1
|
Marei WFA, Moorkens K, Gansemans Y, Van Nieuwerburgh F, Leroy JLMR. Acute and long-term transcriptomic responses of granulosa cells to obesogenic diet and concomitant effects on oocyte quality: insight from an outbred mouse model†. Biol Reprod 2025; 112:692-708. [PMID: 39913328 DOI: 10.1093/biolre/ioaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/08/2024] [Accepted: 02/04/2025] [Indexed: 04/16/2025] Open
Abstract
Diet-induced obesity can cause long-term alterations in ovarian functions, but the acute effects of obesogenic diets on the follicular cells and their progression over time, when intake is continued and obesity develops, remain unclear. We aimed to determine the onset and progression of changes in the granulosa cell transcriptomic profile after starting a high-fat/high sugar (HFHS)-diet feeding in mice. We also examined the changes in oocyte lipid droplet content and mitochondrial ultrastructural abnormalities. Swiss (outbred) mice were sacrificed at 24 h, 3 days, and at 1, 4, 8, 12, and 16 weeks of feeding HFHS and control diets. Lipid droplet content significantly increased in the HFHS oocytes within 24 h compared to controls (P < 0.05). Oocyte mitochondrial abnormalities only increased starting from 8 weeks. Granulosa RNA-seq revealed altered transcriptomic gene-set enrichments (GO terms and KEGG pathways, Padj < 0.05) already at 3 days and 1 week indicating acute endoplasmic reticulum unfolded protein responses, with concomitant fluctuations in several cellular metabolic pathways and gene sets related to mitochondrial bioenergetic functions, some of which persisted after 8 weeks. Interestingly, the short- and long-term patterns of changes in cytochrome P450, steroid hormone biosynthesis, retinol metabolism, bile acid metabolism, fatty acid metabolism, and Pi3K/Akt signaling pathways were most prominent and highly correlated; all being acutely upregulated, then chronically downregulated. These results show that the impact of obesogenic diet on the oocyte and granulosa cells is prompt, while the response depends on the duration of feeding and occurs in a multiphasic cascade together with a progressive deterioration in oocyte quality.
Collapse
Affiliation(s)
- Waleed F A Marei
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Kerlijne Moorkens
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Yannick Gansemans
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | | | - Jo L M R Leroy
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
2
|
Wang Y, Zhang Y, Leung VH, Seradj SH, Sonmez U, Servin-Vences MR, Xiao S, Ren X, Wang L, Mishkanian SA, Kini SA, Long JZ, Lipomi DJ, Ye L, Patapoutian A. A key role of PIEZO2 mechanosensitive ion channel in adipose sensory innervation. Cell Metab 2025; 37:1001-1011.e7. [PMID: 40054462 DOI: 10.1016/j.cmet.2025.02.004] [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: 03/29/2024] [Revised: 12/14/2024] [Accepted: 02/06/2025] [Indexed: 03/12/2025]
Abstract
Compared with the well-established functions of sympathetic innervation, the role of sensory afferents in adipose tissues remains less understood. Recent work has revealed the anatomical and physiological significance of adipose sensory innervation; however, its molecular underpinning remains unclear. Here, using organ-targeted single-cell RNA sequencing, we identified the mechanoreceptor PIEZO2 as one of the most prevalent receptors in fat-innervating dorsal root ganglia (DRG) neurons. PIEZO2 deletion in fat-innervating neurons induced transcriptional programs in adipose tissue resembling sympathetic activation, mirroring DRG ablation. Conversely, a gain-of-function PIEZO2 mutant shifted the adipose phenotypes in the opposite direction. These results indicate that PIEZO2 plays a major role in the sensory regulation of adipose tissues. This discovery opens new avenues for exploring mechanosensation in organs not traditionally considered mechanically active, such as adipose tissues, and therefore sheds light on the broader significance of mechanosensation in regulating organ function and homeostasis.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Yunxiao Zhang
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Verina H Leung
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Saba Heydari Seradj
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Utku Sonmez
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
| | - M Rocio Servin-Vences
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Shuke Xiao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Xiangyu Ren
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Leon Wang
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Sassan A Mishkanian
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Sejal A Kini
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA; The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Darren J Lipomi
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Li Ye
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Ardem Patapoutian
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
3
|
Zeng X, Wang Y, Farias K, Rappa A, Darko C, Sauve A, Huang Q, Alonso LC, Yang Y. NRH, a potent NAD + enhancer, improves glucose homeostasis and lipid metabolism in diet-induced obese mice through an active adenosine kinase pathway. Metabolism 2025; 164:156110. [PMID: 39710001 PMCID: PMC11788054 DOI: 10.1016/j.metabol.2024.156110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
AIMS NAD+ deficiency underlies obesity-induced metabolic disturbances. This study evaluated dihydronicotinamide riboside (NRH), a potent NAD+ enhancer, in lean and obese mice and explored whether NRH operates through a unique mechanism involving adenosine kinase (ADK), an enzyme critical for NRH-driven NAD+ synthesis. METHODS Pharmacokinetic and pharmacodynamic analyses were performed following a single 250 mg/kg intraperitoneal injection of NRH in healthy mice. In long-term studies, lean and high-fat diet-induced obese mice were treated with 250 mg/kg NRH thrice weekly for 7 weeks. Blood NAD+ levels, body composition, energy expenditure, and glucose and lipid metabolism were monitored. To test ADK's role, the ADK inhibitor ABT702 was co-administered with NRH in obese mice. RESULTS NRH entered tissues unassisted and was rapidly metabolized for NAD+ biosynthesis, while ADK inhibition blocked its phosphorylation, leading to NRH accumulation in all examined tissues and possible release back into circulation. The 7-week NRH administration was well-tolerated in both lean and obese mice. In obese mice, NRH improved glucose homeostasis by boosting insulin secretion, enhancing muscle insulin signaling, and reducing hepatic gluconeogenesis. It also lowered fat mass, decreased serum lipids, and improved white adipose function. These benefits were linked to elevated tissue NAD+ levels, enhanced Sirtuin activities, and increased mitochondrial antioxidant defenses. ADK inhibition abolished these effects, confirming that NRH's direct entry into tissues and subsequent phosphorylation is essential for its full benefits. CONCLUSION This study establishes NRH as a promising therapeutic agent for obesity-induced metabolic dysfunction, correcting glucose intolerance and hyperlipidemia through ADK-dependent NAD+ enhancement.
Collapse
Affiliation(s)
- Xinliu Zeng
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongjie Wang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Department of Animal Sciences, North Carolina A&T State University, Greensboro, NC 27411, United States of America
| | - Karina Farias
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Andrew Rappa
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Christine Darko
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Anthony Sauve
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Qingxia Huang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America; Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Laura C Alonso
- Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY 10021, United States of America
| | - Yue Yang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, United States of America
| |
Collapse
|
4
|
Gutierrez MW, van Tilburg Bernardes E, Ren E, Kalbfleisch KN, Day M, Lameu EL, Glatthardt T, Mercer EM, Sharma S, Zhang H, Al-Azawy A, Chleilat F, Hirota SA, Reimer RA, Arrieta MC. Early-life gut mycobiome core species modulate metabolic health in mice. Nat Commun 2025; 16:1467. [PMID: 39922818 PMCID: PMC11807121 DOI: 10.1038/s41467-025-56743-8] [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: 02/12/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
The gut microbiome causally contributes to obesity; however, the role of fungi remains understudied. We previously identified three core species of the infant gut mycobiome (Rhodotorula mucilaginosa, Malassezia restricta and Candida albicans) that correlated with body mass index, however their causal contributions to obesity development are unknown. Here we show the effects of early-life colonization by these fungal species on metabolic health in gnotobiotic mice fed standard (SD) or high-fat-high-sucrose (HFHS) diets. Each species resulted in bacterial microbiome compositional and functional differences. R. mucilaginosa and M. restricta increased adiposity in mice fed SD, while only R. mucilaginosa exacerbated metabolic disease. In contrast, C. albicans resulted in leanness and resistance to diet-induced obesity. Intestinal nutrient transporter expression was unaffected by the presence of fungi in jejunal enteroids, yet the immune landscape in white adipose tissue was distinctly impacted by each fungal species, suggesting that these phenotypes may be a result of fungal immune regulation. This work revealed that three common fungal colonizers have distinct causal influences on obesity and metabolic inflammation and justifies the consideration of fungi in microbiome research on host metabolism.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Erik van Tilburg Bernardes
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ellen Ren
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Kristen N Kalbfleisch
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Madeline Day
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ewandson Luiz Lameu
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Thaís Glatthardt
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Emily M Mercer
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Sunita Sharma
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Hong Zhang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ali Al-Azawy
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Faye Chleilat
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Simon A Hirota
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Raylene A Reimer
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada.
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada.
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
5
|
Ahn M, Dhawan S, McCown EM, Garcia PA, Bhattacharya S, Stein R, Thurmond DC. Beta cell-specific PAK1 enrichment ameliorates diet-induced glucose intolerance in mice by promoting insulin biogenesis and minimising beta cell apoptosis. Diabetologia 2025; 68:152-165. [PMID: 39404845 DOI: 10.1007/s00125-024-06286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/16/2024] [Indexed: 12/22/2024]
Abstract
AIMS/HYPOTHESIS p21 (CDC42/RAC1) activated kinase 1 (PAK1) is depleted in type 2 diabetic human islets compared with non-diabetic human islets, and acute PAK1 restoration in the islets can restore insulin secretory function ex vivo. We hypothesised that beta cell-specific PAK1 enrichment in vivo can mitigate high-fat-diet (HFD)-induced glucose intolerance by increasing the functional beta cell mass. METHODS Human islets expressing exogenous PAK1 specifically in beta cells were used for bulk RNA-seq. Human EndoC-βH1 cells overexpressing myc-tagged PAK1 were used for chromatin immunoprecipitation (ChIP) and ChIP-sequencing (ChIP-seq). Novel doxycycline-inducible beta cell-specific PAK1-expressing (iβPAK1-Tg) mice were fed a 45% HFD pre-induction for 3 weeks and for a further 3 weeks with or without doxycycline induction. These HFD-fed mice were evaluated for GTT, ITT, 6 h fasting plasma insulin and blood glucose, body composition, islet insulin content and apoptosis. RESULTS Beta cell-specific PAK1 enrichment in type 2 diabetes human islets resulted in decreased beta cell apoptosis and increased insulin content. RNA-seq showed an upregulation of INS gene transcription by PAK1. Using clonal human beta cells, we found that PAK1 protein was localised in the cytoplasm and the nucleus. ChIP studies revealed that nuclear PAK1 enhanced pancreatic and duodenal homeobox1 (PDX1) and neuronal differentiation 1 (NEUROD1) binding to the INS promoter in a glucose-responsive manner. Importantly, the iβPAK1-Tg mice, when challenged with HFD and doxycycline induction displayed enhanced glucose tolerance, increased islet insulin content and reduced beta cell apoptosis when compared with iβPAK1-Tg mice without doxycycline induction. CONCLUSIONS/INTERPRETATION PAK1 plays an unforeseen and beneficial role in beta cells by promoting insulin biogenesis via enhancing the expression of PDX1, NEUROD1 and INS, along with anti-apoptotic effects, that culminate in increased insulin content and beta cell mass in vivo and ameliorate diet-induced glucose intolerance. DATA AVAILABILITY The raw and processed RNA-seq data and ChIP-seq data, which has been made publicly available at Gene Expression Omnibus (GEO) at https://www.ncbi.nlm.nih.gov/geo/ , can be accessed in GSE239382.
Collapse
Affiliation(s)
- Miwon Ahn
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Erika M McCown
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Pablo A Garcia
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | | | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Debbie C Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
6
|
Keles U, Kalem-Yapar NE, Hultén H, Zhao LN, Kaldis P. Impact of Short-Term Lipid Overload on Whole-Body Physiology. Mol Cell Biol 2024; 45:47-58. [PMID: 39726368 DOI: 10.1080/10985549.2024.2438814] [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: 05/31/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
Complex metabolic diseases due to overnutrition such as obesity, type 2 diabetes, and fatty liver disease are a major burden on the healthcare system worldwide. Current research primarily focuses on disease endpoints and trying to understand underlying mechanisms at relatively late stages of the diseases, when irreversible damage is already done. However, complex interactions between physiological systems during disease development create a problem regarding how to build cause-and-effect relationships. Therefore, it is essential to understand the early pathophysiological effects of overnutrition, which can help us understand the origin of the disease and to design better treatment strategies. Here, we focus on early metabolic events in response to high-fat diets (HFD) in rodents. Interestingly, insulin resistance, fatty liver, and obesity-promoting systemic inflammatory responses are evident within a week when mice are given consecutive HFD meals. However, as shown in human studies, these effects are usually not visible after a single meal. Overall, these results suggest that sustained HFD-intake within days can create a hyperlipidemic environment, globally remodeling metabolism in all affected organs and resembling some of the important disease features.
