1
|
Satari S, Mota INR, Silva ACL, Brito HO, Oliveira PA, Gil da Costa RM, Medeiros R. Hallmarks of Cancer Cachexia: Sexual Dimorphism in Related Pathways. Int J Mol Sci 2025; 26:3952. [PMID: 40362192 PMCID: PMC12071346 DOI: 10.3390/ijms26093952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Cancer-associated cachexia (CAC), also known as wasting syndrome, is a systemic condition that affects multiple tissues and organs via a variety of metabolic pathways. Systemic inflammation, progressive weight loss, depletion of adipose tissue, and skeletal muscle impairment are some of the hallmark features of cachexia. Despite various studies on the clinical features of CAC, the complexity of the syndrome continues to pose significant challenges in clinical practice, leading to late diagnoses and the absence of a standardised treatment. Men and women respond differently to CAC, which may be prompted by the pre-existing physiologic sex differences. This review presents the sexual dimorphism associated with the hallmark pathways involved in CAC. A comprehensive understanding of sexual dimorphism in these pathways could drive research on cachexia to prioritise the inclusion of more females in related studies in order to achieve personalised sex-based therapeutic approaches and, consequently, enhance treatment efficacy and better patient outcomes.
Collapse
Affiliation(s)
- Setareh Satari
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- The Institute of Public Health, University of Porto (ISPUP), Rua das Taipas 135, 4050-600 Porto, Portugal
| | - Inês N. R. Mota
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal
| | - Ana Carolina Leão Silva
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
| | - Haissa Oliveira Brito
- Research Center For Experimental and Clinical Physiology and Pharmacology (NEC)/Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB)/Bioanalysis Lab (LaBIO), Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil;
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
| | - Rui Miguel Gil da Costa
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Research Center For Experimental and Clinical Physiology and Pharmacology (NEC)/Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB)/Bioanalysis Lab (LaBIO), Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil;
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Pathology and Laboratory Medicine Dep./Clinical Pathology, Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; (S.S.); (I.N.R.M.); (A.C.L.S.); (R.M.G.d.C.)
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
- Biomedical Research Center, Faculty of Health Sciences of the Fernando Pessoa University, 4249-004 Porto, Portugal
- ECO-European Cancer Organization, 1040 Brussels, Belgium
- Research Department of the Portuguese League Against Cancer—Regional Nucleus of the North (Liga Portuguesa Contra o Cancro—Núcleo Regional do Norte), 4200-172 Porto, Portugal
| |
Collapse
|
2
|
Janani KV, Saberian P, Patel HB, Keetha NR, Etemadzadeh A, Patel A, Hashemi SM, Amini-Salehi E, Gurram A. Prevalence of metabolic syndrome in patients with inflammatory bowel disease: a meta-analysis on a global scale. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:112. [PMID: 40205601 PMCID: PMC11983980 DOI: 10.1186/s41043-025-00860-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Metabolic syndrome (MetS) is a cluster of metabolic abnormalities that increase the risk of cardiovascular diseases (CVD). Patients with inflammatory bowel disease (IBD) may be at higher risk of developing MetS due to chronic inflammation, altered adipokine profiles, and the effects of corticosteroid treatment. However, the prevalence of MetS in IBD patients remains inconsistent across studies. This meta-analysis aims to estimate the prevalence of MetS in IBD patients and compare its occurrence between Crohn's disease (CD) and ulcerative colitis (UC). METHODS A systematic search was conducted across PubMed, Scopus, Embase, and Web of Science from their inception up to January 19, 2025. Eligible observational studies reporting MetS prevalence in IBD patients were included. Meta-analysis was performed using a random-effects model, with heterogeneity assessed via the I² statistic. Comprehensive Meta-Analysis (CMA) software, version 4.0 was used for analysis. RESULTS The pooled prevalence of MetS in IBD patients was 21.8% (95% CI: 14.3-31.6%). The prevalence was higher in UC patients (32.7%, 95% CI: 16.0-55.5%) compared to CD patients (14.1%, 95% CI: 8.6-22.3%). Patients with UC had significantly higher odds of MetS than those with CD (OR = 1.38, 95% CI: 1.03-1.85, P = 0.02). Additionally, IBD patients with MetS were significantly older than those without (MD: 9.89, 95% CI: 5.12-14.67, P < 0.01). CONCLUSION In summary, this meta-analysis reveals a notable prevalence of MetS among patients with IBD, particularly in those with UC, where the prevalence is higher than in CD. The analysis also shows that IBD patients with MetS tend to be older, suggesting age as a contributing factor. These findings underscore the need for routine metabolic screening in IBD care, especially in UC and elderly patients.
Collapse
Affiliation(s)
- Khushbu Viresh Janani
- Soundview Medical Associates, Department of Internal Medicine, Hartford Healthcare, 50 Danbury Road, Wilton, CT, 06612, USA
| | - Parsa Saberian
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hardik B Patel
- Department of Internal Medicine, Yale New Haven Health Bridgeport Hospital, 267 Grant Street, Bridgeport, CT, 06610, USA
| | - Narsimha Rao Keetha
- Ohio Kidney and Hypertension Center, 7255, Old Oak Blvd, Ste C111 Middleburg Hts, Fairview Park, OH, 44130, USA
| | - Ardalan Etemadzadeh
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Anya Patel
- , Nashua High School South 36 Riverside St, Nashua, NH, 03062, USA
| | - Seyyed Mohammad Hashemi
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Anoop Gurram
- Department of Hospital Medicine, Cleveland Clinic, 33300 Cleveland Clinic Blvd, Avon, Ohio, ashua, NH, 44011, 03062, USA
| |
Collapse
|
3
|
Pauss SN, Bates EA, Martinez GJ, Bates ZT, Kipp ZA, Gipson CD, Hinds TD. Steroid receptors and coregulators: Dissemination of sex differences and emerging technologies. J Biol Chem 2025; 301:108363. [PMID: 40023399 PMCID: PMC11986243 DOI: 10.1016/j.jbc.2025.108363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
Steroid receptors are ligand-induced transcription factors that have broad functions among all living animal species, ranging from control of sex differences, body weight, stress responses, and many others. Their binding to coregulator proteins is regulated by corepressors and coactivators that interchange upon stimulation with a ligand. Coregulator proteins are an imperative and understudied aspect of steroid receptor signaling. Here, we discuss steroid receptor basics from protein domain structures that allow them to interact with coregulators and other proteins, their essential functions as transcription factors, and other elemental protein-protein interactions. We deliberate about the mechanisms that coregulators control in steroid receptor signaling, sex hormone signaling differences, sex hormone treatment in the opposite sex, and how these affect the coregulator and sex steroid receptor complexes. The steroid receptor-coregulator signaling mechanisms are essential built-in components of the mammalian DNA that mediate physiological and everyday functions. Targeting their crosstalk might be useful when imbalances lead to disease. We introduce novel technologies, such as the PamGene PamStation, which make investigating the heterogeneity of the steroid receptor-coregulator complexes and targeting their binding more feasible. This review provides an extensive understanding of steroid receptor-coregulator signaling and how these interactions are intrinsic to many physiological functions that may offer therapeutic advantages.
Collapse
Affiliation(s)
- Sally N Pauss
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Evelyn A Bates
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Genesee J Martinez
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Zane T Bates
- Department of Bioengineering, University of Toledo College of Engineering, Toledo, Ohio, USA
| | - Zachary A Kipp
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Cassandra D Gipson
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Terry D Hinds
- Drug & Disease Discovery D3 Research Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, USA; Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA; Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA.
| |
Collapse
|
4
|
Feng Y, Mei W, Chen Q, Chen X, Ni Y, Lei M, Liu J. Probiotic Supplementation Alleviates Corticosterone-Induced Fatty Liver Disease by Regulating Hepatic Lipogenesis and Increasing Gut Microbiota Diversity in Broilers. Microorganisms 2025; 13:200. [PMID: 39858968 PMCID: PMC11767375 DOI: 10.3390/microorganisms13010200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/19/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Emerging evidence indicates a close relationship between gut microbiota and fatty liver disease. It has been suggested that gut microbiota modulation with probiotics ameliorates fatty liver disease in rodents and humans, yet it remains unclear whether the same results will also be obtained in poultry. The aim of this study was to investigate whether a mixture of probiotics supplemented after hatching can prevent CORT-induced fatty liver disease in broilers, and to determine how such effects, if any, are associated with hepatic de novo lipogenesis and gut microbiota composition. Ninety-six one-day-old green-legged chickens were divided into a control group (CON) and probiotic group (PB). At 28 days of age, fatty liver was induced in 16 broilers that were randomly selected from the CON or PB group. At the end of the experiment, broilers from four groups, (i) the control group (CON), (ii) corticosterone group (CORT), (iii) probiotic group (PB), and (iv) PB plus CORT group (CORT&PB), were slaughtered for sampling and analysis. The results showed that probiotic administration significantly prevented CORT-induced body weight loss (p < 0.05) but did not alleviate the weight loss of immune organs caused by CORT. Compared to CON, the broilers in the CORT group exhibited a significant increase in triglyceride (TG) levels in plasma and liver (p < 0.01), as well as severe hepatocytic steatosis and hepatocellular ballooning, which was accompanied by the upregulation of hepatic lipogenesis gene expression. However, probiotic supplementation markedly decreased the intrahepatic lipid accumulation and steatosis histological score, which was associated with the downregulation of sterol regulatory element-binding protein-1 (SREBP1) and acetyl-CoA carboxylase (ACC) mRNA (p < 0.05) and the expression of its protein (p = 0.06). The cecal microbiota composition was determined by 16S rRNA high-throughput sequencing. The results showed that CORT treatment induced distinct gut microbiota alterations with a decrease in microbial diversity and an increase in Proteobacteria abundance (p < 0.05). In contrast, probiotic supplementation increased the beta diversity, the community richness, and the diversity index (p > 0.05), as well as the abundance of Intestinimonas (p < 0.05). Our results indicate that CORT treatment induced severe fatty liver disease and altered the gut microbiota composition in broilers. However, post-hatching probiotic supplementation had a beneficial effect on alleviating fatty liver disease by regulating lipogenic gene expression and increasing gut microbiota diversity and the abundance of beneficial bacteria. We demonstrate for the first time that the supplementation of probiotics to chicks had a beneficial effect on preventing fatty liver disease through regulating lipogenic gene expression and improving the gut microbial balance. Thus, our results indicate that probiotics are a potential nutritional agent for preventing fatty liver disease in chickens.
Collapse
Affiliation(s)
- Yuyan Feng
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing 210094, China; (Y.F.); (X.C.)
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Wenqing Mei
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Qu Chen
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Xiaojing Chen
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing 210094, China; (Y.F.); (X.C.)
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Yingdong Ni
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Mingming Lei
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing 210094, China; (Y.F.); (X.C.)
| | - Jie Liu
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing 210094, China; (Y.F.); (X.C.)
| |
Collapse
|
5
|
Li W, Zheng L, Ma X, Xia J, Sheng J, Ge P, Yuan Y, Fan Y, Zhou Y. The sugar moiety in protopanaxadiol ginsenoside affects its ability to target glucocorticoid receptor to regulate lipid metabolism. Bioorg Chem 2024; 153:107885. [PMID: 39442459 DOI: 10.1016/j.bioorg.2024.107885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Ginsenosides are natural products with hydrophobic rings adorned with sugar molecules. The elucidation of the impact of ginsenosides structure on their activity is crucial for facilitating precision-oriented modifications, thereby enhancing their suitability for drug development. Here, utilizing an ob/ob mouse model, we demonstrated that as the number of sugar moiety on the protopanaxadiol-type ginsenosides decreased, the hypolipidemic potency increased, while the aglycon exhibited negligible activity. Mechanistically, we demonstrated the dependency of ginsenosides on the glucocorticoid receptor (GR) for the regulation of lipid metabolism. Interestingly, ginsenoside CK was found to promote the transcription of lipid metabolism-related genes via GR contrast to the effects of glucocorticoids, suggesting a unique mode of action. Furthermore, we observed that a reduction in the number of sugar molecules strengthened the binding affinity of ginsenosides to GR, as determined by microscale thermophoresis. These findings highlight the critical role of the sugar moiety in modulating the lipid-regulating capacity of ginsenosides, providing valuable insights for the development of these compounds as potential therapeutic agents.
Collapse
Affiliation(s)
- Weili Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Lujuan Zheng
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Xiao Ma
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Jing Xia
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Jiaxing Sheng
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Pengyu Ge
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Ye Yuan
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yuying Fan
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| | - Yifa Zhou
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| |
Collapse
|
6
|
Robinson CH, Aman N, Banh T, Brooke J, Chanchlani R, Dhillon V, Langlois V, Levin L, Licht C, McKay A, Noone D, Parikh A, Pearl R, Radhakrishnan S, Rowley V, Teoh CW, Vasilevska-Ristovska J, Parekh RS. Impact of childhood nephrotic syndrome on obesity and growth: a prospective cohort study. Pediatr Nephrol 2024; 39:2667-2677. [PMID: 38637343 DOI: 10.1007/s00467-024-06370-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/22/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Children with nephrotic syndrome are at risk of obesity and growth impairment from repeated steroid treatment. However, incidence and risk factors for obesity and short stature remain uncertain, which is a barrier to preventative care. Our aim was to determine risk, timing, and predictors of obesity and short stature among children with nephrotic syndrome. METHODS We evaluated obesity and longitudinal growth among children (1-18 years) enrolled in Insight into Nephrotic Syndrome: Investigating Genes, Health, and Therapeutics. We included children with nephrotic syndrome diagnosed between 1996-2019 from the Greater Toronto Area, Canada, excluding congenital or secondary nephrotic syndrome. Primary outcomes were obesity (body mass index Z-score ≥ + 2) and short stature (height Z-score ≤ -2). We evaluated prevalence of obesity and short stature at enrolment (< 1-year from diagnosis) and incidence during follow-up. Cox proportional hazards models determined the association between nephrotic syndrome classification and new-onset obesity and short stature. RESULTS We included 531 children with nephrotic syndrome (30% frequently relapsing by 1-year). At enrolment, obesity prevalence was 23.5%, 51.8% were overweight, and 4.9% had short stature. Cumulative incidence of new-onset obesity and short stature over median 4.1-year follow-up was 17.7% and 3.3% respectively. Children with frequently relapsing or steroid dependent nephrotic syndrome within 1-year of diagnosis were at increased risk of new-onset short stature (unadjusted hazard ratio 3.99, 95%CI 1.26-12.62) but not obesity (adjusted hazard ratio 1.56, 95%CI 0.95-2.56). Children with ≥ 7 and ≥ 15 total relapses were more likely to develop obesity and short stature, respectively. CONCLUSIONS Obesity is common among children with nephrotic syndrome early after diagnosis. Although short stature was uncommon overall, children with frequently relapsing or steroid dependent disease are at increased risk of developing short stature. Effective relapse prevention may reduce steroid toxicity and the risk of developing obesity or short stature.
