdeen; UK
| | - Manuel Romero Gómez
- Unidad de Enfermedades Digestivas Hospitales Virgen Macarena-Virgen del Rocío; Instituto de Biomedicina de Sevilla, Universidad de Sevilla; Seville Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd); Madrid Spain
| | - Franz Martín
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER); Universidad Pablo Olavide; Seville Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM); Madrid Spain
| |
Collapse
|
31
|
Nelson JE, Handa P, Aouizerat B, Wilson L, Vemulakonda LA, Yeh MM, Kowdley KV. Increased parenchymal damage and steatohepatitis in Caucasian non-alcoholic fatty liver disease patients with common IL1B and IL6 polymorphisms. Aliment Pharmacol Ther 2016; 44:1253-1264. [PMID: 27730688 PMCID: PMC5118184 DOI: 10.1111/apt.13824] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 07/15/2016] [Accepted: 09/19/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a complex, multifactorial disease affected by diet, lifestyle and genetics. Proinflammatory cytokines like IL-1β and IL-6 have been shown to be elevated in non-alcoholic steatohepatitis (NASH). AIM To investigate the relationship between IL1B and IL6 gene polymorphisms and histological features of NAFLD in the NASH CRN cohort. METHODS A total of 604 adult (≥18 years) non-Hispanic Caucasians with biopsy-proven NAFLD were genotyped for the following SNPs: IL1B, rs16944, rs1143634; IL6, rs1800795, rs10499563. Logistic regression was used to examine the relationship between genotype and a definitive diagnosis and advanced histological features of NASH after controlling for the following variables selected a priori: age, sex, diabetes, obesity and HOMA-IR level. RESULTS The IL6 rs10499563 C allele was independently associated with the presence of definitive NASH, and increased ballooning and Mallory bodies. The IL1B rs1143634 TT genotype was associated with advanced fibrosis and increased Mallory bodies. The IL6 rs1800795 C allele was associated with not only increased risk for severe steatosis, >66% but also decreased risk for advanced fibrosis and lobular inflammation and Mallory body formation. CONCLUSIONS These results suggest that common variants in the IL6 and IL1B genes may increase susceptibility for NASH and confer a higher risk of hepatic parenchymal damage including increased ballooning, increased Mallory bodies, and bridging fibrosis or cirrhosis. In contrast, the IL6 rs1800795 C allele may confer a higher risk for steatosis, but less parenchymal damage. Our findings support the development of therapeutics aimed at IL-1β and IL-6 suppression.
Collapse
Affiliation(s)
- James E. Nelson
- Benaroya Research Institute at Virginia Mason Medical Center, Seattle WA
| | - Priya Handa
- Liver Care Network and Organ Care Research Program, Swedish Medical Center, Seattle WA
| | - Bradley Aouizerat
- Department of Physiological Nursing, University of California at San Francisco, San Francisco, CA
- Institute for Human Genetics, University of California at San Francisco, San Francisco, CA
| | - Laura Wilson
- Johns Hopkins Bloomberg School of Public Health, Dept of Epidemiology, Baltimore, MD
| | - L Akhila Vemulakonda
- Liver Care Network and Organ Care Research Program, Swedish Medical Center, Seattle WA
| | - Matthew M. Yeh
- Department of Pathology, University of Washington School of Medicine, Seattle WA
| | - Kris V. Kowdley
- Benaroya Research Institute at Virginia Mason Medical Center, Seattle WA
- Liver Care Network and Organ Care Research Program, Swedish Medical Center, Seattle WA
| |
Collapse
|
32
|
Paquissi FC. Immune Imbalances in Non-Alcoholic Fatty Liver Disease: From General Biomarkers and Neutrophils to Interleukin-17 Axis Activation and New Therapeutic Targets. Front Immunol 2016; 7:490. [PMID: 27891128 PMCID: PMC5104753 DOI: 10.3389/fimmu.2016.00490] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/25/2016] [Indexed: 12/21/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an increasing problem worldwide and is associated with negative outcomes such as cirrhosis, hepatocellular carcinoma, insulin resistance, diabetes, and cardiovascular events. Current evidence shows that the immune response has an important participation driving the initiation, maintenance, and progression of the disease. So, various immune imbalances, from cellular to cytokines levels, have been studied, either for better compression of the disease pathophysiology or as biomarkers for severity assessment and outcome prediction. In this article, we performed a thorough review of studies that evaluated the role of inflammatory/immune imbalances in the NAFLD. At the cellular level, we gave special focus on the imbalance between neutrophils and lymphocytes counts (the neutrophil-to-lymphocyte ratio), and that which occurs between T helper 17 (Th17) and regulatory T cells as emerging biomarkers. By extension, we reviewed the reflection of these imbalances at the molecular level through pro-inflammatory cytokines including those involved in Th17 differentiation (IL-6, IL-21, IL-23, and transforming growth factor-beta), and those released by Th17 cells (IL-17A, IL-17F, IL-21, and IL-22). We gave particular attention to the role of IL-17, either produced by Th17 cells or neutrophils, in fibrogenesis and steatohepatitis. Finally, we reviewed the potential of these pathways as new therapeutic targets in NAFLD.