Collapse
Affiliation(s)
- Umur Keles
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| | - Nisan Ece Kalem-Yapar
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| | - Hanna Hultén
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| | - Li Na Zhao
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| |
Collapse
|
7
|
Niv D, Anavi E, Yaval L, Abbas A, Rytwo G, Gutman R. Sepiolite-Chitosan-Acetic Acid Biocomposite Attenuates the Development of Obesity and Nonalcoholic Fatty Liver Disease in Mice Fed a High-Fat Diet. Nutrients 2024; 16:3958. [PMID: 39599744 PMCID: PMC11597185 DOI: 10.3390/nu16223958] [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: 10/10/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Background; obesity and nonalcoholic fatty liver disease (NAFLD) reduce life expectancy; nonoperative interventions show poor results. Individually, chitosan (1% w/w), acetic acid (AA 0.3-6.5% w/w), and sepiolite clay (5% w/w) attenuate high-fat-diet-induced obesity (DIO) via reduced energy digestibility and increased energy expenditure. Objectives; therefore, we hypothesized that a chitosan-sepiolite biocomposite suspended in AA would attenuate DIO and NAFLD to a greater extent than AA alone via its more substantial adsorption of nonpolar molecules. Methods; we tested this dietary supplement in C57BL/6J mice fed a high-fat diet (HFD) compared to an unsupplemented HFD and an HFD supplemented with a bile acid sequestrant (cholestyramine) or standalone AA. Results; biocomposite supplementation reduced DIO gain by 60% and abolished hepatic liver accumulation, whereas standalone AA showed mild attenuation of DIO gain and did not prevent HFD-induced hepatic fat accumulation. The biocomposite intake was accompanied by a lower digestibility (-4 point %) counterbalanced by increased intake; hence, it did not affect energy absorption. Therefore, DIO attenuation was suggested to be related to higher energy expenditure, a phenomenon not found with AA alone, as supported by calculated energy expenditure using the energy balance method. Conclusions; these results support further investigation of the biocomposite's efficacy in attenuating obesity and NAFLD, specifically when applied with a restricted diet. Future studies are needed to determine this biocomposite's safety, mechanism of action, and efficacy compared to its components given separately or combined with other ingredients.
Collapse
Affiliation(s)
- Dalia Niv
- Laboratory of Integrative Physiology, The Department of Nutrition and Natural Products, MIGAL—Galilee Research Institute, P.O. Box 831, Kiryat Shmona 11016, Israel
| | - Eli Anavi
- Laboratory of Integrative Physiology, The Department of Nutrition and Natural Products, MIGAL—Galilee Research Institute, P.O. Box 831, Kiryat Shmona 11016, Israel
| | - Laris Yaval
- Laboratory of Integrative Physiology, The Department of Nutrition and Natural Products, MIGAL—Galilee Research Institute, P.O. Box 831, Kiryat Shmona 11016, Israel
| | - Atallah Abbas
- Laboratory of Integrative Physiology, The Department of Nutrition and Natural Products, MIGAL—Galilee Research Institute, P.O. Box 831, Kiryat Shmona 11016, Israel
| | - Giora Rytwo
- Environmental Physical Chemistry Laboratory, MIGAL—Galilee Research Institute, P.O. Box 831, Kiryat Shmona 11016, Israel;
- Departments of Environmental and Water Sciences, Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee 12210, Israel
| | - Roee Gutman
- Laboratory of Integrative Physiology, The Department of Nutrition and Natural Products, MIGAL—Galilee Research Institute, P.O. Box 831, Kiryat Shmona 11016, Israel
- Department of Animal Sciences, Faculty of Sciences and Technology, Tel-Hai College, Upper Galilee 12210, Israel
| |
Collapse
|
8
|
Wang Y, Zhang Y, Leung V, Seradj SH, Sonmez U, Servin-Vences R, Lipomi D, Ye L, Patapoutian A. A key role of PIEZO2 mechanosensitive ion channel in adipose sensory innervation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624210. [PMID: 39605632 PMCID: PMC11601537 DOI: 10.1101/2024.11.18.624210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Compared to the well-established functions of sympathetic innervation, the role of sensory afferents in adipose tissues remains less understood. Recent work revealed the anatomical and physiological significance of adipose sensory innervation; however, its molecular underpinning remains unclear. Here, using organ-targeted single-cell RNA sequencing, we identified the mechanoreceptor PIEZO2 as one of the most prevalent receptors in fat-innervating dorsal root ganglia (DRG) neurons. We found that selective PIEZO2 deletion in fat-innervating neurons phenocopied the molecular alternations in adipose tissue caused by DRG ablation. Conversely, a gain-of-function PIEZO2 mutant shifted the adipose phenotypes in the opposite direction. These results indicate that PIEZO2 plays a major role in the sensory regulation of adipose tissues. This discovery opens new avenues for exploring mechanosensation in organs not traditionally considered mechanically active, such as the adipose tissues, and therefore sheds light on the broader significance of mechanosensation in regulating organ function and homeostasis.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Yunxiao Zhang
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Verina Leung
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, United States
| | - Saba Heydari Seradj
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, United States
| | - Utku Sonmez
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
- Jacobs School of Engineering, UCSD, San Diego, United States
| | - Rocio Servin-Vences
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Darren Lipomi
- Jacobs School of Engineering, UCSD, San Diego, United States
| | - Li Ye
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, United States
| | - Ardem Patapoutian
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, San Diego, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
- Lead contact
| |
Collapse
|
9
|
Budd JM, Notaro NM, MacLeod B, Mutch DM, Dyck DJ. A ketogenic diet, regardless of fish oil content, does not affect glucose homeostasis or muscle insulin response in male rats. Am J Physiol Endocrinol Metab 2024; 327:E449-E458. [PMID: 39140973 DOI: 10.1152/ajpendo.00236.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Ketogenic diets (KDs) are very high in fat and low in carbohydrates. Evidence supports that KDs improve glucose metabolism in humans and rodents that are obese and/or insulin resistant. Conversely, findings in healthy rodents suggest that KDs may impair glucose homeostasis. In addition, most experimental KDs are composed of saturated and monounsaturated fatty acids, with almost no omega-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA). Evidence supports a beneficial role for n-3 LCPUFA on glucose homeostasis in the context of a metabolic challenge. To our knowledge, no study has examined whether the inclusion of n-3 LCPUFA affects the impact of a KD on glucose homeostasis. The objective of this study was to examine the impact of a KD on whole body glucose tolerance and skeletal muscle insulin response in rats and to determine if increasing the n-3 LCPUFA content in a KD with menhaden oil could improve metabolic outcomes. Male Sprague-Dawley rats were pair-fed one of a low-fat diet, high-fat diet, KD, or a KD supplemented with menhaden oil for 8 wk. No significant differences in whole body glucose tolerance, skeletal muscle insulin signaling, or skeletal muscle insulin-stimulated glucose uptake were detected between the dietary groups. Our findings suggest that KD feeding, with or without supplementation of n-3 LCPUFA, does not affect whole body glucose homeostasis or skeletal muscle insulin response under pair-feeding conditions.NEW & NOTEWORTHY Ketogenic diets (KDs) improve glucose metabolism in humans and rodents that are insulin resistant, but their impact is unclear in a healthy context. Furthermore, standard KDs typically lack beneficial omega-3 long-chain polyunsaturated fatty acids (n3-LCPUFA). This study assessed whether supplementing a KD with n3-LCPUFA could alter glucose homeostasis or skeletal muscle insulin response. No differences were observed between a standard KD and a KD with n3-LCPUFA when energy intake was controlled.
Collapse
Affiliation(s)
- Joshua M Budd
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Nicole M Notaro
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Blair MacLeod
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David M Mutch
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David J Dyck
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
10
|
Palatini Jackson KM, Mhawish R, Komarnytsky S. Bitter Phytochemicals Acutely Lower Blood Glucose Levels by Inhibition of Glucose Absorption in the Gut. ENDOCRINES 2024; 5:304-322. [DOI: 10.3390/endocrines5030022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
For early hominids, frequent encounters with plant foods necessitated the ability to discern bitter poisons and adjust the activity of the gastrointestinal system in anticipation of carbohydrate-rich meals. Plants bitters were also used historically to manage a variety of metabolic and digestive disorders despite an immense structural diversity of bitter phytochemicals without a common molecular target. Our study confirms these observations in a standardized C57BL/6J prediabetic mouse model using 24 model compounds by demonstrating acute lower peak blood glucose values and improved glucose tolerance following intragastric, but not intraperitoneal, treatment. The administration of the synthetic bitter compound denatonium benzoate yielded similar results that were attenuated by co-application of the allosteric inhibitor of the bitter TAS2R receptors. We also show that these effects occur dose-dependently; associate with reduced glucose uptake, increased intracellular [Ca2+] fluxes, and enhanced GLP-1 expression; and are attenuated by the TAS2R inhibitor in the neuroendocrine STC-1 intestinal cells. These findings support the view that inhibition of glucose transport from the intestinal lumen to the blood by TAS2R bitter receptor signaling in the gut may represent a common mechanism in the acute response to oral ingestion of bitter phytochemicals.
Collapse
Affiliation(s)
| | - Reham Mhawish
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA
| | - Slavko Komarnytsky
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA
| |
Collapse
|
11
|
Harvei S, Skogen V, Egelandsdal B, Birkeland S, Paulsen JE, Carlsen H. Chronic oral LPS administration does not increase inflammation or induce metabolic dysregulation in mice fed a western-style diet. Front Nutr 2024; 11:1376493. [PMID: 39077160 PMCID: PMC11284168 DOI: 10.3389/fnut.2024.1376493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/25/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Lipopolysaccharides (LPS) present in the intestine are suggested to enter the bloodstream after consumption of high-fat diets and cause systemic inflammation and metabolic dysregulation through a process named "metabolic endotoxemia." This study aimed to determine the role of orally administered LPS to mice in the early stage of chronic low-grade inflammation induced by diet. Methods We supplemented the drinking water with E. coli derived LPS to mice fed either high-fat Western-style diet (WSD) or standard chow (SC) for 7 weeks (n = 16-17). Body weight was recorded weekly. Systemic inflammatory status was assessed by in vivo imaging of NF-κB activity at different time points, and glucose dysregulation was assessed by insulin sensitivity test and glucose tolerance test near the end of the study. Systemic LPS exposure was estimated indirectly via quantification of LPS-binding protein (LBP) and antibodies against LPS in plasma, and directly using an LPS-sensitive cell reporter assay. Results and discussion Our results demonstrate that weight development and glucose regulation are not affected by LPS. We observed a transient LPS dependent upregulation of NF-κB activity in the liver region in both diet groups, a response that disappeared within the first week of LPS administration and remained low during the rest of the study. However, WSD fed mice had overall a higher NF-κB activity compared to SC fed mice at all time points independent of LPS administration. Our findings indicate that orally administered LPS has limited to no impact on systemic inflammation and metabolic dysregulation in mice fed a high-fat western diet and we question the capability of intestinally derived LPS to initiate systemic inflammation through a healthy and uncompromised intestine, even when exposed to a high-fat diet.
Collapse
Affiliation(s)
- Silje Harvei
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Vemund Skogen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Bjørg Egelandsdal
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Signe Birkeland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| | - Jan Erik Paulsen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, As, Norway
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, As, Norway
| |
Collapse
|
12
|
Honce R, Vazquez-Pagan A, Livingston B, Mandarano AH, Wilander BA, Cherry S, Hargest V, Sharp B, Brigleb PH, Kirkpatrick Roubidoux E, Van de Velde LA, Skinner RC, McGargill MA, Thomas PG, Schultz-Cherry S. Diet switch pre-vaccination improves immune response and metabolic status in formerly obese mice. Nat Microbiol 2024; 9:1593-1606. [PMID: 38637722 DOI: 10.1038/s41564-024-01677-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 03/20/2024] [Indexed: 04/20/2024]
Abstract
Metabolic disease is epidemiologically linked to severe complications upon influenza virus infection, thus vaccination is a priority in this high-risk population. Yet, vaccine responses are less effective in these same hosts. Here we examined how the timing of diet switching from a high-fat diet to a control diet affected influenza vaccine efficacy in diet-induced obese mice. Our results demonstrate that the systemic meta-inflammation generated by high-fat diet exposure limited T cell maturation to the memory compartment at the time of vaccination, impacting the recall of effector memory T cells upon viral challenge. This was not improved with a diet switch post-vaccination. However, the metabolic dysfunction of T cells was reversed if weight loss occurred 4 weeks before vaccination, restoring a functional recall response. This corresponded with changes in the systemic obesity-related biomarkers leptin and adiponectin, highlighting the systemic and specific effects of diet on influenza vaccine immunogenicity.