Collapse
Affiliation(s)
- Cal H Robinson
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nowrin Aman
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Tonny Banh
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Josefina Brooke
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rahul Chanchlani
- Division of Nephrology, Department of Pediatrics, McMaster Children's Hospital, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Vaneet Dhillon
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Valerie Langlois
- Division of Nephrology, Department of Paediatrics, Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Leo Levin
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Christoph Licht
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada
- Program in Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ashlene McKay
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Damien Noone
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Alisha Parikh
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rachel Pearl
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada
- Division of Nephrology, William Osler Health Systems, 20 Lynch Street, Brampton, Ontario, L6W 2Z8, Canada
| | - Seetha Radhakrishnan
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada
| | - Veronique Rowley
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chia Wei Teoh
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada
| | | | - Rulan S Parekh
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Paediatrics, The University of Toronto, Toronto, Ontario, Canada.
- Child Health Evaluative Sciences, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Medicine, Women's College Hospital and University of Toronto, 76 Grenville St, Toronto, Ontario, M5S 1B2, Canada.
| |
Collapse
|
7
|
Martinez GJ, Kipp ZA, Lee WH, Bates EA, Morris AJ, Marino JS, Hinds TD. Glucocorticoid resistance remodels liver lipids and prompts lipogenesis, eicosanoid, and inflammatory pathways. Prostaglandins Other Lipid Mediat 2024; 173:106840. [PMID: 38830399 PMCID: PMC11199073 DOI: 10.1016/j.prostaglandins.2024.106840] [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: 02/29/2024] [Revised: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
We have previously demonstrated that the glucocorticoid receptor β (GRβ) isoform induces hepatic steatosis in mice fed a normal chow diet. The GRβ isoform inhibits the glucocorticoid-binding isoform GRα, reducing responsiveness and inducing glucocorticoid resistance. We hypothesized that GRβ regulates lipids that cause metabolic dysfunction. To determine the effect of GRβ on hepatic lipid classes and molecular species, we overexpressed GRβ (GRβ-Ad) and vector (Vec-Ad) using adenovirus delivery, as we previously described. We fed the mice a normal chow diet for 5 days and harvested the livers. We utilized liquid chromatography-mass spectrometry (LC-MS) analyses of the livers to determine the lipid species driven by GRβ. The most significant changes in the lipidome were monoacylglycerides and cholesterol esters. There was also increased gene expression in the GRβ-Ad mice for lipogenesis, eicosanoid synthesis, and inflammatory pathways. These indicate that GRβ-induced glucocorticoid resistance may drive hepatic fat accumulation, providing new therapeutic advantages.
Collapse
Affiliation(s)
- Genesee J Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Drug & Disease Discovery D3 Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Drug & Disease Discovery D3 Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Wang-Hsin Lee
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Drug & Disease Discovery D3 Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Evelyn A Bates
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Drug & Disease Discovery D3 Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Andrew J Morris
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, and Central Arkansas Veterans Affairs Healthcare System, Little Rock, AR 72205, USA
| | - Joseph S Marino
- Department of Applied Physiology, Health, and Clinical Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Drug & Disease Discovery D3 Research Center, University of Kentucky College of Medicine, Lexington, KY, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, USA; Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
8
|
Messeha SS, Agarwal M, Gendy SG, Mehboob SB, Soliman KFA. The Anti-Obesogenic Effects of Muscadine Grapes through Ciliary Neurotrophic Factor Receptor (Cntfr) and Histamine Receptor H1 (Hrh1) Genes in 3T3-L1 Differentiated Mouse Cells. Nutrients 2024; 16:1817. [PMID: 38931172 PMCID: PMC11206641 DOI: 10.3390/nu16121817] [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: 05/11/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Obesity and type 2 diabetes are prevalent metabolic diseases that have significant links to several chronic diseases, including cancer, diabetes, hypertension, and cardiovascular disease. Muscadine grape extracts have shown the potential to reduce adiposity and improve insulin sensitivity and glucose control. Thus, this study was designed to determine the potential of muscadine grape berries extract (Pineapple and Southern Home) for its antiobesity properties in 3T3-L1 cells as a model for obesity research. The current study's data indicated the total phenolic content (TPC) and 2,2-diphenyl-1-picrylhydraziyl (DPPH) activity were higher in cultivar (CV) Southern Home, meanwhile, elevated the total flavonoid content (TFC) in Pineapple. Both extracts were safe across the tested range (0-5 mg/mL). A noticeable reduction in lipid accumulation was also found in extract-treated cells. In preadipocytes and adipocytes, the tested extracts showed significant alterations in various genes involved in glucose homeostasis and obesity. The most remarkable findings of the current study are the upregulation of two genes, Cntfr (+712.715-fold) and Hrh1 (+270.11-fold) in CV Pineapple extract-treated adipocytes 3T3-L1 and the high fold increase in Ramp3 induced by both Pineapple and Southern Home in pre-adipose cells. Furthermore, the tested extracts showed a potential to alter the mRNA of various genes, including Zfp91, B2m, Nr3c1, Insr, Atrn, Il6ra, Hsp90ab1, Sort1, and Npy1r. In conclusion, the data generated from the current study suggested that the two extracts under investigation are considered potential candidates for controlling insulin levels and managing obesity.
Collapse
Affiliation(s)
- Samia S. Messeha
- College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, New Pharmacy Building, 1415 ML King Blvd., Tallahassee, FL 32307, USA
| | - Meenakshi Agarwal
- Center for Viticulture & Small Fruit Research, Florida A&M University, Tallahassee, FL 32317, USA;
| | - Sherif G. Gendy
- School of Allied Health Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Sheikh B. Mehboob
- Center for Viticulture & Small Fruit Research, Florida A&M University, Tallahassee, FL 32317, USA;
| | - Karam F. A. Soliman
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, New Pharmacy Building, 1415 ML King Blvd., Tallahassee, FL 32307, USA
| |
Collapse
|
9
|
Rao F, Xue T. Circadian-independent light regulation of mammalian metabolism. Nat Metab 2024; 6:1000-1007. [PMID: 38831000 DOI: 10.1038/s42255-024-01051-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/16/2024] [Indexed: 06/05/2024]
Abstract
The daily light-dark cycle is a key zeitgeber (time cue) for entraining an organism's biological clock, whereby light sensing by retinal photoreceptors, particularly intrinsically photosensitive retinal ganglion cells, stimulates the suprachiasmatic nucleus of the hypothalamus, a central pacemaker that in turn orchestrates the rhythm of peripheral metabolic activities. Non-rhythmic effects of light on metabolism have also been long known, and their transduction mechanisms are only beginning to unfold. Here, we summarize emerging evidence that, in mammals, light exposure or deprivation profoundly affects glucose homeostasis, thermogenesis and other metabolic activities in a clock-independent manner. Such light regulation could involve melanopsin-based, intrinsically photosensitive retinal ganglion cell-initiated brain circuits via the suprachiasmatic nucleus of the hypothalamus and other nuclei, or direct stimulation of opsins expressed in the hypothalamus, adipose tissue, blood vessels and skin to regulate sympathetic tone, lipolysis, glucose uptake, mitochondrial activation, thermogenesis, food intake, blood pressure and melanogenesis. These photic signalling events may coordinate with circadian-based mechanisms to maintain metabolic homeostasis, with dysregulation of this system underlying metabolic diseases caused by aberrant light exposure, such as environmental night light and shift work.
Collapse
Affiliation(s)
- Feng Rao
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Tian Xue
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
10
|
Perez-Leighton C, Kerr B, Scherer PE, Baudrand R, Cortés V. The interplay between leptin, glucocorticoids, and GLP1 regulates food intake and feeding behaviour. Biol Rev Camb Philos Soc 2024; 99:653-674. [PMID: 38072002 DOI: 10.1111/brv.13039] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 11/21/2023] [Accepted: 12/01/2023] [Indexed: 05/09/2024]
Abstract
Nutritional, endocrine, and neurological signals converge in multiple brain centres to control feeding behaviour and food intake as part of the allostatic regulation of energy balance. Among the several neuroendocrine systems involved, the leptin, glucocorticoid, and glucagon-like peptide 1 (GLP1) systems have been extensively researched. Leptin is at the top hierarchical level since its complete absence is sufficient to trigger severe hyperphagia. Glucocorticoids are key regulators of the energy balance adaptation to stress and their sustained excess leads to excessive adiposity and metabolic perturbations. GLP1 participates in metabolic adaptation to food intake, regulating insulin secretion and satiety by parallel central and peripheral signalling systems. Herein, we review the brain and peripheral targets of these three hormone systems that integrate to regulate food intake, feeding behaviour, and metabolic homeostasis. We examine the functional relationships between leptin, glucocorticoids, and GLP1 at the central and peripheral levels, including the cross-regulation of their circulating levels and their cooperative or antagonistic actions at different brain centres. The pathophysiological roles of these neuroendocrine systems in dysregulated intake are explored in the two extremes of body adiposity - obesity and lipodystrophy - and eating behaviour disorders.
Collapse
Affiliation(s)
- Claudio Perez-Leighton
- Departmento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| | - Bredford Kerr
- Centro de Biología Celular y Biomedicina-CEBICEM, Facultad de Medicina y Ciencia, Universidad San Sebastián, Carmen Sylva 2444, Providencia, Santiago, Chile
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - René Baudrand
- Departmento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
- Centro Translacional de Endocrinología (CETREN), Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| | - Víctor Cortés
- Departmento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| |
Collapse
|
11
|
Sadek SM, Khatri SN, Kipp Z, Dunn KE, Beckmann JS, Stoops WW, Hinds TD, Gipson CD. Impacts of xylazine on fentanyl demand, body weight, and acute withdrawal in rats: A comparison to lofexidine. Neuropharmacology 2024; 245:109816. [PMID: 38128606 PMCID: PMC10843705 DOI: 10.1016/j.neuropharm.2023.109816] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
The opioid use landscape has recently shifted to include xylazine, a veterinary anesthetic, as an adulterant in the fentanyl supply. The health impacts of xylazine as an emerging fentanyl adulterant has raised alarm regarding xylazine as a public health threat, warranting research on the impacts of xylazine on fentanyl's behavioral effects. No prior studies have evaluated the effects of xylazine on fentanyl consumption at various unit doses, fentanyl demand, or withdrawal as compared to the Food and Drug Administration-approved opioid withdrawal medication, lofexidine (Lucemyra®). This is important because lofexidine and xylazine are both adrenergic α2a (A2aR) agonists, however, lofexidine is not a noted fentanyl adulterant. Here we evaluated xylazine and lofexidine combined with self-administered fentanyl doses in male and female rats and evaluated fentanyl demand, body weight, and acute withdrawal. Consumption of fentanyl alone increased at various unit doses compared to saline. Xylazine but not lofexidine shifted fentanyl consumption downward at a number of unit doses, however, both lofexidine and xylazine suppressed fentanyl demand intensity as compared to a fentanyl alone control group. Further, both fentanyl + lofexidine and fentanyl + xylazine reduced behavioral signs of fentanyl withdrawal immediately following SA, but signs increased by 12 h only in the xylazine co-exposed group. Weight loss occurred throughout fentanyl SA and withdrawal regardless of group, although the xylazine group lost significantly more weight during the first 24 h of withdrawal than the other two groups. Severity of weight loss during the first 24 h of withdrawal was also correlated with severity of somatic signs of fentanyl withdrawal. Together, these results suggest that body weight loss may be an important indicator of withdrawal severity during acute withdrawal from the xylazine/fentanyl combination, warranting further translational evaluation.
Collapse
Affiliation(s)
- Safiyah M Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Shailesh N Khatri
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Zachary Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Kelly E Dunn
- Psychiatry and Behavioral Sciences Department, Johns Hopkins University, Baltimore, MD, USA
| | - Joshua S Beckmann
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - William W Stoops
- Department of Behavioral Sciences, University of Kentucky, Lexington, KY, USA
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
12
|
Christakoudi S, Asimakopoulos AG, Riboli E, Tsilidis KK. Links between the genetic determinants of morning plasma cortisol and body shape: a two-sample Mendelian randomisation study. Sci Rep 2024; 14:3230. [PMID: 38332183 PMCID: PMC10853188 DOI: 10.1038/s41598-024-53727-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/04/2024] [Indexed: 02/10/2024] Open
Abstract
High cortisol production in Cushing's syndrome leads to fat centralisation. The influence of modest cortisol variations on body shape, however, is less clear. We examined potentially causal associations between morning plasma cortisol and body shape and obesity with inverse-variance weighted random-effects models in a two-sample Mendelian randomisation analysis. We used publicly available summary statistics from the CORtisol NETwork (CORNET) consortium, UK Biobank, and the Genetic Investigation of Anthropometric Traits (GIANT) consortium. Only in women, morning plasma cortisol (proxied by ten genetic polymorphisms) was associated positively with waist size reflected in waist-to-hip index (WHI, 0.035 standard deviation (SD) units change per one SD cortisol increase; 95% confidence interval (0.002-0.067); p = 0.036) and "a body shape index" (ABSI; 0.039 (0.006-0.071); p = 0.021). There was no evidence for associations with hip index (HI) or body mass index (BMI). Among individual polymorphisms, rs7450600 stood out (chromosome 6; Long Intergenic Non-Protein-Coding RNA 473 gene, LINC00473). Morning plasma cortisol proxied by rs7450600 was associated positively with WHI and inversely with HI and BMI in women and men. Our findings support a causal association of higher morning plasma cortisol with larger waist size in women and highlight LINC00473 as a genetic link between morning plasma cortisol and body shape.