Collapse
|
33
|
Fabbrini E, Magkos F. Hepatic Steatosis as a Marker of Metabolic Dysfunction. Nutrients 2015; 7:4995-5019. [PMID: 26102213 PMCID: PMC4488828 DOI: 10.3390/nu7064995] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/05/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the liver manifestation of the complex metabolic derangements associated with obesity. NAFLD is characterized by excessive deposition of fat in the liver (steatosis) and develops when hepatic fatty acid availability from plasma and de novo synthesis exceeds hepatic fatty acid disposal by oxidation and triglyceride export. Hepatic steatosis is therefore the biochemical result of an imbalance between complex pathways of lipid metabolism, and is associated with an array of adverse changes in glucose, fatty acid, and lipoprotein metabolism across all tissues of the body. Intrahepatic triglyceride (IHTG) content is therefore a very good marker (and in some cases may be the cause) of the presence and the degree of multiple-organ metabolic dysfunction. These metabolic abnormalities are likely responsible for many cardiometabolic risk factors associated with NAFLD, such as insulin resistance, type 2 diabetes mellitus, and dyslipidemia. Understanding the factors involved in the pathogenesis and pathophysiology of NAFLD will lead to a better understanding of the mechanisms responsible for the metabolic complications of obesity, and hopefully to the discovery of novel effective treatments for their reversal.
Collapse
Affiliation(s)
- Elisa Fabbrini
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Faidon Magkos
- Center for Human Nutrition and Atkins Center of Excellence in Obesity Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
34
|
Qizhi Jiangtang Jiaonang Improves Insulin Signaling and Reduces Inflammatory Cytokine Secretion and Reactive Oxygen Species Formation in Insulin Resistant HepG2 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:518639. [PMID: 26074994 PMCID: PMC4436462 DOI: 10.1155/2015/518639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/16/2015] [Accepted: 04/21/2015] [Indexed: 11/18/2022]
Abstract
We analyzed the effects of a traditional Chinese medicine, Qizhi Jiangtang Jiaonang (QJJ), on insulin resistance (IR) in vitro. After an in vitro model of IR was established by treating human liver cancer cells (HepG2 cells) with palmitic acid, the cells were then treated with various concentrations of QJJ. Treatment with 400 µM palmitic acid for 24 h induced IR in HepG2 cells. The survival rate for HepG2 cells in the IR group was significantly lower than that of the untreated control group (P < 0.001); however, QJJ restored HepG2 cell survival (P < 0.001). As compared with HepG2 cells in the IR group, QJJ at all doses analyzed significantly increased glucose consumption (all P < 0.05). Moreover, treatment with all the QJJ doses significantly reduced the mean intracellular reactive oxygen species levels as compared with the IR group (all P < 0.05). Furthermore, high-dose QJJ reduced both TNF-α and IL-6 levels as compared to the IR group (all P < 0.05). QJJ ameliorated the altered PI3K, GLUT4, and RAGE expression observed with IR. In conclusion, QJJ can improve IR in HepG2 cells, which may be mediated through the IRS-1/PI3K/GLUT4 signaling pathway as well as regulation of NF-κB-mediated inflammation and oxidative stress.
Collapse
|