Collapse
Affiliation(s)
- Rebekah Honce
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
- Vermont Lung Center, Division of Pulmonology and Critical Care, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Ana Vazquez-Pagan
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
- Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA
- Weill Cornell Medicine, New York City, NY, USA
- Noguchi Medical Research Institute (NMRI), Accra, Ghana
| | - Brandi Livingston
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Benjamin A Wilander
- Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Sean Cherry
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Virginia Hargest
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Bridgett Sharp
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Pamela H Brigleb
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Lee-Ann Van de Velde
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - R Chris Skinner
- Division of Natural Sciences and Mathematics, University of the Ozarks, Clarksville, AR, USA
- Department of Nutrition and Food Sciences, College of Agriculture and Life Sciences, University of Vermont, Burlington, VT, USA
| | - Maureen A McGargill
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Stacey Schultz-Cherry
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
13
|
Ravaut G, Carneiro A, Mounier C. Exploring the impacts of ketogenic diet on reversible hepatic steatosis: initial analysis in male mice. Front Nutr 2024; 11:1290540. [PMID: 38577162 PMCID: PMC10991688 DOI: 10.3389/fnut.2024.1290540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most common chronic liver disease. Ketogenic diet (KD), a diet with very low intake in carbohydrates, gained popularity as a weight-loss approach. However, in mice models, it has been reported that an excess exposition of dietary fat induces hepatic insulin resistance and steatosis. However, data published is inconsistent. Herein, we investigated in a mouse model, the metabolic effects of KD and its contribution to the pathogenesis of NALFD. Mice were exposed to KD or CHOW diet for 12 weeks while a third group was exposed to KD for also 12 weeks and then switched to CHOW diet for 4 weeks to determine if we can rescue the phenotype. We evaluated the effects of diet treatments on fat distribution, glucose, and insulin homeostasis as well as hepatic steatosis. Mice fed with KD developed glucose intolerance but not insulin resistance accompanied by an increase of inflammation. KD-fed mice showed an increase of fat accumulation in white adipose tissue and liver. This effect could be explained by an increase in fat uptake by the liver with no changes of catabolism leading to MAFLD. Interestingly, we were able to rescue the phenotype by switching KD-fed mice for 4 weeks on a CHOW diet. Our studies demonstrate that even if mice develop hepatic steatosis and glucose intolerance after 12 weeks of KD, they do not develop insulin resistance and more importantly, the phenotype can be reversed by switching the mice from a KD to a CHOW.
Collapse
Affiliation(s)
| | | | - Catherine Mounier
- CERMO-FC Research Center, Molecular Metabolism of Lipids Laboratory, Biological Sciences Department, University of Quebec in Montreal (UQAM), Montreal, QC, Canada
| |
Collapse
|
14
|
Charlot A, Bringolf A, Debrut L, Mallard J, Charles AL, Crouchet E, Duteil D, Geny B, Zoll J. Changes in Macronutrients during Dieting Lead to Weight Cycling and Metabolic Complications in Mouse Model. Nutrients 2024; 16:646. [PMID: 38474774 DOI: 10.3390/nu16050646] [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: 01/26/2024] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Weight cycling is a major challenge in obesity management. Caloric restriction is known to promote this phenomenon, but the impact of macronutrient changes during dieting remains unclear. This study aimed to determine the role of macronutrient changes in weight maintenance without caloric restriction by alternating between two hypercaloric diets: a high-carbohydrate, high-fat Western diet (WD) and a low-carbohydrate, high-fat diet (LCHDF). Obesity was induced in 8-week-old C57BL/6 male mice by 10 weeks of WD feeding. Then, the mice were subjected to 12 weeks of LCHFD interspersed with WD (I-WD), 3 periods of 2-week LCHFD followed by 2 periods of 3-week WD, or 12 weeks of continuous WD (C-WD). C-WD and I-WD mice were compared to standard diet (SD) mice. In the I-WD group, each LCHFD period decreased weight gain, but mice regained weight after WD resumption. I-WD mice exhibited obesity, dyslipidemia, and glucose intolerance, similarly to the C-WD mice. I-WD mice also developed nonalcoholic steatohepatitis, associated with an increase in type-III collagen gene expression and a decrease in FGF21 protein levels, in comparison with SD. I-WD mice developed weight cycling despite maintaining a high caloric consumption, suggesting that changes in macronutrients during dieting are also a trigger of weight regain.
Collapse
Affiliation(s)
- Anouk Charlot
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072, "Mitochondrie, Stress Oxydant et Plasticité Musculaire", University of Strasbourg, 67000 Strasbourg, France
- Faculty of Sport Sciences, University of Strasbourg, 67000 Strasbourg, France
| | - Anthony Bringolf
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072, "Mitochondrie, Stress Oxydant et Plasticité Musculaire", University of Strasbourg, 67000 Strasbourg, France
| | - Léa Debrut
- CNRS, University of Strasbourg, Inserm, IGBMC UMR 7104-UMR-S 1258, 67400 Illkirch, France
| | - Joris Mallard
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072, "Mitochondrie, Stress Oxydant et Plasticité Musculaire", University of Strasbourg, 67000 Strasbourg, France
- Faculty of Sport Sciences, University of Strasbourg, 67000 Strasbourg, France
- Institute of Cancerology Strasbourg Europe (ICANS), 67200 Strasbourg, France
| | - Anne-Laure Charles
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072, "Mitochondrie, Stress Oxydant et Plasticité Musculaire", University of Strasbourg, 67000 Strasbourg, France
- Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Emilie Crouchet
- Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, University of Strasbourg, Inserm, 67000 Strasbourg, France
| | - Delphine Duteil
- CNRS, University of Strasbourg, Inserm, IGBMC UMR 7104-UMR-S 1258, 67400 Illkirch, France
| | - Bernard Geny
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072, "Mitochondrie, Stress Oxydant et Plasticité Musculaire", University of Strasbourg, 67000 Strasbourg, France
- Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
- Service de Physiologie et Explorations Fonctionnelles, University Hospital of Strasbourg, 67091 Strasbourg, France
| | - Joffrey Zoll
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072, "Mitochondrie, Stress Oxydant et Plasticité Musculaire", University of Strasbourg, 67000 Strasbourg, France
- Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
- Service de Physiologie et Explorations Fonctionnelles, University Hospital of Strasbourg, 67091 Strasbourg, France
| |
Collapse
|
15
|
Milhem F, Hamilton LM, Skates E, Wilson M, Johanningsmeier SD, Komarnytsky S. Biomarkers of Metabolic Adaptation to High Dietary Fats in a Mouse Model of Obesity Resistance. Metabolites 2024; 14:69. [PMID: 38276304 PMCID: PMC10819356 DOI: 10.3390/metabo14010069] [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: 12/14/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Obesity-resistant (non-responder, NR) phenotypes that exhibit reduced susceptibility to developing obesity despite being exposed to high dietary fat are crucial in exploring the metabolic responses that protect against obesity. Although several efforts have been made to study them in mice and humans, the individual protective mechanisms are poorly understood. In this exploratory study, we used a polygenic C57BL/6J mouse model of diet-induced obesity to show that NR mice developed healthier fat/lean body mass ratios (0.43 ± 0.05) versus the obesity-prone (super-responder, SR) phenotypes (0.69 ± 0.07, p < 0.0001) by upregulating gene expression networks that promote the accumulation of type 2a, fast-twitch, oxidative muscle tissues. This was achieved in part by a metabolic adaptation in the form of blood glucose sparing, thus aggravating glucose tolerance. Resistance to obesity in NR mice was associated with 4.9-fold upregulated mitoferrin 1 (Slc25a37), an essential mitochondrial iron importer. SR mice also showed fecal volatile metabolite signatures of enhanced short-chain fatty acid metabolism, including increases in detrimental methyl formate and ethyl propionate, and these effects were reversed in NR mice. Continued research into obesity-resistant phenotypes can offer valuable insights into the underlying mechanisms of obesity and metabolic health, potentially leading to more personalized and effective approaches for managing weight and related health issues.
Collapse
Affiliation(s)
- Fadia Milhem
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (F.M.); (E.S.); (M.W.)
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA;
- Department of Nutrition, University of Petra, 317 Airport Road, Amman 11196, Jordan
| | - Leah M. Hamilton
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA;
- College of Agriculture, Virginia State University, 1 Hayden Drive, Petersburg, VA 23806, USA
| | - Emily Skates
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (F.M.); (E.S.); (M.W.)
| | - Mickey Wilson
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (F.M.); (E.S.); (M.W.)
| | - Suzanne D. Johanningsmeier
- United States Department of Agriculture-Agricultural Research Service, Southeast Area, Food Science and Market Quality & Handling Research Unit, North Carolina State University, 322 Schaub Hall, Box 7624, Raleigh, NC 27695, USA;
| | - Slavko Komarnytsky
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (F.M.); (E.S.); (M.W.)
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA;
| |
Collapse
|
16
|
Aimaretti E, Chimienti G, Rubeo C, Di Lorenzo R, Trisolini L, Dal Bello F, Moradi A, Collino M, Lezza AMS, Aragno M, Pesce V. Different Effects of High-Fat/High-Sucrose and High-Fructose Diets on Advanced Glycation End-Product Accumulation and on Mitochondrial Involvement in Heart and Skeletal Muscle in Mice. Nutrients 2023; 15:4874. [PMID: 38068732 PMCID: PMC10708161 DOI: 10.3390/nu15234874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Diets with an elevated content of fat, sucrose, or fructose are recognized models of diet-induced metabolic alterations, since they induce metabolic derangements, oxidative stress, and chronic low-grade inflammation associated with local and systemic accumulation of advanced glycation end-products (AGEs). This study used four-week-old C57BL/6 male mice, randomly assigned to three experimental dietary regimens: standard diet (SD), high-fat high-sucrose diet (HFHS), or high fructose diet (HFr), administered for 12 weeks. Plasma, heart, and tibialis anterior (TA) skeletal muscle were assayed for markers of metabolic conditions, inflammation, presence of AGEs, and mitochondrial involvement. The HFHS diet induced a tissue-specific differential response featuring (1) a remarkable adaptation of the heart to HFHS-induced heavy oxidative stress, demonstrated by an increased presence of AGEs and reduced mitochondrial biogenesis, and efficaciously counteracted by a conspicuous increase in mitochondrial fission and PRXIII expression; (2) the absence of TA adaptation to HFHS, revealed by a heavy reduction in mitochondrial biogenesis, not counteracted by an increase in fission and PRXIII expression. HFr-induced mild oxidative stress elicited tissue-specific responses, featuring (1) a decrease in mitochondrial biogenesis in the heart, likely counteracted by a tendency for increased fission and (2) a mild reduction in mitochondrial biogenesis in TA, likely counteracted by a tendency for increased fusion, showing the adaptability of both tissues to the diet.
Collapse
Affiliation(s)
- Eleonora Aimaretti
- Unit of Experimental Medicine & Clinical Pathology, Department of Clinical and Biological Science, University of Turin, 10125 Turin, Italy; (E.A.); (C.R.); (A.M.); (M.A.)
| | - Guglielmina Chimienti
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (G.C.); (R.D.L.)
| | - Chiara Rubeo
- Unit of Experimental Medicine & Clinical Pathology, Department of Clinical and Biological Science, University of Turin, 10125 Turin, Italy; (E.A.); (C.R.); (A.M.); (M.A.)
| | - Rosa Di Lorenzo
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (G.C.); (R.D.L.)
| | - Lucia Trisolini
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, CNR, 70125 Bari, Italy;
| | - Federica Dal Bello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10125 Turin, Italy;
| | - Atefeh Moradi
- Unit of Experimental Medicine & Clinical Pathology, Department of Clinical and Biological Science, University of Turin, 10125 Turin, Italy; (E.A.); (C.R.); (A.M.); (M.A.)
| | - Massimo Collino
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10125 Turin, Italy;
| | - Angela Maria Serena Lezza
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (G.C.); (R.D.L.)
| | - Manuela Aragno
- Unit of Experimental Medicine & Clinical Pathology, Department of Clinical and Biological Science, University of Turin, 10125 Turin, Italy; (E.A.); (C.R.); (A.M.); (M.A.)
| | - Vito Pesce
- Department of Biosciences Biotechnologies and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (G.C.); (R.D.L.)
| |
Collapse
|
17
|
Almehmadi K, Fourman S, Buesing D, Ulrich-Lai YM. Western diet-induced obesity interferes with the HPA axis-blunting effects of palatable food in male rats. Physiol Behav 2023; 270:114285. [PMID: 37392828 PMCID: PMC10529817 DOI: 10.1016/j.physbeh.2023.114285] [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: 05/02/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
Limited intermittent consumption of palatable food reduces HPA axis responses to stress in chow-fed rats, and this effect is dependent on the rewarding properties of the palatable food. However, obesity may be a state of reduced consummatory food reward, suggesting that palatable foods may be less effective at blunting HPA axis reactivity in the context of diet-induced obesity (DIO). To test this hypothesis, adult male Long-Evans rats were given unlimited access to Western (high-fat, high-sugar) diet (WD) vs. normal chow (controls). After 8 weeks of diet exposure, rats were given limited sucrose intake (LSI) consisting of additional twice-daily access to a small amount (4 ml) of either 3% or 30% sucrose drink, or water (controls) for 2 weeks. Rats then received an acute restraint stress challenge, with collection of tail blood samples for measurement of plasma corticosterone. WD-fed rats had increased caloric intake, body weight and adiposity, as expected. Rats offered LSI (3% or 30%) readily drank the maximal amount allowed (8 ml/day) and reduced their dietary intake to compensate for the sucrose calories, such that LSI did not alter body weight regardless of diet type. In chow-fed lean rats, LSI with either 3% or 30% sucrose reduced the plasma corticosterone response to restraint stress, but this effect was absent in WD-fed DIO rats. Together, these data support the hypothesis that obesity attenuates stress blunting by palatable foods and suggest the possibility that consequently, individuals with obesity may need to consume larger amounts of palatable food to obtain adequate stress relief.