Collapse
Affiliation(s)
- Sofia Christakoudi
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, White City Campus, 90 Wood Lane, London, W12 0BZ, UK.
| | | | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, White City Campus, 90 Wood Lane, London, W12 0BZ, UK
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, White City Campus, 90 Wood Lane, London, W12 0BZ, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| |
Collapse
|
13
|
Martinez GJ, Appleton M, Kipp ZA, Loria AS, Min B, Hinds TD. Glucocorticoids, their uses, sexual dimorphisms, and diseases: new concepts, mechanisms, and discoveries. Physiol Rev 2024; 104:473-532. [PMID: 37732829 PMCID: PMC11281820 DOI: 10.1152/physrev.00021.2023] [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: 05/22/2023] [Revised: 08/07/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023] Open
Abstract
The normal stress response in humans is governed by the hypothalamic-pituitary-adrenal (HPA) axis through heightened mechanisms during stress, raising blood levels of the glucocorticoid hormone cortisol. Glucocorticoids are quintessential compounds that balance the proper functioning of numerous systems in the mammalian body. They are also generated synthetically and are the preeminent therapy for inflammatory diseases. They act by binding to the nuclear receptor transcription factor glucocorticoid receptor (GR), which has two main isoforms (GRα and GRβ). Our classical understanding of glucocorticoid signaling is from the GRα isoform, which binds the hormone, whereas GRβ has no known ligands. With glucocorticoids being involved in many physiological and cellular processes, even small disruptions in their release via the HPA axis, or changes in GR isoform expression, can have dire ramifications on health. Long-term chronic glucocorticoid therapy can lead to a glucocorticoid-resistant state, and we deliberate how this impacts disease treatment. Chronic glucocorticoid treatment can lead to noticeable side effects such as weight gain, adiposity, diabetes, and others that we discuss in detail. There are sexually dimorphic responses to glucocorticoids, and women tend to have a more hyperresponsive HPA axis than men. This review summarizes our understanding of glucocorticoids and critically analyzes the GR isoforms and their beneficial and deleterious mechanisms and the sexual differences that cause a dichotomy in responses. We also discuss the future of glucocorticoid therapy and propose a new concept of dual GR isoform agonist and postulate why activating both isoforms may prevent glucocorticoid resistance.
Collapse
Affiliation(s)
- Genesee J Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Malik Appleton
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
14
|
Penner-Goeke S, Bothe M, Rek N, Kreitmaier P, Pöhlchen D, Kühnel A, Glaser LV, Kaya E, Krontira AC, Röh S, Czamara D, Ködel M, Monteserin-Garcia J, Diener L, Wölfel B, Sauer S, Rummel C, Riesenberg S, Arloth-Knauer J, Ziller M, Labeur M, Meijsing S, Binder EB. High-throughput screening of glucocorticoid-induced enhancer activity reveals mechanisms of stress-related psychiatric disorders. Proc Natl Acad Sci U S A 2023; 120:e2305773120. [PMID: 38011552 DOI: 10.1073/pnas.2305773120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/01/2023] [Indexed: 11/29/2023] Open
Abstract
Exposure to stressful life events increases the risk for psychiatric disorders. Mechanistic insight into the genetic factors moderating the impact of stress can increase our understanding of disease processes. Here, we test 3,662 single nucleotide polymorphisms (SNPs) from preselected expression quantitative trait loci in massively parallel reporter assays to identify genetic variants that modulate the activity of regulatory elements sensitive to glucocorticoids, important mediators of the stress response. Of the tested SNP sequences, 547 were located in glucocorticoid-responsive regulatory elements of which 233 showed allele-dependent activity. Transcripts regulated by these functional variants were enriched for those differentially expressed in psychiatric disorders in the postmortem brain. Phenome-wide Mendelian randomization analysis in 4,439 phenotypes revealed potentially causal associations specifically in neurobehavioral traits, including major depression and other psychiatric disorders. Finally, a functional gene score derived from these variants was significantly associated with differences in the physiological stress response, suggesting that these variants may alter disease risk by moderating the individual set point of the stress response.
Collapse
Affiliation(s)
- Signe Penner-Goeke
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- Graduate School of Systemic Neurosciences, Ludwig Maximilian University of Munich, Planegg 82152, Germany
| | - Melissa Bothe
- Department of Computational Molecular Biology, Max Planck Institute of Molecular Genetics, Berlin 14195, Germany
| | - Nils Rek
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Peter Kreitmaier
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg 85764, Germany
| | - Dorothee Pöhlchen
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Anne Kühnel
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Laura V Glaser
- Department of Computational Molecular Biology, Max Planck Institute of Molecular Genetics, Berlin 14195, Germany
| | - Ezgi Kaya
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- Graduate School of Systemic Neurosciences, Ludwig Maximilian University of Munich, Planegg 82152, Germany
| | - Anthi C Krontira
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Simone Röh
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Maik Ködel
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Jose Monteserin-Garcia
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Laura Diener
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Barbara Wölfel
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Susann Sauer
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Christine Rummel
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Stephan Riesenberg
- Department of Evolutionary Genetics, Max-Planck-Institute for Evolutionary Anthropology, Leipzig 04103, Germany
| | - Janine Arloth-Knauer
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Michael Ziller
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- Department of Psychiatry, University of Muenster, Muenster 48149, Germany
| | - Marta Labeur
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Sebastiaan Meijsing
- Department of Computational Molecular Biology, Max Planck Institute of Molecular Genetics, Berlin 14195, Germany
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| |
Collapse
|
15
|
Yu GR, Kim JE, Lim DW, Park WH. The combination of Ephedrae herba and coixol from Coicis semen attenuate adiposity via glucocorticoid receptor regulation. Sci Rep 2023; 13:20324. [PMID: 37990123 PMCID: PMC10663538 DOI: 10.1038/s41598-023-47553-3] [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: 07/19/2023] [Accepted: 11/15/2023] [Indexed: 11/23/2023] Open
Abstract
The enhanced therapeutic effects and mechanisms of certain herbal combination in various herbal prescriptions are mostly unclear. A combination of two herbs, namely Ephedrae herba (EH) and Coicis semen (CS), has been commonly prescribed for obesity. In our previous work, the combination of EH and CS was studied using network pharmacological approach to predict its pharmacological targets and in vitro experiments to evaluate its efficacy on obesity. Although we demonstrated enhanced anti-adiposity effects of the combination on matured adipocytes, the molecular mechanisms and contributing compounds underlying the effects of EH-CS combination on adiposity or adipogenesis were not fully elucidated. The current study adopted integrated bioinformatics analysis to precisely validate potential targets of EH-CS by screening differentially expressed genes (DEGs) of morbid obesity patients from NCBI gene expression omnibus (GEO). Based on the functional cluster analysis of down-regulated DEGs, the anti-adipogenesis mechanism of EH-CS combination was speculated with KEGG enrichment analysis. Furthermore, we investigated the combinational effects of EH and coixol, or stigmasterol, the two compounds in CS which were expected to have main beneficial effects in metabolic diseases. Moreover, distinct effect of the combination on transcriptional activity of glucocorticoid receptor (GR) was investigated using electrophoretic mobility shift assay (EMSA). The EH-CS combination was predicted to modulate down-regulated genes which are involved in KEGG pathways crucial to metabolic disease in morbidly obese individuals. The combination of EH with CS compounds significantly increased the phosphorylation of acetyl-coA carboxylase (ACC), AMP-activated protein kinase (AMPK), and protein kinase B (AKT) in 3T3-L1 cells and decreased intracellular lipid accumulation. The two CS compounds significantly increased the anti-adipogenesis/lipogenesis effects of EH by inhibiting the gene expression levels. Finally, the combination of EH and coixol inhibited dexamethasone-induced GR translocation to the nucleus and transcriptional binding activity in adipocytes. The combination of EH and CS could be considered a therapeutic strategy for treating metabolic diseases, including obesity.
Collapse
Affiliation(s)
- Ga-Ram Yu
- Department of Diagnostic, College of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea
| | - Jai-Eun Kim
- Department of Pathology, College of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea
| | - Dong-Woo Lim
- Department of Diagnostic, College of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea.
- Institute of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea.
| | - Won-Hwan Park
- Department of Diagnostic, College of Korean Medicine, Dongguk University, Goyang, 10326, Republic of Korea.
| |
Collapse
|
16
|
Lee WH, Najjar SM, Kahn CR, Hinds TD. Hepatic insulin receptor: new views on the mechanisms of liver disease. Metabolism 2023; 145:155607. [PMID: 37271372 PMCID: PMC10330768 DOI: 10.1016/j.metabol.2023.155607] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/06/2023]
Abstract
Over 65 % of people with obesity display the metabolic-associated fatty liver disease (MAFLD), which can manifest as steatohepatitis, fibrosis, cirrhosis, or liver cancer. The development and progression of MAFLD involve hepatic insulin resistance and reduced insulin clearance. This review discusses the relationships between altered insulin signaling, hepatic insulin resistance, and reduced insulin clearance in the development of MAFLD and how this provides the impetus for exploring the use of insulin sensitizers to curb this disease. The review also explores the role of the insulin receptor in hepatocytes and hepatic stellate cells and how it signals in metabolic and end-stage liver diseases. Finally, we discuss new research findings that indicate that advanced hepatic diseases may be an insulin-sensitive state in the liver and deliberate whether insulin sensitizers should be used to manage late-stage liver diseases.
Collapse
Affiliation(s)
- Wang-Hsin Lee
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Sonia M Najjar
- Department of Biomedical Sciences and the Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA; Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, KY, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
17
|
Matsumura Y. Inadequate therapeutic responses to glucocorticoid treatment in bronchial asthma. J Int Med Res 2023; 51:3000605231175746. [PMID: 37296513 DOI: 10.1177/03000605231175746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023] Open
Abstract
Bronchial asthma (BA) is a heterogeneous disease. Some patients benefit greatly from glucocorticoid (GC) treatment, whereas others are non-responders. This could be attributable to differences in pathobiology. Thus, predicting the responses to GC treatment in patients with BA is necessary to increase the success rates of GC therapy and avoid adverse effects. The sustained inflammation in BA decreases glucocorticoid receptor (GR, NR3C1) function. Meanwhile, GRβ overexpression might contribute to GC resistance. Important factors in decreased GR function include p38 mitogen-activated protein kinase-dependent GR phosphorylated at Ser226, reduced expression of histone deacetylase 2 following activation of the phosphatidylinositol 3-kinase-δ signaling pathway, and increased nuclear factor-kappa B activity. MicroRNAs, which are involved in GC sensitivity, are considered biomarkers of the response to inhaled GCs. Some studies revealed that inflammatory phenotypes and disease-related modifiable factors, including infections, the airway microbiome, mental stress, smoking, and obesity, regulate individual sensitivity to GCs. Therefore, future investigations are warranted to improve treatment outcomes.
Collapse
Affiliation(s)
- Yasuhiro Matsumura
- Department of Internal Medicine, Sasaki Foundation Kyoundo Hospital, Tokyo, Japan
| |
Collapse
|
18
|
Wang CJ, Noble PB, Elliot JG, James AL, Wang KCW. From Beneath the Skin to the Airway Wall: Understanding the Pathological Role of Adipose Tissue in Comorbid Asthma-Obesity. Compr Physiol 2023; 13:4321-4353. [PMID: 36715283 DOI: 10.1002/cphy.c220011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article provides a contemporary report on the role of adipose tissue in respiratory dysfunction. Adipose tissue is distributed throughout the body, accumulating beneath the skin (subcutaneous), around organs (visceral), and importantly in the context of respiratory disease, has recently been shown to accumulate within the airway wall: "airway-associated adipose tissue." Excessive adipose tissue deposition compromises respiratory function and increases the severity of diseases such as asthma. The mechanisms of respiratory impairment are inflammatory, structural, and mechanical in nature, vary depending on the anatomical site of deposition and adipose tissue subtype, and likely contribute to different phenotypes of comorbid asthma-obesity. An understanding of adipose tissue-driven pathophysiology provides an opportunity for diagnostic advancement and patient-specific treatment. As an exemplar, the potential impact of airway-associated adipose tissue is highlighted, and how this may change the management of a patient with asthma who is also obese. © 2023 American Physiological Society. Compr Physiol 13:4321-4353, 2023.
Collapse
Affiliation(s)
- Carolyn J Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
19
|
Chen W, Meng F, Zeng X, Cao X, Bu G, Du X, Yu G, Kong F, Li Y, Gan T, Han X. Mechanic Insight into the Distinct and Common Roles of Ovariectomy Versus Adrenalectomy on Adipose Tissue Remodeling in Female Mice. Int J Mol Sci 2023; 24:ijms24032308. [PMID: 36768630 PMCID: PMC9916485 DOI: 10.3390/ijms24032308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Dysfunctions of the ovaries and adrenal glands are both evidenced to cause aberrant adipose tissue (AT) remodeling and resultant metabolic disorders, but their distinct and common roles are poorly understood. In this study, through biochemical, histological and RNA-seq analyses, we comprehensively explored the mechanisms underpinning subcutaneous (SAT) and visceral adipose tissue (VAT) remodeling, in response to ovariectomy (OVX) versus adrenalectomy (ADX) in female mice. OVX promoted adipocyte differentiation and fat accumulation in both SAT and VAT, by potentiating the Pparg signaling, while ADX universally prevented the cell proliferation and extracellular matrix organization in both SAT and VAT, likely by inactivating the Nr3c1 signaling, thus causing lipoatrophy in females. ADX, but not OVX, exerted great effects on the intrinsic difference between SAT and VAT. Specifically, ADX reversed a large cluster of genes differentially expressed between SAT and VAT, by activating 12 key transcription factors, and thereby caused senescent cell accumulation, massive B cell infiltration and the development of selective inflammatory response in SAT. Commonly, both OVX and ADX enhance circadian rhythmicity in VAT, and impair cell proliferation, neurogenesis, tissue morphogenesis, as well as extracellular matrix organization in SAT, thus causing dysfunction of adipose tissues and concomitant metabolic disorders.