Collapse
Affiliation(s)
- Khulood Almehmadi
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, SA
| | - Sarah Fourman
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA
| | - Dana Buesing
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA
| | - Yvonne M Ulrich-Lai
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA.
| |
Collapse
|
18
|
Holly D, Kim H, Woodman CR, Massett MP. Genetic background influences arterial vasomotor function in male and female mice. Physiol Rep 2023; 11:e15824. [PMID: 37771071 PMCID: PMC10539628 DOI: 10.14814/phy2.15824] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/05/2023] [Accepted: 09/09/2023] [Indexed: 09/30/2023] Open
Abstract
The purpose of this study was to assess the influence of genetic background and sex on nitric oxide (NO)-mediated vasomotor function in arteries from different vascular territories. Vasomotor function was assessed in thoracic aorta, abdominal aorta, carotid arteries, and femoral arteries from the following mouse strains: SJL/J, DBA/2J, NZW/LacJ, and C57BL/6J. Contractile responses were assessed using the α1-adrenergic receptor agonist phenylephrine (PE, 10-9 -10-5 M). Vasorelaxation responses were assessed by examining relaxation to an endothelium-dependent vasodilator acetylcholine (ACh, 10-9 -10-5 M) and an endothelium-independent vasodilator sodium nitroprusside (SNP, 10-9 -10-5 M). To evaluate the role of NO, relaxation responses to ACh and SNP were assessed in the absence or presence of a nitric oxide synthase inhibitor (N omega-nitro-l-arginine methyl ester hydrochloride: 10-4 M). Vasomotor responses to ACh and PE varied across strains and among the arteries tested with some strains exhibiting artery-specific impairment. Results indicated some concentration-response heterogeneity in response to ACh and SNP between vessels from females and males, but no significant differences in responses to PE. Collectively, these findings indicate that vasomotor responses vary by genetic background, sex, and artery type.
Collapse
Affiliation(s)
- Dylan Holly
- Department of Kinesiology and Sport ManagementTexas A&M UniversityCollege StationTexasUSA
| | - Hyoseon Kim
- Department of Kinesiology and Sport ManagementTexas Tech UniversityLubbockTexasUSA
| | - Christopher R. Woodman
- Department of Kinesiology and Sport ManagementTexas A&M UniversityCollege StationTexasUSA
| | - Michael P. Massett
- Department of Kinesiology and Sport ManagementTexas Tech UniversityLubbockTexasUSA
| |
Collapse
|
19
|
Yoon SA, Ham YM, Han SC, Hyun HB, Go B, Jung YH, Yoo ES, Yoon WJ. Immature Persimmon ( Diospyros kaki Thunb.) Ethanol Extract Ameliorates High-Fat Diet-Induced Obesity by Modulating Lipid Metabolism. Prev Nutr Food Sci 2023; 28:263-270. [PMID: 37842245 PMCID: PMC10567593 DOI: 10.3746/pnf.2023.28.3.263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 10/17/2023] Open
Abstract
In this study, immature persimmon (Diospyros kaki Thunb.) ethanol extract was administered to an obese animal model fed a high-fat diet to measure weight change, adipose tissue weight, serum lipid level, and expression level of adipose-related genes to evaluate its efficacy. Administration of D. kaki ethanol extract (DKE) (100 and 500 mg/kg/d) decreased the body weight gain, adipose tissue weight, and serum triglyceride levels in mice fed a high-fat diet. Furthermore, it improved the leptin and adiponectin levels in the blood as well as gene expression in the liver. It also inhibited the expression of sterol regulatory element-binding protein-1c, inhibiting the production of triglyceride biosynthetic enzyme fatty acid synthesis and acetyl-CoA carboxylase, and decreased the expressions of peroxisome proliferator-activated receptor γ and CCAAT/enhancer-binding proteins that induce adipocyte differentiation. Therefore, these data suggest that DKE exerts beneficial effects on high-fat diet-induced obesity by modulating lipid metabolism in mice fed a high-fat diet.
Collapse
Affiliation(s)
- Seon-A Yoon
- Biodiversity Research Institute, Jeju Technopark, Jeju 63608, Korea
| | - Young-Min Ham
- Biodiversity Research Institute, Jeju Technopark, Jeju 63608, Korea
| | - Sang-Chul Han
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Korea
| | - Ho Bong Hyun
- Biodiversity Research Institute, Jeju Technopark, Jeju 63608, Korea
| | - Boram Go
- Biodiversity Research Institute, Jeju Technopark, Jeju 63608, Korea
| | - Yong-Hwan Jung
- Biodiversity Research Institute, Jeju Technopark, Jeju 63608, Korea
| | - Eun-Sook Yoo
- Department of Medicine, School of Medicine, Jeju National University, Jeju 63243, Korea
| | - Weon-Jong Yoon
- Biodiversity Research Institute, Jeju Technopark, Jeju 63608, Korea
| |
Collapse
|
20
|
Kang J, Park M, Oh CM, Kim T. High-fat diet-induced dopaminergic dysregulation induces REM sleep fragmentation and ADHD-like behaviors. Psychiatry Res 2023; 327:115412. [PMID: 37607442 DOI: 10.1016/j.psychres.2023.115412] [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: 01/17/2023] [Revised: 07/27/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023]
Abstract
Consumption of a high-fat diet (HFD) has been associated with reduced wakefulness and various behavioral deficits, including anxiety, depression, and anhedonia. The dopaminergic system, which plays a crucial role in sleep and ADHD, is known to be vulnerable to chronic HFD. However, the association between HFD-induced behavioral and molecular changes remains unclear. Therefore, we investigated the effects of a HFD on the dopaminergic system and its association with behavioral deficits in male mice. The mice were divided into normal diet and HFD groups and were analyzed for sleep patterns, behavior tests, and transcription levels of dopamine-related genes in the brain. The HFD group showed decreased wakefulness, increased REM sleep with fragmented patterns, decreased time spent in the center zone of the open field test, shorter immobile time in the tail suspension test, impaired visuospatial memory, and reduced sucrose preference. Additionally, the HFD group had decreased mRNA levels of D1R, COMT, and DAT in the nucleus accumbens, which negatively correlated with REM sleep proportion and REM sleep bout count. The results suggest that HFD-induced behavioral deficits were resemblance to ADHD-like behavioral phenotypes and disturbs REM sleep by dysregulating the dopaminergic system.
Collapse
Affiliation(s)
- Jiseung Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Mincheol Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
21
|
Moorkens K, Leroy JLMR, Quanico J, Baggerman G, Marei WFA. How the Oviduct Lipidomic Profile Changes over Time after the Start of an Obesogenic Diet in an Outbred Mouse Model. BIOLOGY 2023; 12:1016. [PMID: 37508445 PMCID: PMC10376370 DOI: 10.3390/biology12071016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/03/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023]
Abstract
We investigated whether a high-fat/high-sugar (HF/HS) diet alters the lipidomic profile of the oviductal epithelium (OE) and studied the patterns of these changes over time. Female outbred Swiss mice were fed either a control (10% fat) or HF/HS (60% fat, 20% fructose) diet. Mice (n = 3 per treatment per time point) were sacrificed and oviducts were collected at 3 days and 1, 4, 8, 12 and 16 weeks on the diet. Lipids in the OE were imaged using matrix-assisted laser desorption ionisation mass spectrometry imaging. Discriminative m/z values and differentially regulated lipids were determined in the HF/HS versus control OEs at each time point. Feeding the obesogenic diet resulted in acute changes in the lipid profile in the OE already after 3 days, and thus even before the development of an obese phenotype. The changes in the lipid profile of the OE progressively increased and became more persistent after long-term HF/HS diet feeding. Functional annotation revealed a differential abundance of phospholipids, sphingomyelins and lysophospholipids in particular. These alterations appear to be not only caused by the direct accumulation of the excess circulating dietary fat but also a reduction in the de novo synthesis of several lipid classes, due to oxidative stress and endoplasmic reticulum dysfunction. The described diet-induced lipidomic changes suggest alterations in the OE functions and the oviductal microenvironment which may impact crucial reproductive events that take place in the oviduct, such as fertilization and early embryo development.
Collapse
Affiliation(s)
- Kerlijne Moorkens
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Jo L M R Leroy
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Jusal Quanico
- Centre for Proteomics, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Geert Baggerman
- Centre for Proteomics, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
- Health Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium
| | - Waleed F A Marei
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| |
Collapse
|
22
|
Mattar P, Jaque C, Teske JA, Morselli E, Kerr B, Cortés V, Baudrand R, Perez-Leighton CE. Impact of short and long exposure to cafeteria diet on food intake and white adipose tissue lipolysis mediated by glucagon-like peptide 1 receptor. Front Endocrinol (Lausanne) 2023; 14:1164047. [PMID: 37293487 PMCID: PMC10244886 DOI: 10.3389/fendo.2023.1164047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction The modern food environment facilitates excessive calorie intake, a major driver of obesity. Glucagon-like peptide 1 (GLP1) is a neuroendocrine peptide that has been the basis for developing new pharmacotherapies against obesity. The GLP1 receptor (GLP1R) is expressed in central and peripheral tissues, and activation of GLP1R reduces food intake, increases the expression of thermogenic proteins in brown adipose tissue (BAT), and enhances lipolysis in white adipose tissue (WAT). Obesity decreases the efficiency of GLP1R agonists in reducing food intake and body weight. Still, whether palatable food intake before or during the early development of obesity reduces the effects of GLP1R agonists on food intake and adipose tissue metabolism remains undetermined. Further, whether GLP1R expressed in WAT contributes to these effects is unclear. Methods Food intake, expression of thermogenic BAT proteins, and WAT lipolysis were measured after central or peripheral administration of Exendin-4 (EX4), a GLP1R agonist, to mice under intermittent-short exposure to CAF diet (3 h/d for 8 days) or a longer-continuous exposure to CAF diet (24 h/d for 15 days). Ex-vivo lipolysis was measured after EX4 exposure to WAT samples from mice fed CAF or control diet for 12 weeks. . Results During intermittent-short exposure to CAF diet (3 h/d for 8 days), third ventricle injection (ICV) and intra-peritoneal administration of EX4 reduced palatable food intake. Yet, during a longer-continuous exposure to CAF diet (24 h/d for 15 days), only ICV EX4 administration reduced food intake and body weight. However, this exposure to CAF diet blocked the increase in uncoupling protein 1 (UCP1) caused by ICV EX4 administration in mice fed control diet. Finally, GLP1R expression in WAT was minimal, and EX4 failed to increase lipolysis ex-vivo in WAT tissue samples from mice fed CAF or control diet for 12 weeks. . Discussion Exposure to a CAF diet during the early stages of obesity reduces the effects of peripheral and central GLP1R agonists, and WAT does not express a functional GLP1 receptor. These data support that exposure to the obesogenic food environment, without the development or manifestation of obesity, can alter the response to GLP1R agonists. .
Collapse
Affiliation(s)
- Pamela Mattar
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian Jaque
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jennifer A. Teske
- Department of Physiology, School of Nutritional Sciences and Wellness, Graduate Interdisciplinary Programs in Physiological Sciences and Neuroscience, University of Arizona, Tucson, AZ, United States
- Department of Food Science and Nutrition at the University of Minnesota, Saint Paul, MN, United States
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Bredford Kerr
- Centro de Biología Celular y Biomedicina-CEBICEM, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Víctor Cortés
- Department of Nutrition, Diabetes, and Metabolism, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rene Baudrand
- Department of Endocrinology, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Centro Traslacional de Endocrinologia UC CETREN, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | | |
Collapse
|
23
|
Watson MA, Brar H, Gibbs ET, Wong HS, Dighe PA, McKibben B, Riedmaier S, Siu A, Polakowski JS, Segreti JA, Liu X, Chung S, Pliushchev YM, Gesmundo N, Wang Z, Vortherms TA, Brand MD. Suppression of superoxide/hydrogen peroxide production at mitochondrial site I Q decreases fat accumulation, improves glucose tolerance and normalizes fasting insulin concentration in mice fed a high-fat diet. Free Radic Biol Med 2023; 204:276-286. [PMID: 37217089 DOI: 10.1016/j.freeradbiomed.2023.05.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
We developed S1QEL1.719, a novel bioavailable S1QEL (suppressor of site IQ electron leak). S1QEL1.719 prevented superoxide/hydrogen peroxide production at site IQ of mitochondrial complex I in vitro. The free concentration giving half-maximal suppression (IC50) was 52 nM. Even at 50-fold higher concentrations S1QEL1.719 did not inhibit superoxide/hydrogen peroxide production from other sites. The IC50 for inhibition of complex I electron flow was 500-fold higher than the IC50 for suppression of superoxide/hydrogen peroxide production from site IQ. S1QEL1.719 was used to test the metabolic effects of suppressing superoxide/hydrogen peroxide production from site IQin vivo. C57BL/6J male mice fed a high-fat chow for one, two or eight weeks had increased body fat, decreased glucose tolerance, and increased fasting insulin concentrations, classic symptoms of metabolic syndrome. Daily prophylactic or therapeutic oral treatment of high-fat-fed animals with S1QEL1.719 decreased fat accumulation, strongly protected against decreased glucose tolerance and prevented or reversed the increase in fasting insulin level. Free exposures in plasma and liver at Cmax were 1-4 fold the IC50 for suppression of superoxide/hydrogen peroxide production at site IQ and substantially below levels that inhibit electron flow through complex I. These results show that the production of superoxide/hydrogen peroxide from mitochondrial site IQin vivo is necessary for the induction and maintenance of glucose intolerance caused by a high-fat diet in mice. They raise the possibility that oral administration of S1QELs may be beneficial in metabolic syndrome.
Collapse
Affiliation(s)
- Mark A Watson
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| | - Harmanmeet Brar
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| | - Edwin T Gibbs
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| | - Hoi-Shan Wong
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| | - Pratiksha A Dighe
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| | - Bryan McKibben
- AbbVie Inc, 1 North Waukegan Road, North Chicago, IL, 60064, USA.
| | | | - Amy Siu
- AbbVie Inc, 1 North Waukegan Road, North Chicago, IL, 60064, USA.
| | | | - Jason A Segreti
- AbbVie Inc, 1 North Waukegan Road, North Chicago, IL, 60064, USA.
| | - Xiaoqin Liu
- AbbVie Inc, 1 North Waukegan Road, North Chicago, IL, 60064, USA.
| | - SeungWon Chung
- AbbVie Inc, 1 North Waukegan Road, North Chicago, IL, 60064, USA.
| | | | - Nathan Gesmundo
- AbbVie Inc, 1 North Waukegan Road, North Chicago, IL, 60064, USA.
| | - Zhi Wang
- AbbVie Inc, 1 North Waukegan Road, North Chicago, IL, 60064, USA.
| | | | - Martin D Brand
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| |
Collapse
|
24
|
Wei JH, Lee WJ, Luo JL, Huang HL, Wang SC, Chou RH, Huang PH, Lin SJ. Vertical Sleeve Gastrectomy Offers Protection against Disturbed Flow-Induced Atherosclerosis in High-Fat Diet-Fed Mice. Int J Mol Sci 2023; 24:ijms24065669. [PMID: 36982743 PMCID: PMC10051344 DOI: 10.3390/ijms24065669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Bariatric surgery reduces body weight, enhances metabolic and diabetic control, and improves outcomes on obesity-related comorbidities. However, the mechanisms mediating this protection against cardiovascular diseases remain unclear. We investigated the effect of sleeve gastrectomy (SG) on vascular protection in response to shear stress-induced atherosclerosis using an overweighted and carotid artery ligation mouse model. Eight-week-old male wild-type mice (C57BL/6J) were fed a high-fat diet (HFD) for two weeks to induce weight gain and dysmetabolism. SG was performed in HFD-fed mice. Two weeks after the SG procedure, partial carotid-artery ligation was performed to promote disturbed flow-induced atherosclerosis. Compared with the control mice, HFD-fed wild-type mice exhibited increased body weight, total cholesterol level, hemoglobin A1c, and enhanced insulin resistance; SG significantly reversed these adverse effects. As expected, HFD-fed mice exhibited greater neointimal hyperplasia and atherosclerotic plaques than the control group, and the SG procedure attenuated HFD-promoted ligation-induced neointimal hyperplasia and arterial elastin fragmentation. Besides, HFD promoted ligation-induced macrophage infiltration, matrix metalloproteinase-9 expression, upregulation of inflammatory cytokines, and increased vascular endothelial growth factor secretion. SG significantly reduced the above-mentioned effects. Moreover, HFD restriction partially reversed the intimal hyperplasia caused by carotid artery ligation; however, this protective effect was significantly lower than that observed in SG-operated mice. Our study demonstrated that HFD deteriorates shear stress-induced atherosclerosis and SG mitigates vascular remodeling, and this protective effect was not comparable in HFD restriction group. These findings provide a rationale for using bariatric surgery to counter atherosclerosis in morbid obesity.
Collapse
Affiliation(s)
- Jih-Hua Wei
- Division of Cardiology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan 330, Taiwan; (J.-H.W.)
- School of Medicine, National Defense Medical Center, Taipei 114, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wei-Jei Lee
- Department of Surgery, Min-Sheng General Hospital, Taoyuan 330, Taiwan
| | - Jing-Lin Luo
- Division of Cardiology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan 330, Taiwan; (J.-H.W.)
| | - Hsin-Lei Huang
- School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei 112, Taiwan
| | - Shen-Chih Wang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Ruey-Hsing Chou
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Anesthesiology, Taipei Veteran General Hospital, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei 112, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Anesthesiology, Taipei Veteran General Hospital, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei 112, Taiwan
- Correspondence: ; Tel.: +886-2-2875-7374; Fax: +886-2-2875-7375
| | - Shing-Jong Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Anesthesiology, Taipei Veteran General Hospital, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Rd., Beitou District, Taipei 112, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
25
|
Both prolonged high-fat diet consumption and calorie restriction boost hepatic NAD+ metabolism in mice. J Nutr Biochem 2023; 115:109296. [PMID: 36849030 DOI: 10.1016/j.jnutbio.2023.109296] [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: 06/27/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
Hepatic NAD+ homeostasis is essential to metabolic flexibility upon energy balance challenges. The molecular mechanism is unclear. This study aimed to determine how the enzymes involved in NAD+ salvage (Nampt, Nmnat1, Nrk1), clearance (Nnmt, Aox1, Cyp2e1), and consumption pathways (Sirt1, Sirt3, Sirt6, Parp1, Cd38) were regulated in the liver upon energy overload or shortage, as well as their relationships with glucose and lipid metabolism. Male C57BL/6N mice were fed ad libitum with the CHOW diet, high-fat diet (HFD), or subjected to 40% calorie restriction (CR) CHOW diet for 16 weeks respectively. HFD feeding increased hepatic lipids content and inflammatory markers, while lipids accumulation was not changed by CR. Both HFD feeding and CR elevated the hepatic NAD+ levels, as well as gene and protein levels of Nampt and Nmnat1. Furthermore, both HFD feeding and CR lowered acetylation of PGC-1α in parallel with the reduced hepatic lipogenesis and enhanced fatty acid oxidation, while CR enhanced hepatic AMPK activity and gluconeogenesis. Hepatic Nampt and Nnmt gene expression negatively correlated with fasting plasma glucose levels concomitant with positive correlations with Pck1 gene expression. Nrk1 and Cyp2e1 gene expression positively correlated with fat mass and plasma cholesterol levels, as well as Srebf1 gene expression. These data highlight that hepatic NAD+ metabolism will be induced for either the down-regulation of lipogenesis upon over nutrition or up-regulation of gluconeogenesis in response to CR, thus contributing to the hepatic metabolic flexibility upon energy balance challenges.
Collapse
|
26
|
Habibovic A, Hristova M, Morris CR, Lin MCJ, Cruz LC, Ather JL, Geiszt M, Anathy V, Janssen-Heininger YMW, Poynter ME, Dixon AE, van der Vliet A. Diet-induced obesity worsens allergen-induced type 2/type 17 inflammation in airways by enhancing DUOX1 activation. Am J Physiol Lung Cell Mol Physiol 2023; 324:L228-L242. [PMID: 36625485 PMCID: PMC9942905 DOI: 10.1152/ajplung.00331.2022] [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: 10/03/2022] [Revised: 12/16/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
More than 50% of people with asthma in the United States are obese, and obesity often worsens symptoms of allergic asthma and impairs response to treatment. Based on previously established roles of the epithelial NADPH oxidase DUOX1 in allergic airway inflammation, we addressed the potential involvement of DUOX1 in altered allergic inflammation in the context of obesity. Intranasal house dust mite (HDM) allergen challenge of subjects with allergic asthma induced rapid secretion of IL-33, then IL-13, into the nasal lumen, responses that were significantly enhanced in obese asthmatic subjects (BMI >30). Induction of diet-induced obesity (DIO) in mice by high-fat diet (HFD) feeding similarly enhanced acute airway responses to intranasal HDM challenge, particularly with respect to secretion of IL-33 and type 2/type 3 cytokines, and this was associated with enhanced epithelial DUOX1 expression and was avoided in DUOX1-deficient mice. DIO also enhanced DUOX1-dependent features of chronic HDM-induced allergic inflammation. Although DUOX1 did not affect overall weight gain by HFD feeding, it contributed to glucose intolerance, suggesting a role in glucose metabolism. However, glucose intolerance induced by short-term HFD feeding, in the absence of adiposity, was not sufficient to alter HDM-induced acute airway responses. DIO was associated with enhanced presence of the adipokine leptin in the airways, and leptin enhanced DUOX1-dependent IL-13 and mucin production in airway epithelial cells. In conclusion, augmented inflammatory airway responses to HDM in obesity are associated with increases in airway epithelial DUOX1, and by increased airway epithelial leptin signaling.
Collapse
Affiliation(s)
- Aida Habibovic
- Departments of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Milena Hristova
- Departments of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Carolyn R Morris
- Departments of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Miao-Chong Joy Lin
- Departments of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Litiele C Cruz
- Departments of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Jennifer L Ather
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Miklós Geiszt
- Department of Physiology and "Lendület" Peroxidase Enzyme Research Group, Semmelweis University, Budapest, Hungary
| | - Vikas Anathy
- Departments of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Yvonne M W Janssen-Heininger
- Departments of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Matthew E Poynter
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Anne E Dixon
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Albert van der Vliet
- Departments of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
27
|
Moorkens K, Leroy JLMR, Verheyen S, Marei WFA. Effects of an obesogenic diet on the oviduct depend on the duration of feeding. PLoS One 2022; 17:e0275379. [PMID: 36174086 PMCID: PMC9522283 DOI: 10.1371/journal.pone.0275379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/15/2022] [Indexed: 12/04/2022] Open
Abstract
Research question How long does it take for an obesogenic (high-fat/high-sugar, HF/HS) diet to influence the oviductal microenvironment? What are the affected cellular pathways and are they dependent on the genetic background of the mouse model? Design Female Swiss (outbred) and C57BL/6N (B6, inbred) mice were fed either a control (10% fat) or HF/HS (60% fat, 20% fructose) diet. Body weight was measured weekly. Mice were sacrificed at 3 days (3d), 1 week (1w), 4w, 8w, 12w and 16w on the diet (n = 5 per treatment per time point). Total cholesterol concentrations and inflammatory cytokines were measured in serum. Oviductal epithelial cells (OECs) were used to study the expression of genes involved in (mitochondrial) oxidative stress (OS), endoplasmic reticulum (ER) stress and inflammation using qPCR. Results Body weight and blood cholesterol increased significantly in the HF/HS mice in both strains compared to controls. In Swiss mice, HF/HS diet acutely increased ER-stress and OS-related genes in the OECs already after 3d. Subsequently, mitochondrial and cytoplasmic antioxidants were upregulated and ER-stress was alleviated at 1w. After 4-8w (mid-phase), the expression of ER-stress and OS-related genes was increased again and persisted throughout the late-phase (12-16w). Serum inflammatory cytokines and inflammatory marker-gene expression in the OECs were increased only in the late-phase. Some of the OEC stress responses were stronger or earlier in the B6. Conclusions OECs are sensitive to an obesogenic diet and may exhibit acute stress responses already after a few days of feeding. This may impact the oviductal microenvironment and contribute to diet-induced subfertility.
Collapse
Affiliation(s)
- Kerlijne Moorkens
- Department of Veterinary Sciences, Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Antwerp, Belgium
- * E-mail:
| | - Jo L. M. R. Leroy
- Department of Veterinary Sciences, Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Sara Verheyen
- Department of Veterinary Sciences, Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Waleed F. A. Marei
- Department of Veterinary Sciences, Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Antwerp, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
28
|
Fryklund C, Neuhaus M, Morén B, Borreguero-Muñoz A, Lundmark R, Stenkula KG. Expansion of the Inguinal Adipose Tissue Depot Correlates With Systemic Insulin Resistance in C57BL/6J Mice. Front Cell Dev Biol 2022; 10:942374. [PMID: 36158197 PMCID: PMC9489915 DOI: 10.3389/fcell.2022.942374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
To accommodate surplus energy, the adipose tissue expands by increasing adipocyte size (hypertrophy) and number (hyperplasia). The presence of hypertrophic adipocytes is a key characteristic of adipose tissue dysfunction. High-fat diet (HFD) fed C57BL/6J mice are a commonly used model to study obesity and obesity-related complications. In the present study, we have characterized adipose plasticity, at both the cellular and tissue level, by examining the temporal development of systemic insulin resistance and adiposity in response to HFD-feeding for 4, 8, and 12 weeks (4w, 8w, and 12w). Within the same time frame, we examined systemic metabolic flexibility and adipose plasticity when switching from HFD- to chow-diet during the last 2 weeks of diet intervention (referred to as the reverse (REV) group: 4wREV (2w HFD+2w chow), 8wREV (6w HFD+2w chow), 12wREV (10w HFD+2w chow)). In response to HFD-feeding over time, the 12w group had impaired systemic insulin sensitivity compared to both the 4w and 8w groups, accompanied by an increase in hypertrophic inguinal adipocytes and liver triglycerides. After reversing from HFD- to chow-feeding, most parameters were completely restored to chow control levels for 4wREV and 8wREV groups. In contrast, the 12wREV group had a significantly increased number of hypertrophic adipocytes, liver triglycerides accumulation, and impaired systemic insulin sensitivity compared to chow-fed mice. Further, image analysis at the single-cell level revealed a cell-size dependent organization of actin filaments for all feeding conditions. Indeed, the impaired adipocyte size plasticity in the 12wREV group was accompanied by increased actin filamentation and reduced insulin-stimulated glucose uptake compared with chow-fed mice. In summary, these results demonstrate that the C57BL/6J HFD-feeding model has a large capacity to restore adipocyte cell size and systemic insulin sensitivity, and that a metabolic tipping point occurs between 8 and 12w of HFD-feeding where this plasticity deteriorates. We believe these findings provide substantial understanding of C57BL/6J mice as an obesity model, and that an increased pool of hypertrophic ING adipocytes could contribute to aggravated insulin resistance.