Collapse
|
20
|
Sultana N, Islam R. Modulation of the dynamics and cellularity of adipose tissues in different fat depots in broilers by dietary dexamethasone. J Adv Vet Anim Res 2022; 9:583-590. [PMID: 36714508 PMCID: PMC9868779 DOI: 10.5455/javar.2022.i627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/16/2022] [Accepted: 09/26/2022] [Indexed: 01/15/2023] Open
Abstract
Objective The objective of this investigation was to determine the effects of dexamethasone (DEX) on the weight and cellularity of abdominal and subcutaneous fat depots. Materials and Methods The study was conducted on four broiler chicks (20 chicks per group) fed commercial feed and water ad libitum. The DEX was supplied with feed at 0 mg/kg (non-DEX), 3 mg/kg (DEX-1), 5 mg/kg (DEX-2), and 7 mg/kg (DEX-3) from day 0 to day 28. The entire abdominal and subcutaneous fat depots were collected and weighed after sacrificing five birds from each group on days 14 and 28. Results The DEX groups had considerably lower (p < 0.05) fat depot weights with dose-related variation noted among the DEX groups. The histological findings revealed the presence of unilocular, round to oval-shaped adipocytes. The DEX-1 and DEX-2 had way lower (p < 0.05) numbers of adipocytes while the DEX-3 had considerably higher (p < 0.05) numbers of adipocytes than the non-DEX. DEX-1 and DEX-2 had larger (p < 0.05) adipocytes whereas DEX-3 had smaller adipocytes than the non-DEX. Adipocyte sizes and fat depot weights were found to have very strong negative relationships. Conclusion Dietary DEX affects the growth and distribution of abdominal and subcutaneous fat depots and adipocyte cellularity subjected to both dose and duration of DEX treatment.
Collapse
Affiliation(s)
- Nasrin Sultana
- Department of Anatomy and Histology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Rafiqul Islam
- Department of Anatomy and Histology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| |
Collapse
|
21
|
Srivastava RK, Ruiz de Azua I, Conrad A, Purrio M, Lutz B. Cannabinoid CB1 Receptor Deletion from Catecholaminergic Neurons Protects from Diet-Induced Obesity. Int J Mol Sci 2022; 23:ijms232012635. [PMID: 36293486 PMCID: PMC9604114 DOI: 10.3390/ijms232012635] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
High-calorie diets and chronic stress are major contributors to the development of obesity and metabolic disorders. These two risk factors regulate the activity of the sympathetic nervous system (SNS). The present study showed a key role of the cannabinoid type 1 receptor (CB1) in dopamine β-hydroxylase (dbh)-expressing cells in the regulation of SNS activity. In a diet-induced obesity model, CB1 deletion from these cells protected mice from diet-induced weight gain by increasing sympathetic drive, resulting in reduced adipogenesis in white adipose tissue and enhanced thermogenesis in brown adipose tissue. The deletion of CB1 from catecholaminergic neurons increased the plasma norepinephrine levels, norepinephrine turnover, and sympathetic activity in the visceral fat, which coincided with lowered neuropeptide Y (NPY) levels in the visceral fat of the mutant mice compared with the controls. Furthermore, the mutant mice showed decreased plasma corticosterone levels. Our study provided new insight into the mechanisms underlying the roles of the endocannabinoid system in regulating energy balance, where the CB1 deletion in dbh-positive cells protected from diet-induced weight gain via multiple mechanisms, such as increased SNS activity, reduced NPY activity, and decreased basal hypothalamic-pituitary-adrenal (HPA) axis activity.
Collapse
Affiliation(s)
- Raj Kamal Srivastava
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, 55128 Mainz, Germany
- Department of Zoology, Indira Gandhi National Tribal University, Amarkantak, Anuppur 484887, India
| | - Inigo Ruiz de Azua
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, 55128 Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Andrea Conrad
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, 55128 Mainz, Germany
| | - Martin Purrio
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, 55128 Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University of Mainz, 55128 Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
- Correspondence:
| |
Collapse
|
22
|
de Souza MLM, Borçoi AR, Dutra BAB, Dos Santos Vieira T, Mendes SO, Nascimento IAA, Quaioto BR, Olinda AS, Cunha ER, Freitas FV, Pinheiro JA, Dos Santos JG, Sorroche BP, Arantes LMRB, Sartório CL, da Silva AMA. Lifestyle and NR3C1 exon 1F gene methylation is associated with changes in glucose levels and insulin resistance. Life Sci 2022; 309:120940. [PMID: 36108769 DOI: 10.1016/j.lfs.2022.120940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/03/2022] [Accepted: 09/05/2022] [Indexed: 10/31/2022]
Abstract
AIMS the present study aimed to investigate how glucose and insulin levels may be associated with changes in NR3C1 gene methylation levels in adults. MAIN METHODS 375 volunteers users of the Brazilian Public Unified Health System (SUS) were recruited to assess socioeconomic status, lifestyle, anthropometric data, blood glucose and serum cortisol levels, insulin resistance, and NR3C1 gene methylation assessment. Factors associated with glucose levels and insulin resistance were investigated using multivariate analysis GLzM at 5 % significance (p < 0.05). KEY FINDINGS our results verified that glucose levels and insulin resistance were directly related to NR3C1 gene methylation and age, while not being overweight and obese and no tobacco consumption were indirectly related to glucose levels and insulin resistance. SIGNIFICANCE habits and lifestyle may influence NR3C1 gene regulation, revealing the complexity of environmental impacts on NR3C1 methylation. Furthermore, associated risk factors must be taken into account in epigenetic studies as they directly interfere with blood glucose levels and insulin resistance.
Collapse
Affiliation(s)
| | - Aline Ribeiro Borçoi
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | | | - Tamires Dos Santos Vieira
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | | | | | - Barbara Risse Quaioto
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Amanda Sgrancio Olinda
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Ester Ribeiro Cunha
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Flávia Vitorino Freitas
- Department of Pharmacy and Nutrition, Universidade Federal do Espírito Santo, Alegre, ES, Brazil
| | - Julia Assis Pinheiro
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | | | - Bruna Pereira Sorroche
- Molecular Oncology Research Center, Hospital do Câncer de Barretos, Barretos, São Paulo, Brazil
| | | | - Carmem Luíza Sartório
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | | |
Collapse
|
23
|
Ruan H, Tang Q, Zhao X, Zhang Y, Zhao X, Xiang Y, Geng W, Feng Y, Cai W. The levels of osteopontin in human milk of Chinese mothers and its associations with maternal body composition. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.04.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
24
|
Molecular mechanisms of metabolic associated fatty liver disease (MAFLD): functional analysis of lipid metabolism pathways. Clin Sci (Lond) 2022; 136:1347-1366. [PMID: 36148775 PMCID: PMC9508552 DOI: 10.1042/cs20220572] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 01/30/2023]
Abstract
The metabolic-associated fatty liver disease (MAFLD) is a condition of fat accumulation in the liver in combination with metabolic dysfunction in the form of overweight or obesity and insulin resistance. It is also associated with an increased cardiovascular disease risk, including hypertension and atherosclerosis. Hepatic lipid metabolism is regulated by a combination of the uptake and export of fatty acids, de novo lipogenesis, and fat utilization by β-oxidation. When the balance between these pathways is altered, hepatic lipid accumulation commences, and long-term activation of inflammatory and fibrotic pathways can progress to worsen the liver disease. This review discusses the details of the molecular mechanisms regulating hepatic lipids and the emerging therapies targeting these pathways as potential future treatments for MAFLD.
Collapse
|
25
|
Tavares MEA, Veras ASC, Thorpe HHA, Baptista DB, Teixeira GR. Physical exercise regulates apoptosis and prostatic inflammatory effects induced by high-fat diet in PPAR-alpha deleted mice. Prostaglandins Other Lipid Mediat 2022; 163:106669. [PMID: 35963510 DOI: 10.1016/j.prostaglandins.2022.106669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022]
Abstract
The high-fat diet (HFD) promotes obesity and develops inflammation, causing dysregulation of energy metabolism and prostatic neoplastic tissue changes. PPARɑ deletion leads to loss of homeostasis between the pro and anti-inflammatory response, and dysregulation of lipid metabolism, causing changes in different physiological processes and damage to the prostate. On the other hand, aerobic physical exercise has been suggested as a non-pharmacological tool to improve energy metabolism and cellular metabolism in the prostate, however, the underlying molecular mechanism remains unclear. the current study aimed to evaluate PPARα as a possible regulator of the protective effects of aerobic physical exercise in the prostate by examining prostatic alterations in wild-type and PPARα deletion mice fed a standard diet or an HFD. Wild-type and PPARα-null mice were fed a standard or HFD diet for 12 weeks, and submitted to aerobic physical exercise for 8 weeks. The HFD promoted the increase of inflammatory markers IL-6, TNF-α, NF-kB, and an increase of inflammatory foci in animals in both genotypes. Although the PPARα deletion animals submitted to the aerobic physical exercise were not able to regulate response pro-inflammatory, but promoted an increase in IL-10 in the prostate. In animals WT, the aerobic physical exercise, reduced all inflammatory markers, improve the inflammatory response, and showed a higher expression of BAX and IL-10 proteins was protective against prostatic tissue lesions. Suggested that PPARα deletion associated with HFD suppressed apoptosis and increased damage prostate. On other hand, aerobic physical exercise improves prostatic tissue by increasing the response to anti-inflammatory and apoptosis protein.
Collapse
Affiliation(s)
- Maria Eduarda Almeida Tavares
- Department of Physical Education, São Paulo State University-UNESP, campus of Presidente Prudente, São Paulo, SP, Brazil; Multicenter Graduate Program in Physiological Sciences, SBFis, São Paulo State University (UNESP), Araçatuba, SP, Brazil; Experimental Laboratory of Exercise Biology (LEBioEx), São Paulo State University (UNESP), Presidente Prudent, SP, Brazil
| | - Allice Santos Cruz Veras
- Department of Physical Education, São Paulo State University-UNESP, campus of Presidente Prudente, São Paulo, SP, Brazil; Multicenter Graduate Program in Physiological Sciences, SBFis, São Paulo State University (UNESP), Araçatuba, SP, Brazil; Experimental Laboratory of Exercise Biology (LEBioEx), São Paulo State University (UNESP), Presidente Prudent, SP, Brazil
| | | | - Danilo Bianchini Baptista
- Experimental Laboratory of Exercise Biology (LEBioEx), São Paulo State University (UNESP), Presidente Prudent, SP, Brazil
| | - Giovana Rampazzo Teixeira
- Department of Physical Education, São Paulo State University-UNESP, campus of Presidente Prudente, São Paulo, SP, Brazil; Multicenter Graduate Program in Physiological Sciences, SBFis, São Paulo State University (UNESP), Araçatuba, SP, Brazil; Experimental Laboratory of Exercise Biology (LEBioEx), São Paulo State University (UNESP), Presidente Prudent, SP, Brazil.
| |
Collapse
|
26
|
Dai J, Wang H, Liao Y, Tan L, Sun Y, Song C, Liu W, Ding C, Luo T, Qiu X. Non-Targeted Metabolomic Analysis of Chicken Kidneys in Response to Coronavirus IBV Infection Under Stress Induced by Dexamethasone. Front Cell Infect Microbiol 2022; 12:945865. [PMID: 35909955 PMCID: PMC9335950 DOI: 10.3389/fcimb.2022.945865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Stress in poultry can lead to changes in body metabolism and immunity, which can increase susceptibility to infectious diseases. However, knowledge regarding chicken responses to viral infection under stress is limited. Dexamethasone (Dex) is a synthetic glucocorticoid similar to that secreted by animals under stress conditions, and has been widely used to induce stress in chickens. Herein, we established a stress model in 7-day-old chickens injected with Dex to elucidate the effects of stress on IBV replication in the kidneys. The metabolic changes, immune status and growth of the chickens under stress conditions were comprehensively evaluated. Furthermore, the metabolic profile, weight gain, viral load, serum cholesterol levels, cytokines and peripheral blood lymphocyte ratio were compared in chickens treated with Dex and infected with IBV. An LC-MS/MS-based metabolomics method was used to examine differentially enriched metabolites in the kidneys. A total of 113 metabolites whose abundance was altered after Dex treatment were identified, most of which were lipids and lipid-like molecules. The principal metabolic alterations in chicken kidneys caused by IBV infection included fatty acid, valine, leucine and isoleucine metabolism. Dex treatment before and after IBV infection mainly affected the host’s tryptophan, phenylalanine, amino sugar and nucleotide sugar metabolism. In addition, Dex led to up-regulation of serum cholesterol levels and renal viral load in chickens, and to the inhibition of weight gain, peripheral blood lymphocytes and IL-6 production. We also confirmed that the exogenous cholesterol in DF-1 cells promoted the replication of IBV. However, whether the increase in viral load in kidney tissue is associated with the up-regulation of cholesterol levels induced by Dex must be demonstrated in future experiments. In conclusion, chick growth and immune function were significantly inhibited by Dex. Host cholesterol metabolism and the response to IBV infection are regulated by Dex. This study provides valuable insights into the molecular regulatory mechanisms in poultry stress, and should support further research on the intrinsic link between cholesterol metabolism and IBV replication under stress conditions.