Collapse
Affiliation(s)
- Claes Fryklund
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- *Correspondence: Claes Fryklund,
| | - Mathis Neuhaus
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Björn Morén
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | | | - Karin G. Stenkula
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
29
|
Bradey AL, Fitter S, Duggan J, Wilczek V, Williams CMD, Cheney EA, Noll JE, Tangseefa P, Panagopoulos V, Zannettino ACW. Calorie restriction has no effect on bone marrow tumour burden in a Vk*MYC transplant model of multiple myeloma. Sci Rep 2022; 12:13128. [PMID: 35908046 PMCID: PMC9338941 DOI: 10.1038/s41598-022-17403-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/25/2022] [Indexed: 12/05/2022] Open
Abstract
Multiple myeloma (MM) is an incurable haematological malignancy, caused by the uncontrolled proliferation of plasma cells within the bone marrow (BM). Obesity is a known risk factor for MM, however, few studies have investigated the potential of dietary intervention to prevent MM progression. Calorie restriction (CR) is associated with many health benefits including reduced cancer incidence and progression. To investigate if CR could reduce MM progression, dietary regimes [30% CR, normal chow diet (NCD), or high fat diet (HFD)] were initiated in C57BL/6J mice. Diet-induced changes were assessed, followed by inoculation of mice with Vk*MYC MM cells (Vk14451-GFP) at 16 weeks of age. Tumour progression was monitored by serum paraprotein, and at endpoint, BM and splenic tumour burden was analysed by flow cytometry. 30% CR promoted weight loss, improved glucose tolerance, increased BM adiposity and elevated serum adiponectin compared to NCD-fed mice. Despite these metabolic changes, CR had no significant effect on serum paraprotein levels. Furthermore, endpoint analysis found that dietary changes were insufficient to affect BM tumour burden, however, HFD resulted in an average two-fold increase in splenic tumour burden. Overall, these findings suggest diet-induced BM changes may not be key drivers of MM progression in the Vk14451-GFP transplant model of myeloma.
Collapse
Affiliation(s)
- Alanah L Bradey
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Stephen Fitter
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Jvaughn Duggan
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Vicki Wilczek
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Connor M D Williams
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Emma Aj Cheney
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Jacqueline E Noll
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Pawanrat Tangseefa
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Vasilios Panagopoulos
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia. .,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia.
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide, Australia.,Precision Cancer Medicine Theme, Solid Tumour Program, South Australian Health and Medical Research Institute, Adelaide, Australia.,Department of Haematology, Royal Adelaide Hospital, Adelaide, Australia.,Central Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
30
|
Macrophages, Low-Grade Inflammation, Insulin Resistance and Hyperinsulinemia: A Mutual Ambiguous Relationship in the Development of Metabolic Diseases. J Clin Med 2022; 11:jcm11154358. [PMID: 35955975 PMCID: PMC9369133 DOI: 10.3390/jcm11154358] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic derangement with poor glycemic control accompanying overweight and obesity is associated with chronic low-grade inflammation and hyperinsulinemia. Macrophages, which present a very heterogeneous population of cells, play a key role in the maintenance of normal tissue homeostasis, but functional alterations in the resident macrophage pool as well as newly recruited monocyte-derived macrophages are important drivers in the development of low-grade inflammation. While metabolic dysfunction, insulin resistance and tissue damage may trigger or advance pro-inflammatory responses in macrophages, the inflammation itself contributes to the development of insulin resistance and the resulting hyperinsulinemia. Macrophages express insulin receptors whose downstream signaling networks share a number of knots with the signaling pathways of pattern recognition and cytokine receptors, which shape macrophage polarity. The shared knots allow insulin to enhance or attenuate both pro-inflammatory and anti-inflammatory macrophage responses. This supposedly physiological function may be impaired by hyperinsulinemia or insulin resistance in macrophages. This review discusses the mutual ambiguous relationship of low-grade inflammation, insulin resistance, hyperinsulinemia and the insulin-dependent modulation of macrophage activity with a focus on adipose tissue and liver.
Collapse
|
31
|
de Campos Zani SC, Son M, Bhullar KS, Chan CB, Wu J. IRW (Isoleucine-Arginine-Tryptophan) Improves Glucose Tolerance in High Fat Diet Fed C57BL/6 Mice via Activation of Insulin Signaling and AMPK Pathways in Skeletal Muscle. Biomedicines 2022; 10:biomedicines10061235. [PMID: 35740257 PMCID: PMC9220315 DOI: 10.3390/biomedicines10061235] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 02/01/2023] Open
Abstract
IRW (Isoleucine−Arginine−Tryptophan), has antihypertensive and anti-inflammatory properties in cells and animal models and prevents angiotensin-II- and tumor necrosis factor (TNF)-α-induced insulin resistance (IR) in vitro. We investigated the effects of IRW on body composition, glucose homeostasis and insulin sensitivity in a high-fat diet (HFD) induced insulin resistant (IR) model. C57BL/6 mice were fed HFD for 6 weeks, after which IRW was incorporated into the diet (45 or 15 mg/kg body weight (BW)) until week 14. IRW45 (at a dose of 45 mg/kg BW) reduced BW (p = 0.0327), fat mass gain (p = 0.0085), and preserved lean mass of HFD mice (p = 0.0065), concomitant with enhanced glucose tolerance and reduced fasting glucose (p < 0.001). In skeletal muscle, IRW45 increased insulin-stimulated protein kinase B (AKT) phosphorylation (p = 0.0132) and glucose transporter 4 (GLUT4) translocation (p < 0.001). Angiotensin 2 receptor (AT2R) (p = 0.0024), phosphorylated 5′-AMP-activated protein kinase (AMPKα) (p < 0.0124) and peroxisome proliferator-activated receptor gamma (PPARγ) (p < 0.001) were enhanced in skeletal muscle of IRW45-treated mice, as was the expression of genes involved in myogenesis. Plasma angiotensin converting enzyme-2 (ACE2) activity was increased (p = 0.0016). Uncoupling protein-1 in white adipose tissue (WAT) was partially restored after IRW supplementation. IRW improves glucose tolerance and body composition in HFD-fed mice and promotes glucose uptake in skeletal muscle via multiple signaling pathways, independent of angiotensin converting enzyme (ACE) inhibition.
Collapse
Affiliation(s)
| | - Myoungjin Son
- Department of Agricultural Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (M.S.); (K.S.B.)
| | - Khushwant S. Bhullar
- Department of Agricultural Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (M.S.); (K.S.B.)
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Catherine B. Chan
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada; (S.C.d.C.Z.); (C.B.C.)
- Department of Agricultural Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (M.S.); (K.S.B.)
| | - Jianping Wu
- Department of Agricultural Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (M.S.); (K.S.B.)
- Correspondence: ; Tel.: +1-780-492-6885; Fax: +1-780-492-4346
| |
Collapse
|
32
|
The Prebiotic Potential of Geraniin and Geraniin-Enriched Extract against High-Fat-Diet-Induced Metabolic Syndrome in Sprague Dawley Rats. Antioxidants (Basel) 2022; 11:antiox11040632. [PMID: 35453317 PMCID: PMC9029067 DOI: 10.3390/antiox11040632] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/02/2023] Open
Abstract
Geraniin, an ellagitannin, has ameliorative properties against high-fat diet (HFD)-induced metabolic syndrome. Since geraniin has poor bioavailability, we hypothesised the interaction of this compound with gut microbiota as the main mechanism for improving metabolic aberrations. Male Sprague Dawley rats were divided into normal diet (ND)- and HFD-fed animals and treated with geraniin and an enriched extract of geraniin (GEE). We observed that 5 mg geraniin and 115 mg GEE supplementation significantly attenuated glucose intolerance, lipopolysaccharide-binding protein, total cholesterol, triacylglyceride, and low-density lipoprotein; improved insulin sensitivity; and significantly increased adiponectin and hepatic PPARα expression. Although geraniin and GEE did not significantly alter the gut microbial composition, we found an increment in the relative abundance of a few butyrate producers such as Alloprevotella, Blautia, Lachnospiraceae NK4A136 group, and Clostridium sensu stricto 1. Geraniin and its enriched extract’s ability to ameliorate metabolic syndrome parameters while positively affecting the growth of butyrate-producing bacteria suggests its potential prebiotic role.
Collapse
|
33
|
Panzarin C, Simino LADP, Mancini MCS, Ignácio-Souza LM, Milanski M, Torsoni MA, Torsoni AS. Hepatic microRNA modulation might be an early event to non-alcoholic fatty liver disease development driven by high-fat diet in male mice. Mol Biol Rep 2022; 49:2655-2666. [PMID: 35048271 DOI: 10.1007/s11033-021-07072-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/08/2021] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Metabolic alterations caused by an imbalance of macronutrient consumption are often related to the modulation of microRNAs (miRNAs), which could alter mRNAs expression profile and accelerate the development of non-alcoholic fatty liver disease (NAFLD). AIMS This study aimed to investigate the contribution of miRNAs in modulating early stages of NAFLD in mice submitted to a high-fat diet (HFD). METHODS AND RESULTS Male Swiss mice, fed either a control diet or an HFD for 1, 3, 7, 15, 30, 56 days, were assessed for metabolic alterations, gene expression and NAFLD markers. A hepatocyte cell line was used to investigate the effects of miR-370 modulation on enzymes involved in β-oxidation. Body weight and adiposity were higher after 7 days of HFD. Fasting glucose and insulin increased after 3 and 7 days of HFD, respectively. While hepatic lipid content increased from the first day on, hepatic glycogen had a decrease after 3 days of HFD consumption. miR-370 and Let-7 expression increased with acute and chronic exposure to HFD, accompanied by carnitine palmitoyltransferase 1A (Cpt1a), acyl-CoA dehydrogenase very long chain (Acadvl) and protein kinase AMP-activated Catalytic Subunit 2 (Prkaa2) downregulation, while decreased miR-122 expression was accompanied by 1-acylglycerol-3-phosphate-O-acyltransferase (Agpat) upregulation after 56 days of HFD consumption, some of them confirmed by in vitro experiments. Despite fluctuations in TNFa and IL6 mRNA levels, molecular modulation was consistent with hepatic TG and NAFLD development. CONCLUSION Hepatic miR-370-122-Let7 miRNA modulation could be the first insult to NAFLD development, preceding changes in glycemic homeostasis and adiposity.
Collapse
Affiliation(s)
- Carolina Panzarin
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, 1300, Pedro Zaccaria St, Limeira, São Paulo, 13484-350, Brazil
| | - Laís Angélica de Paula Simino
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, 1300, Pedro Zaccaria St, Limeira, São Paulo, 13484-350, Brazil
| | - Mariana Camargo Silva Mancini
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, 1300, Pedro Zaccaria St, Limeira, São Paulo, 13484-350, Brazil
| | - Leticia Martins Ignácio-Souza
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, 1300, Pedro Zaccaria St, Limeira, São Paulo, 13484-350, Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, 1300, Pedro Zaccaria St, Limeira, São Paulo, 13484-350, Brazil
| | - Márcio Alberto Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, 1300, Pedro Zaccaria St, Limeira, São Paulo, 13484-350, Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas - UNICAMP, 1300, Pedro Zaccaria St, Limeira, São Paulo, 13484-350, Brazil.
| |
Collapse
|
34
|
Barnes CN, Wallace CW, Jacobowitz BS, Fordahl SC. Reduced phasic dopamine release and slowed dopamine uptake occur in the nucleus accumbens after a diet high in saturated but not unsaturated fat. Nutr Neurosci 2022; 25:33-45. [PMID: 31914869 PMCID: PMC7343597 DOI: 10.1080/1028415x.2019.1707421] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
High-fat diets are linked with obesity and changes in dopamine neurotransmission. Mounting evidence shows that saturated fat impacts dopamine neurons and their terminal fields, but little is known about the effect a diet high in unsaturated fat has on the dopamine system. This study sought to determine whether fat type, saturated vs. unsaturated, differentially affected body weight, blood glucose regulation, locomotor behavior, and control of dopamine release and uptake at dopamine neuron terminals in the nucleus accumbens (NAc). C57BL/6 mice were fed a control diet or a nutrient-matched diet high in saturated fat (SF), unsaturated flaxseed oil (Flax) or a blend of the two fats. After 6-weeks, mice from each high-fat diet group gained significantly more weight than Controls, but the group fed Flax gained less weight than the SF group and had fasting blood glucose levels similar to Controls. Ex-vivo fast scan cyclic voltammetry revealed the SF group also had significantly slower synaptic dopamine clearance and a reduced capacity for phasic dopamine release in the nucleus accumbens (NAc), but the Flax and Blend groups resembled Controls. These data show that different types of dietary fat have substantially different effects on metabolic phenotype and influence how dopamine terminals in the NAc regulate dopamine neurotransmission. Our data also suggests that a diet high in unsaturated fat may preserve normal metabolic and behavioral parameters as well as dopamine signaling in the NAc.
Collapse
Affiliation(s)
| | | | | | - Steve C Fordahl
- Corresponding Author: Steve C. Fordahl, Ph.D., Department of Nutrition, UNC Greensboro, 319 College Ave.; 338 Stone Bldg., Greensboro, NC 27402, Tel: 336.334.5313, Fax: 336.334.4129,
| |
Collapse
|
35
|
Lee H, Ha TY, Jung CH, Nirmala FS, Park SY, Huh YH, Ahn J. Mitochondrial dysfunction in skeletal muscle contributes to the development of acute insulin resistance in mice. J Cachexia Sarcopenia Muscle 2021; 12:1925-1939. [PMID: 34605225 PMCID: PMC8718067 DOI: 10.1002/jcsm.12794] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/12/2021] [Accepted: 08/23/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Although mounting evidence indicates that insulin resistance (IR) co-occurs with mitochondrial dysfunction in skeletal muscle, there is no clear causal link between mitochondrial dysfunction and IR pathogenesis. In this study, the exact role of mitochondria in IR development was determined. METHODS Six-week-old C57BL/6 mice were fed a high-fat diet for 2 weeks to induce acute IR or for 24 weeks to induce chronic IR (n = 8 per group). To characterize mitochondrial function, we measured citrate synthase activity, ATP content, mitochondrial DNA (mtDNA) content, and oxygen consumption rate in gastrocnemius and liver tissues. We intraperitoneally administered mitochondrial division inhibitor 1 (mdivi-1) to mice with acute IR and measured mitochondrial adaptive responses such as mitophagy, mitochondrial unfolded protein response (UPRmt), and oxidative stress (n = 6 per group). RESULTS Acute IR occurred coincidently with impaired mitochondrial function, including reduced citrate synthase activity (-37.8%, P < 0.01), ATP production (-88.0%, P < 0.01), mtDNA (-53.1%, P < 0.01), and mitochondrial respiration (-52.2% for maximal respiration, P < 0.05) in skeletal muscle but not in liver. Administration of mdivi-1 attenuated IR development by increasing mitochondrial function (+58.5% for mtDNA content, P < 0.01; 4.06 ± 0.69 to 5.84 ± 0.95 pmol/min/mg for citrate synthase activity, P < 0.05; 13.06 ± 0.70 to 34.87 ± 0.70 pmol/min/g for maximal respiration, P < 0.001). Western blot analysis showed acute IR resulted in increased autophagy (mitophagy) and UPRmt induction in muscle tissue. This adaptive response was inhibited by mdivi-1, which reduced the mitochondrial oxidative stress of skeletal muscle during acute IR. CONCLUSIONS Acute IR induced mitochondrial oxidative stress that impaired mitochondrial function in skeletal muscle. Improving mitochondrial function has important potential for treating acute IR.
Collapse
Affiliation(s)
- Hyunjung Lee
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea
| | - Tae Youl Ha
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Chang Hwa Jung
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Farida Sukma Nirmala
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Yang Hoon Huh
- Center for Electron Microscopy Research, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Jiyun Ahn
- Research Group of Natural Material and Metabolism, Korea Food Research Institute, Wanju, Republic of Korea.,Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
36
|
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a central metabolite involved in energy and redox homeostasis as well as in DNA repair and protein deacetylation reactions. Pharmacological or genetic inhibition of NAD+-degrading enzymes, external supplementation of NAD+ precursors, and transgenic overexpression of NAD+-generating enzymes have wide positive effects on metabolic health and age-associated diseases. NAD+ pools tend to decline with normal aging, obesity, and hypertension, which are all major risk factors for cardiovascular disease, and NAD+ replenishment extends healthspan, avoids metabolic syndrome, and reduces blood pressure in preclinical models. In addition, experimental elevation of NAD+ improves atherosclerosis, ischemic, diabetic, arrhythmogenic, hypertrophic, or dilated cardiomyopathies, as well as different modalities of heart failure. Here, we critically discuss cardiomyocyte-specific circuitries of NAD+ metabolism, comparatively evaluate distinct NAD+ precursors for their preclinical efficacy, and raise outstanding questions on the optimal design of clinical trials in which NAD+ replenishment or supraphysiological NAD+ elevations are assessed for the prevention or treatment of major cardiac diseases. We surmise that patients with hitherto intractable cardiac diseases such as heart failure with preserved ejection fraction may profit from the administration of NAD+ precursors. The development of such NAD+-centered treatments will rely on technological and conceptual progress on the fine regulation of NAD+ metabolism.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.).,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France (M.A., G.K.).,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM) U1138, Institut Universitaire de France (M.A., G.K.)
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.).,Institute of Physiology, Faculty of Medicine, University of Maribor, Slovenia (S.S.)
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France (M.A., G.K.).,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM) U1138, Institut Universitaire de France (M.A., G.K.).,Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris 7015, France (G.K.)
| |
Collapse
|
37
|
Nurmasitoh T, Khoiriyah U, Fidianingsih I, Arjana AZ, Devita N. Impact of Obesity on Physical Activity. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Obesity occurs due to an imbalance between the calories and the energy released. On the animal model, obesity is considered as the ground for low physical activity. This is caused by low dopamine D2 receptor in the striatum. However, this suggestion is still unproven in the human condition.
AIM: The aim of this study was to find out difference in dopamine expression in obese subjects compared to non-obese subjects when triggered by the stimuli of physical activity.
METHODS: This is a quasi-experimental study. The sample was obese and non-obese (control) female who met inclusion and exclusion criteria. Before treatment was given, subjects were asked to fill out a depression, anxiety, and exercise motivation questionnaire. All subjects were tested for vital signs, anthropometrics, and neurological examinations to determine the initial condition. Then, the subjects saw video about physical activity and were taken for blood to measure blood dopamine levels using enzyme-linked immunosorbent assay. Differences in dopamine levels between the obese and control groups were analyzed using independent t-test. The relationship between dopamine levels and exercise motivation was analyzed using Pearson.
RESULTS: The obese group’s dopamine level was 71.19 ±3.02ng/ml and the control group was 81.15 ± 3.17ng/ml (independent t-test, p = 0.032). The obese group’s motivation score was 58.46 ± 1.59 and the control group score was 62.38 ± 1.54 (independent t-test, p = 0.09). Furthermore, there was no correlation between dopamine levels and motivation scores (Pearson test, p = 0.09).
CONCLUSION: There are significant differences in dopamine levels between the obese group and the control group but no correlation between dopamine levels and exercise motivation scores.
Collapse
|
38
|
Smits A, Marei WFA, De Neubourg D, Leroy JLMR. Diet normalization or caloric restriction as a preconception care strategy to improve metabolic health and oocyte quality in obese outbred mice. Reprod Biol Endocrinol 2021; 19:166. [PMID: 34736458 PMCID: PMC8567997 DOI: 10.1186/s12958-021-00848-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Maternal metabolic disorders are linked to reduced metabolic health and oocyte quality. Obese women are advised to lose weight before conception to increase pregnancy chances. However, as human studies show no univocal guidelines, more research is necessary to provide fundamental insights in the consequences of dietary weight loss on oocyte quality. Therefore, we investigated the impact of diet normalization or calorie restricted diet for two, four or six weeks, as preconception care intervention (PCCI), in obese mice on metabolic health and oocyte quality. METHODS Outbred female mice were fed a control (CTRL) or high-fat (HF) diet for 7 weeks (7w). Afterwards, HF-mice were put on different PCCIs, resulting in four treatment groups: 1) control diet up to 13w, 2) HF diet up to 13w (HF_HF), switch from a HF (7w) to 3) an ad libitum control diet (HF_CTRL) or 4) 30% calorie restricted control diet (HF_CR) for two, four or six weeks. Body weight, metabolic health, oocyte quality and overall fertility results were assessed. RESULTS Negative effects of HF diet on metabolic health, oocyte quality and pregnancy rates were confirmed. HF_CTRL mice progressively improved insulin sensitivity, glucose tolerance, serum insulin and cholesterol from PCCI w2 to w4. No further improvements in metabolic health were present at PCCI w6. However, PCCI w6 showed best oocyte quality improvements. Mature oocytes still showed elevated lipid droplet volume and mitochondrial activity but a significant reduction in ROS levels and ROS: active mitochondria ratio compared with HF_HF mice. HF_CR mice restored overall insulin sensitivity and glucose tolerance by PCCI w4. However, serum insulin, cholesterol and ALT remained abnormal. At PCCI w6, glucose tolerance was again reduced. However, only at PCCI w6, oocytes displayed reduced ROS levels and restored mitochondrial activity compared with HF_HF mice. In addition, at PCCI w6, both PCCI groups showed decreased mitochondrial ultrastructural abnormalities compared with the HF_HF group and restored pregnancy rates. CONCLUSIONS Diet normalization for 4 weeks showed to be the shortest, most promising intervention to improve metabolic health. Most promising improvements in oocyte quality were seen after 6 weeks of intervention in both PCCI groups. This research provides fundamental insights to be considered in developing substantiated preconception guidelines for obese women planning for pregnancy.
Collapse
Affiliation(s)
- Anouk Smits
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610, Wilrijk, Belgium.
| | - Waleed F A Marei
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610, Wilrijk, Belgium
| | - Diane De Neubourg
- Centre for Reproductive Medicine - Antwerp University Hospital, University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Jo L M R Leroy
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610, Wilrijk, Belgium
| |
Collapse
|
39
|
Cartwright DM, Oakey LA, Fletcher RS, Doig CL, Heising S, Larner DP, Nasteska D, Berry CE, Heaselgrave SR, Ludwig C, Hodson DJ, Lavery GG, Garten A. Nicotinamide riboside has minimal impact on energy metabolism in mouse models of mild obesity. J Endocrinol 2021; 251:111-123. [PMID: 34370682 PMCID: PMC8494379 DOI: 10.1530/joe-21-0123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022]
Abstract
Supplementation with precursors of NAD has been shown to prevent and reverse insulin resistance, mitochondrial dysfunction, and liver damage in mouse models of diet-induced obesity. We asked whether the beneficial effects of supplementation with the NAD precursor nicotinamide riboside (NR) are dependent on mouse strain. We compared the effects of NR supplementation on whole-body energy metabolism and mitochondrial function in mildly obese C57BL/6N and C57BL/6J mice, two commonly used strains to investigate metabolism. Male C57BL/6N and C57BL/6J mice were fed a high-fat diet (HFD) or standard chow with or without NR supplementation for 8 weeks. Body and organ weights, glucose tolerance, and metabolic parameters as well as mitochondrial O2 flux in liver and muscle fibers were assessed. We found that NR supplementation had no influence on body or organ weight, glucose metabolism or hepatic lipid accumulation, energy expenditure, or metabolic flexibility but increased mitochondrial respiration in soleus muscle in both mouse strains. Strain-dependent differences were detected for body and fat depot weight, fasting blood glucose, hepatic lipid accumulation, and energy expenditure. We conclude that, in mild obesity, NR supplementation does not alter metabolic phenotype in two commonly used laboratory mouse strains.
Collapse
Affiliation(s)
- David M Cartwright
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Lucy A Oakey
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rachel S Fletcher
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Craig L Doig
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Silke Heising
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Dean P Larner
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Daniela Nasteska
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Caitlin E Berry
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sam R Heaselgrave
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Christian Ludwig
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Antje Garten
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Pediatric Research Center, Hospital for Child and Adolescent Medicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
40
|
Kim SJ, Choi SI, Jang M, Jeong YA, Kang CH, Kim GH. Combination of Limosilactobacillus fermentum MG4231 and MG4244 attenuates lipid accumulation in high-fat diet-fed obese mice. Benef Microbes 2021; 12:479-491. [PMID: 34348593 DOI: 10.3920/bm2020.0205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We investigated the anti-obesity effect and the underlying mechanisms of action of human-derived Limosilactobacillus fermentum MG4231, MG4244, and their combination, in high-fat diet-induced obese mice. Administration of the Limosilactobacillus strains decreased body weight gain, liver and adipose tissue weight, and glucose tolerance. Serum levels of total cholesterol, low-density lipoprotein-cholesterol, and leptin were reduced, while adiponectin increased. The administration of Limosilactobacillus strains improved the histopathological features of liver tissue, such as hepatic atrophy and inflammatory penetration, and significantly reduced the content of triglyceride in the liver. Limosilactobacillus administration discovered a significant reduction in the size of the adipocytes in the epididymal tissue. Limosilactobacillus treatment significantly reduced the expression of important regulators in lipid metabolism, including peroxisome proliferator-activated receptor γ, CCAAT/enhancer-binding protein α, fatty acid synthase (FAS), adipocyte-protein 2, and lipoprotein lipase in the epididymal tissue. Also, Limosilactobacillus lowered sterol regulatory element-binding protein 1-c and FAS in the liver tissue. Such changes in the expression of these regulators in both liver and epididymis tissue were caused by Limosilactobacillus upregulating phosphorylation of AMP-activated protein kinase and acetyl-CoA carboxylase. Therefore, we suggest that the use of the combination of L. fermentum MG4231 and MG4244, as probiotics could effectively inhibit adipogenesis and lipogenesis from preventing obesity.