Collapse
Affiliation(s)
- Jun Dai
- Laboratory of Veterinary Microbiology and Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Huan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Lei Tan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yingjie Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Cuiping Song
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Weiwei Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Tingrong Luo
- Laboratory of Veterinary Microbiology and Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China
- *Correspondence: Xusheng Qiu, ; Tingrong Luo,
| | - Xusheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- *Correspondence: Xusheng Qiu, ; Tingrong Luo,
| |
Collapse
|
27
|
Shah MA, Haris M, Faheem HI, Hamid A, Yousaf R, Rasul A, Shah GM, Khalil AAK, Wahab A, Khan H, Alhasani RH, Althobaiti NA. Cross-Talk between Obesity and Diabetes: Introducing Polyphenols as an Effective Phytomedicine to Combat the Dual Sword Diabesity. Curr Pharm Des 2022; 28:1523-1542. [PMID: 35762558 DOI: 10.2174/1381612828666220628123224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/06/2022] [Indexed: 12/15/2022]
Abstract
: Obesity-associated diabetes mellitus, a chronic metabolic affliction accounting for 90% of all diabetic patients, has been affecting humanity extremely badly and escalating the risk of developing other serious disorders. It is observed that 0.4 billion people globally have diabetes, whose major cause is obesity. Currently, innumerable synthetic drugs like alogliptin and rosiglitazone are being used to get through diabetes, but they have certain complications, restrictions with severe side effects, and toxicity issues. Recently, the frequency of plant-derived phytochemicals as advantageous substitutes against diabesity is increasing progressively due to their unparalleled benefit of producing less side effects and toxicity. Of these phytochemicals, dietary polyphenols have been accepted as potent agents against the dual sword "diabesity". These polyphenols target certain genes and molecular pathways through dual mechanisms such as adiponectin upregulation, cannabinoid receptor antagonism, free fatty acid oxidation, ghrelin antagonism, glucocorticoid inhibition, sodium-glucose cotransporter inhibition, oxidative stress and inflammation inhibition etc. which sequentially help to combat both diabetes and obesity. In this review, we have summarized the most beneficial natural polyphenols along with their complex molecular pathways during diabesity.
Collapse
Affiliation(s)
| | - Muhammad Haris
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Hafiza Ishmal Faheem
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Ayesha Hamid
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Rimsha Yousaf
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Ghulam Mujtaba Shah
- Department of Pharmacy, Hazara University, Mansehra, Pakistan.,Department of Botany, Hazara University, Mansehra, Pakistan
| | - Atif Ali Khan Khalil
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science & Technology, Kohat, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Reem Hasaballah Alhasani
- Department of Biology, Faculty of Applied Science, Umm Al-Qura University, 21961 Makkah, Saudi Arabia
| | - Nora A Althobaiti
- Department of Biology, College of Science and Humanities-Al Quwaiiyah, Shaqra University, Al Quwaiiyah, Saudi Arabia
| |
Collapse
|
28
|
Yang S, Liu T, Hu C, Li W, Meng Y, Li H, Song C, He C, Zhou Y, Fan Y. Ginsenoside Compound K Protects against Obesity through Pharmacological Targeting of Glucocorticoid Receptor to Activate Lipophagy and Lipid Metabolism. Pharmaceutics 2022; 14:pharmaceutics14061192. [PMID: 35745765 PMCID: PMC9231161 DOI: 10.3390/pharmaceutics14061192] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/10/2022] Open
Abstract
(1) Background: The glucocorticoid receptor (GR) plays a key role in lipid metabolism, but investigations of GR activation as a potential therapeutic approach have been hampered by a lack of selective agonists. Ginsenoside compound K (CK) is natural small molecule with a steroid-like structure that offers a variety of therapeutic benefits. Our study validates CK as a novel GR agonist for the treatment of obesity. (2) Methods: By using pulldown and RNA interference, we determined that CK binds to GR. The anti-obesity potential effects of CK were investigated in obese mice, including through whole-body energy homeostasis, glucose and insulin tolerance, and biochemical and proteomic analysis. Using chromatin immunoprecipitation, we identified GR binding sites upstream of lipase ATGL. (3) Results: We demonstrated that CK reduced the weight and blood lipids of mice more significantly than the drug Orlistat. Proteomics data showed that CK up-regulated autophagy regulatory proteins, enhanced fatty acid oxidation proteins, and decreased fatty acid synthesis proteins. CK induced lipophagy with the initial formation of the phagophore via AMPK/ULK1 activation. However, a blockade of autophagy did not disturb the increase in CK on lipase expression, suggesting that autophagy and lipase are independent pathways in the function of CK. The pulldown and siRNA experiments showed that GR is the critical target. After binding to GR, CK not only activated lipophagy, but also promoted the binding of GR to the ATGL promoter. (4) Conclusions: Our findings indicate that CK is a natural food candidate for reducing fat content and weight.
Collapse
Affiliation(s)
- Siwen Yang
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Ting Liu
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Chenxing Hu
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Weili Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Yuhan Meng
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Haiyang Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Chengcheng Song
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
| | - Congcong He
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Yifa Zhou
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
- Correspondence: (Y.Z.); (Y.F.)
| | - Yuying Fan
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China; (S.Y.); (T.L.); (C.H.); (W.L.); (Y.M.); (H.L.); (C.S.)
- Correspondence: (Y.Z.); (Y.F.)
| |
Collapse
|
29
|
The newborn metabolome: associations with gestational diabetes, sex, gestation, birth mode, and birth weight. Pediatr Res 2022; 91:1864-1873. [PMID: 34526650 DOI: 10.1038/s41390-021-01672-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Pathways towards many adult-onset conditions begin early in life, even in utero. Maternal health in pregnancy influences this process, but little is known how it affects neonatal metabolism. We investigated associations between pregnancy and birth factors and cord blood metabolomic profile in a large, population-derived cohort. METHODS Metabolites were measured using nuclear magnetic resonance in maternal (28 weeks gestation) and cord serum from 912 mother-child pairs in the Barwon Infant Study pre-birth cohort. Associations between maternal (metabolites, age, BMI, smoking), pregnancy (pre-eclampsia, gestational diabetes (GDM)), and birth characteristics (delivery mode, gestational age, weight, infant sex) with 72 cord blood metabolites were examined by linear regression. RESULTS Delivery mode, sex, gestational age, and birth weight were associated with specific metabolite levels in cord blood, including amino acids, fatty acids, and cholesterols. GDM was associated with higher cord blood levels of acetoacetate and 3-hydroxybutyrate. CONCLUSIONS Neonatal factors, particularly delivery mode, were associated with many cord blood metabolite differences, including those implicated in later risk of cardiometabolic disease. Associations between GDM and higher offspring ketone levels at birth are consistent with maternal ketosis in diabetic pregnancies. Further work is needed to determine whether these neonatal metabolome differences associate with later health outcomes. IMPACT Variations in blood metabolomic profile have been linked to health status in adults and children, but corresponding data in neonates are scarce. We report evidence that pregnancy complications, mode of delivery, and offspring characteristics, including sex, are independently associated with a range of circulating metabolites at birth, including ketone bodies, amino acids, cholesterols, and inflammatory markers. Independent of birth weight, exposure to gestational diabetes is associated with higher cord blood ketone bodies and citrate. These findings suggest that pregnancy complications, mode of delivery, gestational age, and measures of growth influence metabolic pathways prior to birth, potentially impacting later health and development.
Collapse
|
30
|
Lu H, Lei X, Winkler R, John S, Kumar D, Li W, Alnouti Y. Crosstalk of hepatocyte nuclear factor 4a and glucocorticoid receptor in the regulation of lipid metabolism in mice fed a high-fat-high-sugar diet. Lipids Health Dis 2022; 21:46. [PMID: 35614477 PMCID: PMC9134643 DOI: 10.1186/s12944-022-01654-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/06/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Hepatocyte nuclear factor 4α (HNF4α) and glucocorticoid receptor (GR), master regulators of liver metabolism, are down-regulated in fatty liver diseases. The present study aimed to elucidate the role of down-regulation of HNF4α and GR in fatty liver and hyperlipidemia. METHODS Adult mice with liver-specific heterozygote (HET) and knockout (KO) of HNF4α or GR were fed a high-fat-high-sugar diet (HFHS) for 15 days. Alterations in hepatic and circulating lipids were determined with analytical kits, and changes in hepatic mRNA and protein expression in these mice were quantified by real-time PCR and Western blotting. Serum and hepatic levels of bile acids were quantified by LC-MS/MS. The roles of HNF4α and GR in regulating hepatic gene expression were determined using luciferase reporter assays. RESULTS Compared to HFHS-fed wildtype mice, HNF4α HET mice had down-regulation of lipid catabolic genes, induction of lipogenic genes, and increased hepatic and blood levels of lipids, whereas HNF4α KO mice had fatty liver but mild hypolipidemia, down-regulation of lipid-efflux genes, and induction of genes for uptake, synthesis, and storage of lipids. Serum levels of chenodeoxycholic acid and deoxycholic acid tended to be decreased in the HNF4α HET mice but dramatically increased in the HNF4α KO mice, which was associated with marked down-regulation of cytochrome P450 7a1, the rate-limiting enzyme for bile acid synthesis. Hepatic mRNA and protein expression of sterol-regulatory-element-binding protein-1 (SREBP-1), a master lipogenic regulator, was induced in HFHS-fed HNF4α HET mice. In reporter assays, HNF4α cooperated with the corepressor small heterodimer partner to potently inhibit the transactivation of mouse and human SREBP-1C promoter by liver X receptor. Hepatic nuclear GR proteins tended to be decreased in the HNF4α KO mice. HFHS-fed mice with liver-specific KO of GR had increased hepatic lipids and induction of SREBP-1C and PPARγ, which was associated with a marked decrease in hepatic levels of HNF4α proteins in these mice. In reporter assays, GR and HNF4α synergistically/additively induced lipid catabolic genes. CONCLUSIONS induction of lipid catabolic genes and suppression of lipogenic genes by HNF4α and GR may mediate the early resistance to HFHS-induced fatty liver and hyperlipidemia.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Xiaohong Lei
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Rebecca Winkler
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Savio John
- Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Devendra Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wenkuan Li
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
31
|
Lustig RH, Collier D, Kassotis C, Roepke TA, Ji Kim M, Blanc E, Barouki R, Bansal A, Cave MC, Chatterjee S, Choudhury M, Gilbertson M, Lagadic-Gossmann D, Howard S, Lind L, Tomlinson CR, Vondracek J, Heindel JJ. Obesity I: Overview and molecular and biochemical mechanisms. Biochem Pharmacol 2022; 199:115012. [PMID: 35393120 PMCID: PMC9050949 DOI: 10.1016/j.bcp.2022.115012] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a chronic, relapsing condition characterized by excess body fat. Its prevalence has increased globally since the 1970s, and the number of obese and overweight people is now greater than those underweight. Obesity is a multifactorial condition, and as such, many components contribute to its development and pathogenesis. This is the first of three companion reviews that consider obesity. This review focuses on the genetics, viruses, insulin resistance, inflammation, gut microbiome, and circadian rhythms that promote obesity, along with hormones, growth factors, and organs and tissues that control its development. It shows that the regulation of energy balance (intake vs. expenditure) relies on the interplay of a variety of hormones from adipose tissue, gastrointestinal tract, pancreas, liver, and brain. It details how integrating central neurotransmitters and peripheral metabolic signals (e.g., leptin, insulin, ghrelin, peptide YY3-36) is essential for controlling energy homeostasis and feeding behavior. It describes the distinct types of adipocytes and how fat cell development is controlled by hormones and growth factors acting via a variety of receptors, including peroxisome proliferator-activated receptor-gamma, retinoid X, insulin, estrogen, androgen, glucocorticoid, thyroid hormone, liver X, constitutive androstane, pregnane X, farnesoid, and aryl hydrocarbon receptors. Finally, it demonstrates that obesity likely has origins in utero. Understanding these biochemical drivers of adiposity and metabolic dysfunction throughout the life cycle lends plausibility and credence to the "obesogen hypothesis" (i.e., the importance of environmental chemicals that disrupt these receptors to promote adiposity or alter metabolism), elucidated more fully in the two companion reviews.
Collapse
Affiliation(s)
- Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California, San Francisco, CA 94143, United States
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Christopher Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, United States
| | - Troy A Roepke
- School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, United States
| | - Min Ji Kim
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Etienne Blanc
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Robert Barouki
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, United States
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, United States
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, United States
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland, United Kingdom
| | - Dominique Lagadic-Gossmann
- Research Institute for Environmental and Occupational Health, University of Rennes, INSERM, EHESP, Rennes, France
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| | - Lars Lind
- Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States.
| |
Collapse
|
32
|
Sun W, Li P, Cai J, Ma J, Zhang X, Song Y, Liu Y. Lipid Metabolism: Immune Regulation and Therapeutic Prospectives in Systemic Lupus Erythematosus. Front Immunol 2022; 13:860586. [PMID: 35371016 PMCID: PMC8971568 DOI: 10.3389/fimmu.2022.860586] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous disease characterized by the production of abnormal autoantibodies and immune complexes that can affect the organ and organ systems, particularly the kidneys and the cardiovascular system. Emerging evidence suggests that dysregulated lipid metabolism, especially in key effector cells, such as T cells, B cells, and innate immune cells, exerts complex effects on the pathogenesis and progression of SLE. Beyond their important roles as membrane components and energy storage, different lipids can also modulate different cellular processes, such as proliferation, differentiation, and survival. In this review, we summarize altered lipid metabolism and the associated mechanisms involved in the pathogenesis and progression of SLE. Furthermore, we discuss the recent progress in the role of lipid metabolism as a potential therapeutic target in SLE.
Collapse
Affiliation(s)
- Wei Sun
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Southeast University, Nanjing, China
| | - Pengchong Li
- Department of Rheumatology and Clinical Immunology, The Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Beijing, China
- Department of Gastroenterology, Beijing Friendship Hospital, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Capital Medical University, Beijing, China
| | - Jianping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontolog, Beijing, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Southeast University, Nanjing, China
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
- *Correspondence: Yudong Liu, ; Yong Song,
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Center of Biotherapy, Beijing Hospital, National Center of Gerontolog, Beijing, China
- *Correspondence: Yudong Liu, ; Yong Song,
| |
Collapse
|
33
|
Lavin KM, Coen PM, Baptista LC, Bell MB, Drummer D, Harper SA, Lixandrão ME, McAdam JS, O’Bryan SM, Ramos S, Roberts LM, Vega RB, Goodpaster BH, Bamman MM, Buford TW. State of Knowledge on Molecular Adaptations to Exercise in Humans: Historical Perspectives and Future Directions. Compr Physiol 2022; 12:3193-3279. [PMID: 35578962 PMCID: PMC9186317 DOI: 10.1002/cphy.c200033] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
For centuries, regular exercise has been acknowledged as a potent stimulus to promote, maintain, and restore healthy functioning of nearly every physiological system of the human body. With advancing understanding of the complexity of human physiology, continually evolving methodological possibilities, and an increasingly dire public health situation, the study of exercise as a preventative or therapeutic treatment has never been more interdisciplinary, or more impactful. During the early stages of the NIH Common Fund Molecular Transducers of Physical Activity Consortium (MoTrPAC) Initiative, the field is well-positioned to build substantially upon the existing understanding of the mechanisms underlying benefits associated with exercise. Thus, we present a comprehensive body of the knowledge detailing the current literature basis surrounding the molecular adaptations to exercise in humans to provide a view of the state of the field at this critical juncture, as well as a resource for scientists bringing external expertise to the field of exercise physiology. In reviewing current literature related to molecular and cellular processes underlying exercise-induced benefits and adaptations, we also draw attention to existing knowledge gaps warranting continued research effort. © 2021 American Physiological Society. Compr Physiol 12:3193-3279, 2022.
Collapse
Affiliation(s)
- Kaleen M. Lavin
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Human Health, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, Florida, USA
| | - Paul M. Coen
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Liliana C. Baptista
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Margaret B. Bell
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Devin Drummer
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sara A. Harper
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manoel E. Lixandrão
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeremy S. McAdam
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samia M. O’Bryan
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sofhia Ramos
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Lisa M. Roberts
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rick B. Vega
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Bret H. Goodpaster
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Marcas M. Bamman
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Human Health, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, Florida, USA
| | - Thomas W. Buford
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
34
|
Völker J, Ashcroft F, Vedøy Å, Zimmermann L, Wagner M. Adipogenic Activity of Chemicals Used in Plastic Consumer Products. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022. [PMID: 35080176 DOI: 10.1101/2021.07.29.454199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Bisphenols and phthalates, chemicals frequently used in plastic products, promote obesity in cell and animal models. However, these well-known metabolism-disrupting chemicals (MDCs) represent only a minute fraction of all compounds found in plastics. To gain a comprehensive understanding of plastics as a source of exposure to MDCs, we characterized the chemicals present in 34 everyday products using nontarget high-resolution mass spectrometry and analyzed their joint adipogenic activities by high-content imaging. We detected 55,300 chemical features and tentatively identified 629 unique compounds, including 11 known MDCs. Importantly, the chemicals extracted from one-third of the products caused murine 3T3-L1 preadipocytes to proliferate, and differentiate into adipocytes, which were larger and contained more triglycerides than those treated with the reference compound rosiglitazone. Because the majority of plastic extracts did not activate the peroxisome proliferator-activated receptor γ and the glucocorticoid receptor, the adipogenic effects are mediated via other mechanisms and, thus, likely to be caused by unknown MDCs. Our study demonstrates that daily-use plastics contain potent mixtures of MDCs and can, therefore, be a relevant yet underestimated environmental factor contributing to obesity.
Collapse
Affiliation(s)
- Johannes Völker
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Felicity Ashcroft
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Åsa Vedøy
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Lisa Zimmermann
- Department of Aquatic Ecotoxicology, Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - Martin Wagner
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| |
Collapse
|
35
|
Völker J, Ashcroft F, Vedøy Å, Zimmermann L, Wagner M. Adipogenic Activity of Chemicals Used in Plastic Consumer Products. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:2487-2496. [PMID: 35080176 PMCID: PMC8851687 DOI: 10.1021/acs.est.1c06316] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/03/2021] [Accepted: 12/27/2021] [Indexed: 05/28/2023]
Abstract
Bisphenols and phthalates, chemicals frequently used in plastic products, promote obesity in cell and animal models. However, these well-known metabolism-disrupting chemicals (MDCs) represent only a minute fraction of all compounds found in plastics. To gain a comprehensive understanding of plastics as a source of exposure to MDCs, we characterized the chemicals present in 34 everyday products using nontarget high-resolution mass spectrometry and analyzed their joint adipogenic activities by high-content imaging. We detected 55,300 chemical features and tentatively identified 629 unique compounds, including 11 known MDCs. Importantly, the chemicals extracted from one-third of the products caused murine 3T3-L1 preadipocytes to proliferate, and differentiate into adipocytes, which were larger and contained more triglycerides than those treated with the reference compound rosiglitazone. Because the majority of plastic extracts did not activate the peroxisome proliferator-activated receptor γ and the glucocorticoid receptor, the adipogenic effects are mediated via other mechanisms and, thus, likely to be caused by unknown MDCs. Our study demonstrates that daily-use plastics contain potent mixtures of MDCs and can, therefore, be a relevant yet underestimated environmental factor contributing to obesity.
Collapse
Affiliation(s)
- Johannes Völker
- Department
of Biology, Norwegian University of Science
and Technology (NTNU), 7491 Trondheim, Norway
| | - Felicity Ashcroft
- Department
of Biology, Norwegian University of Science
and Technology (NTNU), 7491 Trondheim, Norway
| | - Åsa Vedøy
- Department
of Biology, Norwegian University of Science
and Technology (NTNU), 7491 Trondheim, Norway
| | - Lisa Zimmermann
- Department
of Aquatic Ecotoxicology, Goethe University
Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - Martin Wagner
- Department
of Biology, Norwegian University of Science
and Technology (NTNU), 7491 Trondheim, Norway
| |
Collapse
|
36
|
Reactive Oxygen Species (ROS) and Antioxidants as Immunomodulators in Exercise: Implications for Heme Oxygenase and Bilirubin. Antioxidants (Basel) 2022; 11:antiox11020179. [PMID: 35204062 PMCID: PMC8868548 DOI: 10.3390/antiox11020179] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
Exercise is commonly prescribed as a lifestyle treatment for chronic metabolic diseases as it functions as an insulin sensitizer, cardio-protectant, and essential lifestyle tool for effective weight maintenance. Exercise boosts the production of reactive oxygen species (ROS) and subsequent transient oxidative damage, which also upregulates counterbalancing endogenous antioxidants to protect from ROS-induced damage and inflammation. Exercise elevates heme oxygenase-1 (HO-1) and biliverdin reductase A (BVRA) expression as built-in protective mechanisms, which produce the most potent antioxidant, bilirubin. Together, these mitigate inflammation and adiposity. Moderately raising plasma bilirubin protects in two ways: (1) via its antioxidant capacity to reduce ROS and inflammation, and (2) its newly defined function as a hormone that activates the nuclear receptor transcription factor PPARα. It is now understood that increasing plasma bilirubin can also drive metabolic adaptions, which improve deleterious outcomes of weight gain and obesity, such as inflammation, type II diabetes, and cardiovascular diseases. The main objective of this review is to describe the function of bilirubin as an antioxidant and metabolic hormone and how the HO-1-BVRA-bilirubin-PPARα axis influences inflammation, metabolic function and interacts with exercise to improve outcomes of weight management.
Collapse
|
37
|
Tsai SF, Hung HC, Shih MMC, Chang FC, Chung BC, Wang CY, Lin YL, Kuo YM. High-fat diet-induced increases in glucocorticoids contribute to the development of non-alcoholic fatty liver disease in mice. FASEB J 2021; 36:e22130. [PMID: 34959259 DOI: 10.1096/fj.202101570r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022]
Abstract
This study aimed to investigate the causal relationship between chronic ingestion of a high-fat diet (HFD)-induced secretion of glucocorticoids (GCs) and the development of non-alcoholic fatty liver disease (NAFLD). We have produced a strain of transgenic mice (termed L/L mice) that have normal levels of circulating corticosterone (CORT), the major type of GCs in rodents, but unlike wild-type (WT) mice, their circulating CORT was not affected by HFD. Compared to WT mice, 12-week HFD-induced fatty liver was less pronounced with higher plasma levels of triglycerides in L/L mice. These changes were reversed by CORT supplement to L/L mice. By analyzing a sort of lipid metabolism-related proteins, we found that expressions of the hepatic cluster of differentiation 36 (CD36) were upregulated by HFD-induced CORT and involved in CORT-mediated fatty liver. Dexamethasone, an agonist of the glucocorticoid receptor (GR), upregulated expressions of CD36 in HepG2 hepatocytes and facilitated lipid accumulation in the cells. In conclusion, the fat ingestion-induced release of CORT contributes to NAFLD. This study highlights the pathogenic role of CORT-mediated upregulation of hepatic CD 36 in diet-induced NAFLD.
Collapse
Affiliation(s)
- Sheng-Feng Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hao-Chang Hung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | | | - Fu-Chuan Chang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bon-Chu Chung
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chia-Yih Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ling Lin
- Division of Gastroenterology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
38
|
Hinds TD, Kipp ZA, Xu M, Yiannikouris FB, Morris AJ, Stec DF, Wahli W, Stec DE. Adipose-Specific PPARα Knockout Mice Have Increased Lipogenesis by PASK-SREBP1 Signaling and a Polarity Shift to Inflammatory Macrophages in White Adipose Tissue. Cells 2021; 11:4. [PMID: 35011564 PMCID: PMC8750478 DOI: 10.3390/cells11010004] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/16/2022] Open
Abstract
The nuclear receptor PPARα is associated with reducing adiposity, especially in the liver, where it transactivates genes for β-oxidation. Contrarily, the function of PPARα in extrahepatic tissues is less known. Therefore, we established the first adipose-specific PPARα knockout (PparaFatKO) mice to determine the signaling position of PPARα in adipose tissue expansion that occurs during the development of obesity. To assess the function of PPARα in adiposity, female and male mice were placed on a high-fat diet (HFD) or normal chow for 30 weeks. Only the male PparaFatKO animals had significantly more adiposity in the inguinal white adipose tissue (iWAT) and brown adipose tissue (BAT) with HFD, compared to control littermates. No changes in adiposity were observed in female mice compared to control littermates. In the males, the loss of PPARα signaling in adipocytes caused significantly higher cholesterol esters, activation of the transcription factor sterol regulatory element-binding protein-1 (SREBP-1), and a shift in macrophage polarity from M2 to M1 macrophages. We found that the loss of adipocyte PPARα caused significantly higher expression of the Per-Arnt-Sim kinase (PASK), a kinase that activates SREBP-1. The hyperactivity of the PASK-SREBP-1 axis significantly increased the lipogenesis proteins fatty acid synthase (FAS) and stearoyl-Coenzyme A desaturase 1 (SCD1) and raised the expression of genes for cholesterol metabolism (Scarb1, Abcg1, and Abca1). The loss of adipocyte PPARα increased Nos2 in the males, an M1 macrophage marker indicating that the population of macrophages had changed to proinflammatory. Our results demonstrate the first adipose-specific actions for PPARα in protecting against lipogenesis, inflammation, and cholesterol ester accumulation that leads to adipocyte tissue expansion in obesity.
Collapse
Affiliation(s)
- Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA; (Z.A.K.); (M.X.); (F.B.Y.)
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Zachary A. Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA; (Z.A.K.); (M.X.); (F.B.Y.)
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA; (Z.A.K.); (M.X.); (F.B.Y.)
| | - Frederique B. Yiannikouris
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40508, USA; (Z.A.K.); (M.X.); (F.B.Y.)
- Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY 40508, USA
| | - Andrew J. Morris
- Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY 40508, USA;
- Lexington Veterans Affairs Medical Center, Lexington, KY 40508, USA
| | - Donald F. Stec
- Small Molecule NMR Facility Core, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA;
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, Singapore 308232, Singapore;
- Toxalim Research Center in Food Toxicology (UMR 1331), INRAE, ENVT, INP—PURPAN, UPS, Université de Toulouse, F-31300 Toulouse, France
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| | - David E. Stec
- Department of Physiology & Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
39
|
Dashti-Khavidaki S, Saidi R, Lu H. Current status of glucocorticoid usage in solid organ transplantation. World J Transplant 2021; 11:443-465. [PMID: 34868896 PMCID: PMC8603633 DOI: 10.5500/wjt.v11.i11.443] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/16/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoids (GCs) have been the mainstay of immunosuppressive therapy in solid organ transplantation (SOT) for decades, due to their potent effects on innate immunity and tissue protective effects. However, some SOT centers are reluctant to administer GCs long-term because of the various related side effects. This review summarizes the advantages and disadvantages of GCs in SOT. PubMed and Scopus databases were searched from 2011 to April 2021 using search syntaxes covering “transplantation” and “glucocorticoids”. GCs are used in transplant recipients, transplant donors, and organ perfusate solution to improve transplant outcomes. In SOT recipients, GCs are administered as induction and maintenance immunosuppressive therapy. GCs are also the cornerstone to treat acute antibody- and T-cell-mediated rejections. Addition of GCs to organ perfusate solution and pretreatment of transplant donors with GCs are recommended by some guidelines and protocols, to reduce ischemia-reperfusion injury peri-transplant. GCs with low bioavailability and high potency for GC receptors, such as budesonide, nanoparticle-mediated targeted delivery of GCs to specific organs, and combination use of dexamethasone with inducers of immune-regulatory cells, are new methods of GC application in SOT patients to reduce side effects or induce immune-tolerance instead of immunosuppression. Various side effects involving different non-targeted organs/tissues, such as bone, cardiovascular, neuromuscular, skin and gastrointestinal tract, have been noted for GCs. There are also potential drug-drug interactions for GCs in SOT patients.
Collapse
Affiliation(s)
- Simin Dashti-Khavidaki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155, Iran
| | - Reza Saidi
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| |
Collapse
|
40
|
Rondini EA, Ramseyer VD, Burl RB, Pique-Regi R, Granneman JG. Single cell functional genomics reveals plasticity of subcutaneous white adipose tissue (WAT) during early postnatal development. Mol Metab 2021; 53:101307. [PMID: 34298199 PMCID: PMC8385178 DOI: 10.1016/j.molmet.2021.101307] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The current study addresses the cellular complexity and plasticity of subcutaneous (inguinal) white adipose tissue (iWAT) in mice during the critical periods of perinatal growth and establishment. METHODS We performed a large-scale single cell transcriptomic (scRNA-seq) and epigenomic (snATAC-seq) characterization of cellular subtypes (adipose stromal cells (ASC) and adipocyte nuclei) during inguinal WAT (subcutaneous; iWAT) development in mice, capturing the early postnatal period (postnatal days (PND) 06 and 18) through adulthood (PND56). RESULTS Perinatal and adult iWAT contain 3 major ASC subtypes that can be independently identified by RNA expression profiles and DNA transposase accessibility. Furthermore, the transcriptomes and enhancer landscapes of both ASC and adipocytes dynamically change during postnatal development. Perinatal ASC (PND06) are highly enriched for several imprinted genes (IGs; e.g., Mest, H19, Igf2) and extracellular matrix proteins whose expression then declines prior to weaning (PND18). By comparison, adult ASC (PND56) are more enriched for transcripts associated with immunoregulation, oxidative stress, and integrin signaling. Two clusters of mature adipocytes, identified through single nuclei RNA sequencing (snRNA-seq), were distinctive for proinflammatory/immune or metabolic gene expression patterns that became more transcriptionally diverse in adult animals. Single nuclei assay for transposase-accessible chromatin (snATAC-seq) revealed that differences in gene expression were associated with developmental changes in chromatin accessibility and predicted transcription factor motifs (e.g., Plagl1, Ar) in both stromal cells and adipocytes. CONCLUSIONS Our data provide new insights into transcriptional and epigenomic signaling networks important during iWAT establishment at a single cell resolution, with important implications for the field of metabolic programming.
Collapse
Affiliation(s)
- Elizabeth A Rondini
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Vanesa D Ramseyer
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Rayanne B Burl
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Center for Integrative Metabolic and Endocrine Research, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
41
|
Smedlund KB, Sanchez ER, Hinds TD. FKBP51 and the molecular chaperoning of metabolism. Trends Endocrinol Metab 2021; 32:862-874. [PMID: 34481731 PMCID: PMC8516732 DOI: 10.1016/j.tem.2021.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/31/2021] [Accepted: 08/06/2021] [Indexed: 01/30/2023]
Abstract
The molecular chaperone FK506-binding protein 51 (FKBP51) is gaining attention as a meaningful biomarker of metabolic dysfunction. This review examines the emerging contributions of FKBP51 in adipogenesis and lipid metabolism, myogenesis and protein catabolism, and glucocorticoid-induced skin hypoplasia and dermal adipocytes. The FKBP51 signaling mechanisms that may explain these metabolic consequences are discussed. These mechanisms are diverse, with FKBP51 independently and directly regulating phosphorylation cascades and nuclear receptors. We provide a discussion of the newly developed compounds that antagonize FKBP51, which may offer therapeutic advantages for adiposity. These observations suggest we are only beginning to uncover the complex nature of FKBP51 and its molecular chaperoning of metabolism.
Collapse
Affiliation(s)
- Kathryn B Smedlund
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Edwin R Sanchez
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Terry D Hinds
- Barnstable Brown Diabetes Center, Markey Cancer Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA.
| |
Collapse
|
42
|
Eley VA, Thuzar M, Navarro S, Dodd BR, Zundert AAV. Obesity, metabolic syndrome, and inflammation: an update for anaesthetists caring for patients with obesity. Anaesth Crit Care Pain Med 2021; 40:100947. [PMID: 34534700 DOI: 10.1016/j.accpm.2021.100947] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/11/2021] [Accepted: 03/20/2021] [Indexed: 11/25/2022]
Abstract
Our understanding of chronic inflammation in obesity is evolving. Suggested mechanisms include hypoxia of adipose tissue and a subsequent increase in circulating cytokines. It is now known that adipose tissue, far from being an inert tissue, produces and secretes multiple peptides that influence inflammation and metabolism, including substrates of the renin-angiotensin-aldosterone system (RAAS). RAAS blocking antihypertensive medication and cholesterol-lowering agents are now being evaluated for their metabolic and inflammation-modulating effects. Surgery also has pro-inflammatory effects, which may be exacerbated in patients with obesity. This narrative review will summarise the recent literature surrounding obesity, metabolic syndrome, inflammation, and interplay with the RAAS, with evidence-based recommendations for the optimisation of patients with obesity, prior to surgery and anaesthesia.
Collapse
Affiliation(s)
- Victoria A Eley
- Department of Anaesthesia and Perioperative Medicine, The Royal Brisbane and Women's Hospital, Butterfield St, Herston, 4006 Queensland, Australia; Faculty of Medicine, The University of Queensland, St Lucia, 4067 Queensland, Australia.
| | - Moe Thuzar
- Faculty of Medicine, The University of Queensland, St Lucia, 4067 Queensland, Australia; Department of Endocrinology and Diabetes, Princess Alexandra Hospital, Ipswich Road Woolloongabba, 4102 Queensland, Australia; Endocrine Hypertension Research Centre, The University of Queensland Diamantina Institute, Ipswich Road Woolloongabba, 4102 Queensland, Australia
| | - Séverine Navarro
- Department of Immunology, QIMR Berghofer Medical Research Institute Herston Rd, Herston, 4006 Queensland, Australia; Woolworths Centre for Childhood Nutrition Research, Faculty of Health, School of Exercise and Nutrition Sciences, Queensland University of Technology, Kelvin Grove, 4059 Queensland, Australia
| | - Benjamin R Dodd
- Faculty of Medicine, The University of Queensland, St Lucia, 4067 Queensland, Australia; Department of Upper GI and Bariatric Surgery, The Royal Brisbane and Women's Hospital, Butterfield St, Herston, 4006 Queensland, Australia
| | - André A Van Zundert
- Department of Anaesthesia and Perioperative Medicine, The Royal Brisbane and Women's Hospital, Butterfield St, Herston, 4006 Queensland, Australia; Faculty of Medicine, The University of Queensland, St Lucia, 4067 Queensland, Australia
| |
Collapse
|
43
|
Di S, Han L, An X, Kong R, Gao Z, Yang Y, Wang X, Zhang P, Ding Q, Wu H, Wang H, Zhao L, Tong X. In silico network pharmacology and in vivo analysis of berberine-related mechanisms against type 2 diabetes mellitus and its complications. JOURNAL OF ETHNOPHARMACOLOGY 2021; 276:114180. [PMID: 33957209 DOI: 10.1016/j.jep.2021.114180] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 04/24/2021] [Accepted: 04/28/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Berberine (BBR), extracted from the traditional medicinal plant Coptis chinensis Franch., has been widely used for the treatment of type 2 diabetes mellitus (T2DM) and its complications. AIM OF THE STUDY To determine the potential pharmacological mechanisms underlying BBR therapeutic effect on T2DM and its complications by in silico network pharmacology and experimental in vivo validation. MATERIALS AND METHODS A predictive network depicting the relationship between BBR and T2DM was designed based on information collected from several databases, namely STITCH, CHEMBL, PharmMapper, TTD, Drugbank, and PharmGKB. Identified overlapping targets related to both BBR and T2DM were crossed with information on biological processes (BPs) and molecular/signaling pathways using the DAVID platform and Cytoscape software. Three candidate targets identified with the BBR-T2DM network (RXRA, KCNQ1 and NR3C1) were evaluated in the C57BL/6J mouse model of T2DM. The mice were treated with BBR or metformin for 10 weeks. Weight, fasting blood glucose (FBG), oral glucose tolerance, and expression levels of the three targets were evaluated. RESULTS A total of 31 targets of BBR that were also related to T2DM were identified, of which 14 had already been reported in previous studies. Furthermore, these 31 overlapping targets were enriched in 21 related BPs and 18 pathways involved in T2DM treatment. The identified BP-target-pathway network revealed the underlying mechanisms of BBR antidiabetic activity were mediated by core targets such as RXRA, KCNQ1, and NR3C1. In vivo experiments further confirmed that treatment with BBR significantly reduced weight and FBG and alleviated insulin resistance in T2DM mice. Moreover, BBR treatment promoted RXRA expression, whereas it reduced KCNQ1 and NR3C1 expression in the liver. CONCLUSION Using network pharmacology and a T2DM mouse model, this study revealed that BBR can effectively prevent T2DM symptoms through vital targets and multiple signaling pathways. Network pharmacology provides an efficient, time-saving approach for therapeutic research and the development of new drugs.
Collapse
Affiliation(s)
- Sha Di
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Lin Han
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China; Laboratory of Molecular and Biology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Xuedong An
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Ran Kong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
| | - Zezheng Gao
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yingying Yang
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Xinmiao Wang
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China; Endocrinology Department, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China.
| | - Pei Zhang
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Qiyou Ding
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Haoran Wu
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Han Wang
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Linhua Zhao
- Department of Endocrinology, Guang'anmen Hospital of China, Academy of Chinese Medical Sciences, Beijing, 100053, China; Endocrinology Department, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China.
| | - Xiaolin Tong
- Endocrinology Department, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China.
| |
Collapse
|
44
|
Van Wyngene L, Vanderhaeghen T, Petta I, Timmermans S, Corbeels K, Van der Schueren B, Vandewalle J, Van Looveren K, Wallaeys C, Eggermont M, Dewaele S, Catrysse L, van Loo G, Beyaert R, Vangoitsenhoven R, Nakayama T, Tavernier J, De Bosscher K, Libert C. ZBTB32 performs crosstalk with the glucocorticoid receptor and is crucial in glucocorticoid responses to starvation. iScience 2021; 24:102790. [PMID: 34337361 PMCID: PMC8324811 DOI: 10.1016/j.isci.2021.102790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 03/25/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022] Open
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis forms a complex neuroendocrine system that regulates the body’s response to stress such as starvation. In contrast with the glucocorticoid receptor (GR), Zinc finger and BTB domain containing 32 (ZBTB32) is a transcription factor with poorly described functional relevance in physiology. This study shows that ZBTB32 is essential for the production of glucocorticoids (GCs) in response to starvation, since ZBTB32−/− mice fail to increase their GC production in the absence of nutrients. In terms of mechanism, GR-mediated upregulation of adrenal Scarb1 gene expression was absent in ZBTB32−/− mice, implicating defective cholesterol import as the cause of the poor GC synthesis. These lower GC levels are further associated with aberrations in the metabolic adaptation to starvation, which could explain the progressive weight gain of ZBTB32−/− mice. In conclusion, ZBTB32 performs a crosstalk with the GR in the metabolic adaptation to starvation via regulation of adrenal GC production. ZBTB32 is involved in the glucocorticoid production in response to starvation GR-mediated upregulation of adrenal Scarb1 regulates cholesterol import The weight gain of ZBTB32−/− mice is associated with aberrant metabolic adaptations
Collapse
Affiliation(s)
- Lise Van Wyngene
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Tineke Vanderhaeghen
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Ioanna Petta
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Ghent Gut Inflammation Group (GGIG), Ghent University, 9000 Ghent, Belgium.,Department of Rheumatology, Ghent University, 9000 Ghent, Belgium
| | - Steven Timmermans
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Katrien Corbeels
- Department of Chronic Diseases and Metabolism - Endocrinology, KU Leuven, Leuven, Belgium
| | - Bart Van der Schueren
- Department of Chronic Diseases and Metabolism - Endocrinology, KU Leuven, Leuven, Belgium
| | - Jolien Vandewalle
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Kelly Van Looveren
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Charlotte Wallaeys
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Melanie Eggermont
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Sylviane Dewaele
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Leen Catrysse
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Geert van Loo
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.,Ghent Gut Inflammation Group (GGIG), Ghent University, 9000 Ghent, Belgium
| | - Rudi Beyaert
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.,Ghent Gut Inflammation Group (GGIG), Ghent University, 9000 Ghent, Belgium
| | - Roman Vangoitsenhoven
- Department of Chronic Diseases and Metabolism - Endocrinology, KU Leuven, Leuven, Belgium
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jan Tavernier
- Center for Medical Biotechnology, VIB Center for Medical Biotechnology, 9000 Ghent, Belgium.,Cytokine Receptor Laboratory (CRL), Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, 3 Albert Baertsoenkaai, 9000 Ghent, Belgium
| | - Karolien De Bosscher
- Center for Medical Biotechnology, VIB Center for Medical Biotechnology, 9000 Ghent, Belgium.,Translational Nuclear Receptor Research Lab, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, 3 Albert Baertsoenkaai,9000 Ghent, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB Center for Inflammation Research, 9000 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.,Ghent Gut Inflammation Group (GGIG), Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
45
|
Neonatal nicotine exposure changes insulin status in fat depots: sex-related differences. J Dev Orig Health Dis 2021; 13:252-262. [PMID: 33818369 DOI: 10.1017/s2040174421000131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nicotine is the main psychoactive substance present in cigarette smoke that is transferred to the baby by breast milk. In rats, maternal nicotine exposure during breastfeeding induces obesogenesis and hormone dysfunctions in adult male offspring. As glucocorticoid (GC), insulin, and vitamin D change both adipogenesis and lipogenesis processes, we assessed parameters related to metabolism and action of these hormones in visceral and subcutaneous adipose tissues (VAT and SAT) of adult male and female rats in a model of neonatal nicotine exposure. At postnatal (PN) day 2, dams were kept with six pups (three per sex) and divided into nicotine and control groups for implantation of osmotic minipumps that released 6 mg/kg nicotine or saline, respectively. At PN180, fat mass, hormone levels, and protein contents of biomarkers of the GC activation and receptor (11beta-hydroxysteroid dehydrogenase type 1 and glucocorticoid receptor alpha), insulin signaling pathway [insulin receptor beta (IRβ), phosphorylated insulin receptor substrate 1, insulin receptor substrate 1 (IRS1), phosphorylated serine/threonine kinase (pAKT), serine/threonine kinase, glucose transporter type 4 (GLUT4)], and vitamin D activation and receptor (1α-hydroxylase and vitamin D receptor) were evaluated. While nicotine-exposed males showed increased fat mass, hypercorticosteronemia, hyperinsulinemia, and higher 25-hydroxyvitamin D, these alterations were not observed in nicotine-exposed females. Nicotine-exposed males only showed lower IRS1 in VAT, while the females had hyperglycemia, higher pAKT in VAT, while lower IRβ, IRS1, and GLUT4 in SAT. Parameters related to metabolism and action of GC and vitamin D were unaltered in both sexes. We evidence that exposure exclusively to nicotine during breastfeeding affects the hormone status and fat depots of the adult progeny in a sex-dependent manner.
Collapse
|
46
|
Huang W, Zhou H, Hodgkinson C, Montero A, Goldman D, Chang SL. Network Meta-Analysis on the Mechanisms Underlying Alcohol Augmentation of COVID-19 Pathologies. Alcohol Clin Exp Res 2021; 45:675-688. [PMID: 33583045 PMCID: PMC8014161 DOI: 10.1111/acer.14573] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic is a worldwide crisis caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Many COVID-19 patients present with fever in the early phase, with some progressing to a hyperinflammatory phase. Ethanol (EtOH) exposure may lead to systemic inflammation. Network meta-analysis was conducted to examine possible relationships between EtOH consumption and COVID-19 pathologies. METHODS Molecules affected by EtOH exposure were identified by analysis with QIAGEN Knowledge Base. Molecules affected by COVID-19 were identified from studies in MEDLINE, bioRxiv, and medRxiv reporting gene expression profiles in COVID-19 patients, QIAGEN Coronavirus Network Explorer, and analysis of the RNA-sequencing data of autopsied lungs of COVID-19 patients retrieved from the GEO database. Network meta-analysis was then conducted on these molecules using QIAGEN Ingenuity Pathway Analysis (IPA). RESULTS Twenty-eight studies reporting significant gene expression changes in COVID-19 patients were identified. One RNA-sequencing dataset on autopsied lungs of COVID-19 patients was retrieved from GEO. Our network meta-analysis suggests that EtOH exposure may augment the effects of SARS-CoV-2 infection on hepatic fibrosis signaling pathway, cellular metabolism and homeostasis, inflammation, and neuroinflammation. EtOH may also enhance the activity of key mediators including cytokines, such as IL-1β, IL-6, and TNF, and transcription factors, such as JUN and STAT, while inhibiting the activity of anti-inflammatory mediators including glucocorticoid receptor. Furthermore, IL-1β, IL-6, TNF, JUN, and STAT were mapped to 10 pathways predicted to associate with SARS-CoV-2 proteins, including HMGB1, IL-1, and IL-6 signaling pathways. CONCLUSIONS Our meta-analyses demonstrate that EtOH exposure may augment SARS-CoV-2-induced inflammation by altering the activity of key inflammatory mediators. Our findings suggest that it is important for clinicians to caution patients about the risk of alcohol consumption, which has increased during the COVID-19 pandemic. The findings also call for further investigation into how alcohol exposure affects viral infections.
Collapse
Affiliation(s)
- Wenfei Huang
- Institute of Neuroimmune PharmacologySeton Hall UniversitySouth OrangeNJUSA
- Department of Biological SciencesSeton Hall UniversitySouth OrangeNJUSA
| | - Heping Zhou
- Department of Biological SciencesSeton Hall UniversitySouth OrangeNJUSA
| | | | - Angelo Montero
- Institute of Neuroimmune PharmacologySeton Hall UniversitySouth OrangeNJUSA
- Department of Biological SciencesSeton Hall UniversitySouth OrangeNJUSA
| | | | - Sulie L. Chang
- Institute of Neuroimmune PharmacologySeton Hall UniversitySouth OrangeNJUSA
- Department of Biological SciencesSeton Hall UniversitySouth OrangeNJUSA
| |
Collapse
|
47
|
de Medeiros SF, Rodgers RJ, Norman RJ. Adipocyte and steroidogenic cell cross-talk in polycystic ovary syndrome. Hum Reprod Update 2021; 27:771-796. [PMID: 33764457 DOI: 10.1093/humupd/dmab004] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Metabolic and endocrine alterations in women with polycystic ovary syndrome (PCOS) affect adipose tissue mass and distribution. PCOS is characterised by hyperandrogenism, obesity and adipocyte dysfunction. Hyperandrogenism in PCOS drives dysfunctional adipocyte secretion of potentially harmful adipocytokines. Glucocorticoids and sex-steroids modulate adipocyte development and function. For their part, adipocyte products interact with adrenal and ovarian steroidogenic cells. Currently, the relationship between adipocyte and steroidogenic cells is not clear, and for these reasons, it is important to elucidate the interrelationship between these cells in women with and without PCOS. OBJECTIVE AND RATIONALE This comprehensive review aims to assess current knowledge regarding the interrelationship between adipocytes and adrenal and ovarian steroidogenic cells in animal models and humans with or without PCOS. SEARCH METHODS We searched for articles published in English and Portuguese in PubMed. Keywords were as follows: polycystic ovary syndrome, steroidogenesis, adrenal glands, theca cells, granulosa cells, adipocytes, adipocytokines, obesity, enzyme activation, and cytochrome P450 enzymes. We expanded the search into the references from the retrieved articles. OUTCOMES Glucocorticoids and sex-steroids modulate adipocyte differentiation and function. Dysfunctional adipocyte products play important roles in the metabolic and endocrine pathways in animals and women with PCOS. Most adipokines participate in the regulation of the hypothalamic-pituitary-adrenal and ovarian axes. In animal models of PCOS, hyperinsulinemia and poor fertility are common; various adipokines modulate ovarian steroidogenesis, depending on the species. Women with PCOS secrete unbalanced levels of adipocyte products, characterised by higher levels of leptin and lower levels of adiponectin. Leptin expression positively correlates with body mass index, waist/hip ratio and levels of total cholesterol, triglyceride, luteinising hormone, oestradiol and androgens. Leptin inhibits the production of oestradiol and, in granulosa cells, may modulate 17-hydroxylase and aromatase enzyme activities. Adiponectin levels negatively correlate with fat mass, body mass index, waist-hip ratio, glucose, insulin and triglycerides, and decrease androgen production by altering expression of luteinising hormone receptor, steroidogenic acute regulatory protein, cholesterol-side-chain cleavage enzyme and 17-hydroxylase. Resistin expression positively correlates with body mass index and testosterone, and promotes the expression of 17-hydroxylase enzyme in theca cells. The potential benefits of adipokines in the treatment of women with PCOS require more investigation. WIDER IMPLICATIONS The current data regarding the relationship between adipocyte products and steroidogenic cells are conflicting in animals and humans. Polycystic ovary syndrome is an excellent model to investigate the interrelationship among adipocyte and steroidogenic cells. Women with PCOS manifest some pathological conditions associated with hyperandrogenism and adipocyte products. In animals, cross-talk between cells may vary according to species, and the current review suggests opportunities to test new medications to prevent or even reverse several harmful sequelae of PCOS in humans. Further studies are required to investigate the possible therapeutic application of adipokines in women with obese and non-obese PCOS. Meanwhile, when appropriate, metformin use alone, or associated with flutamide, may be considered for therapeutic purposes.
Collapse
Affiliation(s)
- Sebastião Freitas de Medeiros
- Department of Gynecology and Obstetrics, Medical School, Federal University of Mato Grosso; and Tropical Institute of Reproductive Medicine,Cuiabá, MT, Brazil
| | - Raymond Joseph Rodgers
- Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Robert John Norman
- Robinson Research Institute Adelaide Medical School, Adelaide, South Australia, Australia
| |
Collapse
|
48
|
Lu Y, Wang E, Chen Y, Zhou B, Zhao J, Xiang L, Qian Y, Jiang J, Zhao L, Xiong X, Lu Z, Wu D, Liu B, Yan J, Zhang R, Zhang H, Hu C, Li X. Obesity-induced excess of 17-hydroxyprogesterone promotes hyperglycemia through activation of glucocorticoid receptor. J Clin Invest 2021; 130:3791-3804. [PMID: 32510471 DOI: 10.1172/jci134485] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/08/2020] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) has become an expanding global public health problem. Although the glucocorticoid receptor (GR) is an important regulator of glucose metabolism, the relationship between circulating glucocorticoids (GCs) and the features of T2DM remains controversial. Here, we show that 17-hydroxyprogesterone (17-OHP), an intermediate steroid in the biosynthetic pathway that converts cholesterol to cortisol, binds to and stimulates the transcriptional activity of GR. Hepatic 17-OHP concentrations are increased in diabetic mice and patients due to aberrantly increased expression of Cyp17A1. Systemic administration of 17-OHP or overexpression of Cyp17A1 in the livers of lean mice promoted the pathogenesis of hyperglycemia and insulin resistance, whereas knockdown of Cyp17A1 abrogated metabolic disorders in obese mice. Therefore, our results identify a Cyp17A1/17-OHP/GR-dependent pathway in the liver that mediates obesity-induced hyperglycemia, suggesting that selectively targeting hepatic Cyp17A1 may provide a therapeutic avenue for treating T2DM.
Collapse
Affiliation(s)
- Yan Lu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - E Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Ying Chen
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Bing Zhou
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Jiejie Zhao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Liping Xiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Yiling Qian
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Jingjing Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Lin Zhao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Xuelian Xiong
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Zhiqiang Lu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| | - Duojiao Wu
- Institute of Clinical Science, Shanghai Institute of Clinical Bioinformatics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Liu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and.,Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Jing Yan
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, and
| | - Rong Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, and.,Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, and.,Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai, China.,Institute for Metabolic Disease, Fengxian Central Hospital, Southern Medical University, Shanghai, China
| | - Xiaoying Li
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Endocrinology and Metabolism, and
| |
Collapse
|
49
|
Creeden JF, Gordon DM, Stec DE, Hinds TD. Bilirubin as a metabolic hormone: the physiological relevance of low levels. Am J Physiol Endocrinol Metab 2021; 320:E191-E207. [PMID: 33284088 PMCID: PMC8260361 DOI: 10.1152/ajpendo.00405.2020] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recent research on bilirubin, a historically well-known waste product of heme catabolism, suggests an entirely new function as a metabolic hormone that drives gene transcription by nuclear receptors. Studies are now revealing that low plasma bilirubin levels, defined as "hypobilirubinemia," are a possible new pathology analogous to the other end of the spectrum of extreme hyperbilirubinemia seen in patients with jaundice and liver dysfunction. Hypobilirubinemia is most commonly seen in patients with metabolic dysfunction, which may lead to cardiovascular complications and possibly stroke. We address the clinical significance of low bilirubin levels. A better understanding of bilirubin's hormonal function may explain why hypobilirubinemia might be deleterious. We present mechanisms by which bilirubin may be protective at mildly elevated levels and research directions that could generate treatment possibilities for patients with hypobilirubinemia, such as targeting of pathways that regulate its production or turnover or the newly designed bilirubin nanoparticles. Our review here calls for a shift in the perspective of an old molecule that could benefit millions of patients with hypobilirubinemia.
Collapse
Affiliation(s)
- Justin F Creeden
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Darren M Gordon
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - David E Stec
- Department of Physiology & Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
50
|
Hinds TD, Creeden JF, Gordon DM, Stec DF, Donald MC, Stec DE. Bilirubin Nanoparticles Reduce Diet-Induced Hepatic Steatosis, Improve Fat Utilization, and Increase Plasma β-Hydroxybutyrate. Front Pharmacol 2020; 11:594574. [PMID: 33390979 PMCID: PMC7775678 DOI: 10.3389/fphar.2020.594574] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/13/2020] [Indexed: 12/18/2022] Open
Abstract
The inverse relationship of plasma bilirubin levels with liver fat accumulation has prompted the possibility of bilirubin as a therapeutic for non-alcoholic fatty liver disease. Here, we used diet-induced obese mice with non-alcoholic fatty liver disease treated with pegylated bilirubin (bilirubin nanoparticles) or vehicle control to determine the impact on hepatic lipid accumulation. The bilirubin nanoparticles significantly reduced hepatic fat, triglyceride accumulation, de novo lipogenesis, and serum levels of liver dysfunction marker aspartate transaminase and ApoB100 containing very-low-density lipoprotein. The bilirubin nanoparticles improved liver function and activated the hepatic β-oxidation pathway by increasing PPARα and acyl-coenzyme A oxidase 1. The bilirubin nanoparticles also significantly elevated plasma levels of the ketone β-hydroxybutyrate and lowered liver fat accumulation. This study demonstrates that bilirubin nanoparticles induce hepatic fat utilization, raise plasma ketones, and reduce hepatic steatosis, opening new therapeutic avenues for NAFLD.
Collapse
Affiliation(s)
- Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Justin F Creeden
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH, United States
| | - Darren M Gordon
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH, United States
| | - Donald F Stec
- Small Molecule NMR Facility Core, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States
| | - Matthew C Donald
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|