Collapse
Affiliation(s)
- S J Kim
- Department of Health Functional Materials, Duksung Women's University, 144 gil, Dobong-gu, Seoul, 01369, Republic of Korea
| | - S-I Choi
- Department of Health Functional Materials, Duksung Women's University, 144 gil, Dobong-gu, Seoul, 01369, Republic of Korea
| | - M Jang
- Department of Food and Life Science, Inje University, Gimhae, Republic of Korea
| | - Y-A Jeong
- R&D Center, MEDIOGEN Co., Ltd., Seoul, Republic of Korea
| | - C-H Kang
- R&D Center, MEDIOGEN Co., Ltd., Seoul, Republic of Korea
| | - G-H Kim
- Department of Food and Nutrition, Duksung Women's University, 33, Samyang-ro 144-gil, Dobong-gu, Seoul, 01369, Republic of Korea
| |
Collapse
|
41
|
Sexual Dimorphism in Changes That Occur in Tissues, Organs and Plasma during the Early Stages of Obesity Development. BIOLOGY 2021; 10:biology10080717. [PMID: 34439950 PMCID: PMC8389333 DOI: 10.3390/biology10080717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
Simple Summary Obesity is a global health concern with numerous associated comorbidities. This study aims to provide a qualitative assessment of changes that may occur in tissues, organs, and plasma during the early stages of obesity development and how it may differ between male and female using a mouse model of diet induced obesity. Notable changes, not previously reported, were observed in the lungs, liver, kidney, spleen, and heart, which may suggest early signs of developing an obesity associated comorbidity. Leptin levels with notable sexual dimorphisms changes significantly in early obesity and was observed to also correlate with insulin levels. Interestingly, males and females showed different inflammatory cytokine profiles with females exhibiting a more anti-inflammatory cytokine profile, notably the IL-6/IL-10 axis of cytokine regulation may account for their significantly lower weight gain compared to males. Thus, this study provides valuable information which may aid in understanding the development of some obesity associated diseases at the early stages and could assist in developing effective intervention strategies in males and females. Abstract Despite obesity being a major health concern, information on the early clinical changes that occur in plasma and tissues during obesity development and the influence of sexual dimorphism is lacking. This study investigated changes in tissue and organ histology, macrophage infiltration, plasma hormones, lipid, and chemokine and cytokine levels in mice fed on a high fat diet for 11-weeks. An increase in adiposity, accompanied by adipocyte hypertrophy and macrophage infiltration, was observed to be significantly greater in males than females. Important changes in cell morphology and histology were noted in the lungs, liver, kidney, spleen, and heart, which may indicate early signs for developing obesity associated comorbidities. Leptin, but not adiponectin, was significantly altered during weight gain. Additionally, leptin, but not adiposity, correlated with insulin levels. Interestingly, GM-CSF, TNFα, and IL-12 (p70) were not produced in the early stages of obesity development. Meanwhile, the production of MCP-1, IP-10, RANTES, IL-10, IL-6, KC, and IL-9 were greatly influenced by sexual dimorphism. Importantly, IL-6/IL-10 axis of anti-inflammatory cytokine regulation was observed only in females and may account for their significantly lower weight gain compared to males. This study provides new knowledge on how sexual dimorphism may influence the development of obesity and associated comorbidities.
Collapse
|
42
|
Obesity-associated hyperleptinemia alters the gliovascular interface of the hypothalamus to promote hypertension. Cell Metab 2021; 33:1155-1170.e10. [PMID: 33951475 PMCID: PMC8183500 DOI: 10.1016/j.cmet.2021.04.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/27/2021] [Accepted: 04/12/2021] [Indexed: 12/21/2022]
Abstract
Pathologies of the micro- and macrovascular systems are a hallmark of the metabolic syndrome, which can lead to chronically elevated blood pressure. However, the underlying pathomechanisms involved still need to be clarified. Here, we report that an obesity-associated increase in serum leptin triggers the select expansion of the micro-angioarchitecture in pre-autonomic brain centers that regulate hemodynamic homeostasis. By using a series of cell- and region-specific loss- and gain-of-function models, we show that this pathophysiological process depends on hypothalamic astroglial hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling downstream of leptin signaling. Importantly, several distinct models of HIF1α-VEGF pathway disruption in astrocytes are protected not only from obesity-induced hypothalamic angiopathy but also from sympathetic hyperactivity or arterial hypertension. These results suggest that hyperleptinemia promotes obesity-induced hypertension via a HIF1α-VEGF signaling cascade in hypothalamic astrocytes while establishing a novel mechanistic link that connects hypothalamic micro-angioarchitecture with control over systemic blood pressure.
Collapse
|
43
|
Sergi D, Luscombe-Marsh N, Naumovski N, Abeywardena M, O'Callaghan N. Palmitic Acid, but Not Lauric Acid, Induces Metabolic Inflammation, Mitochondrial Fragmentation, and a Drop in Mitochondrial Membrane Potential in Human Primary Myotubes. Front Nutr 2021; 8:663838. [PMID: 34136519 PMCID: PMC8200524 DOI: 10.3389/fnut.2021.663838] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/22/2021] [Indexed: 12/21/2022] Open
Abstract
The chain length of saturated fatty acids may dictate their impact on inflammation and mitochondrial dysfunction, two pivotal players in the pathogenesis of insulin resistance. However, these paradigms have only been investigated in animal models and cell lines so far. Thus, the aim of this study was to compare the effect of palmitic (PA) (16:0) and lauric (LA) (12:0) acid on human primary myotubes mitochondrial health and metabolic inflammation. Human primary myotubes were challenged with either PA or LA (500 μM). After 24 h, the expression of interleukin 6 (IL-6) was assessed by quantitative polymerase chain reaction (PCR), whereas Western blot was used to quantify the abundance of the inhibitor of nuclear factor κB (IκBα), electron transport chain complex proteins and mitofusin-2 (MFN-2). Mitochondrial membrane potential and dynamics were evaluated using tetraethylbenzimidazolylcarbocyanine iodide (JC-1) and immunocytochemistry, respectively. PA, contrarily to LA, triggered an inflammatory response marked by the upregulation of IL-6 mRNA (11-fold; P < 0.01) and a decrease in IκBα (32%; P < 0.05). Furthermore, whereas PA and LA did not differently modulate the levels of mitochondrial electron transport chain complex proteins, PA induced mitochondrial fragmentation (37%; P < 0.001), decreased MFN-2 (38%; P < 0.05), and caused a drop in mitochondrial membrane potential (11%; P < 0.01) compared to control, with this effect being absent in LA-treated cells. Thus, LA, as opposed to PA, did not trigger pathogenetic mechanisms proposed to be linked with insulin resistance and therefore represents a healthier saturated fatty acid choice to potentially preserve skeletal muscle metabolic health.
Collapse
Affiliation(s)
- Domenico Sergi
- Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | | | - Nenad Naumovski
- Faculty of Health, University of Canberra, Canberra, ACT, Australia.,Functional Foods and Nutrition Research Laboratory, University of Canberra, Bruce, ACT, Australia
| | - Mahinda Abeywardena
- Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide, SA, Australia
| | - Nathan O'Callaghan
- Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Adelaide, SA, Australia
| |
Collapse
|
44
|
Whitson RH, Li SL, Zhang G, Larson GP, Itakura K. Mice with Fabp4-Cre ablation of Arid5b are resistant to diet-induced obesity and hepatic steatosis. Mol Cell Endocrinol 2021; 528:111246. [PMID: 33757861 PMCID: PMC8956154 DOI: 10.1016/j.mce.2021.111246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/19/2021] [Accepted: 03/15/2021] [Indexed: 11/29/2022]
Abstract
Mice with global deletion of Arid5b expression are lean and resistant to diet-induced obesity, and Arid5b is required for adipogenesis in a variety of in vitro models. To determine whether the lean phenotype of Arid5b-/- mice can be explained by its absence in adipose tissues, we generated mice with Fabp4-mediated ablation of Arid5b. Arid5b expression was ablated in adipocytes, from Fabp4-CREpos; Arid5bFLOX/FLOX (FSKO) mice. FSKO mice were not lean when maintained on standard chow, but males were resistant to weight gains when placed on high-fat diets (HFD). This was mainly due to decreased lipid accumulation in subcutaneous (inguinal) white adipose tissue (IWAT), and the liver. Lipid accumulation proceeded normally in gonadal WAT (GWAT) and glucose intolerance developed to the same degree in FSKO and WT controls when subjected to HFD. CD68-positive macrophages were also significantly reduced in both inguinal and gonadal fat depots. RNA-Seq analysis of IWAT adipocytes from FSKO mice on HFD revealed significant decreases in the expression of genes associated with inflammation. Although Arid5b expression was normal in livers of FSKO mice, tissue weight gains and triglyceride accumulation, and expression of genes involved in lipid metabolism were markedly reduced in livers of FSKO mice on HFD. These results suggest that Arid5b plays a critical role in lipid accumulation in specific WAT depots, and in the inflammatory signaling from WAT depots to liver that lead to lipid accumulation and hepatic steatosis.
Collapse
Affiliation(s)
- Robert H Whitson
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Shu-Lian Li
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Guoxiang Zhang
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Garrett P Larson
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| | - Keiichi Itakura
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| |
Collapse
|
45
|
Learoyd AE, Calmus R, Cunningham CN, England TJ, Farr TD, Fone KC, Kendall DA, O’Sullivan SE, Trueman RC. A pilot of the feasibility and usefulness of an aged obese model for use in stroke research. Wellcome Open Res 2021; 6:104. [PMID: 34095511 PMCID: PMC8167504 DOI: 10.12688/wellcomeopenres.16592.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
Abstract
Background: Animal models of stroke have been criticised as having poor predictive validity, lacking risk factors prevalent in an aging population. This pilot study examined the development of comorbidities in a combined aged and high-fat diet model, and then examined the feasibility of modelling stroke in such rats. Methods: Twelve-month old male Wistar-Han rats (n=15) were fed a 60% fat diet for 8 months during which monthly serial blood samples were taken to assess the development of metabolic syndrome and pro-inflammatory markers. Following this, to pilot the suitability of these rats for undergoing surgical models of stroke, they underwent 30min of middle cerebral artery occlusion (MCAO) alongside younger controls fed a standard diet (n=10). Survival, weight and functional outcome were monitored, and blood vessels and tissues collected for analysis. Results: A high fat diet in aged rats led to substantial obesity. These rats did not develop type 2 diabetes or hypertension. There was thickening of the thoracic arterial wall and vacuole formation in the liver; but of the cytokines examined changes were not seen. MCAO surgery and behavioural assessment was possible in this model (with some caveats discussed in manuscript). Conclusions: This study shows MCAO is possible in aged, obese rats. However, this model is not ideal for recapitulating the complex comorbidities commonly seen in stroke patients.
Collapse
Affiliation(s)
| | - Ryan Calmus
- Newcastle University Biosciences Institute, Newcastle University, Newcastle, NE2 4HH, UK
| | | | - Tim J. England
- Division of Medical Sciences & GEM, School of Medicine, University of Nottingham, Nottingham, DE22 3DT, UK
- University Hospitals of Derby and Burton, NHS trust, Derby, DE22 3NE, UK
| | - Tracy D. Farr
- School of Life Science, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Kevin C.F. Fone
- School of Life Science, University of Nottingham, Nottingham, NG7 2UH, UK
| | | | - Saoirse E. O’Sullivan
- Division of Medical Sciences & GEM, School of Medicine, University of Nottingham, Nottingham, DE22 3DT, UK
| | - Rebecca C. Trueman
- School of Life Science, University of Nottingham, Nottingham, NG7 2UH, UK
| |
Collapse
|
46
|
Porsche CE, Delproposto JB, Geletka L, O'Rourke R, Lumeng CN. Obesity results in adipose tissue T cell exhaustion. JCI Insight 2021; 6:139793. [PMID: 33724954 PMCID: PMC8119198 DOI: 10.1172/jci.insight.139793] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 03/10/2021] [Indexed: 12/18/2022] Open
Abstract
Despite studies implicating adipose tissue T cells (ATT) in the initiation and persistence of adipose tissue inflammation, fundamental gaps in knowledge regarding ATT function impedes progress toward understanding how obesity influences adaptive immunity. We hypothesized that ATT activation and function would have tissue-resident–specific properties and that obesity would potentiate their inflammatory properties. We assessed ATT activation and inflammatory potential within mouse and human stromal vascular fraction (SVF). Surprisingly, murine and human ATTs from obese visceral white adipose tissue exhibited impaired inflammatory characteristics upon stimulation. Both environmental and cell-intrinsic factors are implicated in ATT dysfunction. Soluble factors from obese SVF inhibit ATT activation. Additionally, chronic signaling from macrophage major histocompatibility complex II (MHCII) is necessary for ATT impairment in obese adipose tissue but is independent of increased PD1 expression. To assess intracellular signaling mechanisms responsible for ATT inflammation impairments, single-cell RNA sequencing of ATTs was performed. ATTs in obese adipose tissue exhibit enrichment of genes characteristic of T cell exhaustion and increased expression of coinhibitory receptor Btla. In sum, this work suggests that obesity-induced ATTs have functional characteristics and gene expression resembling T cell exhaustion induced by local soluble factors and cell-to-cell interactions in adipose tissue.
Collapse
Affiliation(s)
| | | | - Lynn Geletka
- Department of Pediatrics and Communicable Diseases, and
| | - Robert O'Rourke
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Surgery, Ann Arbor Veterans Affairs Healthcare System, Ann Arbor, Michigan, USA
| | - Carey N Lumeng
- Graduate Program in Immunology.,Department of Pediatrics and Communicable Diseases, and
| |
Collapse
|
47
|
Dietary-challenged mice with Alzheimer-like pathology show increased energy expenditure and reduced adipocyte hypertrophy and steatosis. Aging (Albany NY) 2021; 13:10891-10919. [PMID: 33864446 PMCID: PMC8109068 DOI: 10.18632/aging.202978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer’s disease (AD) is frequently accompanied by progressing weight loss, correlating with mortality. Counter-intuitively, weight loss in old age might predict AD onset but obesity in midlife increases AD risk. Furthermore, AD is associated with diabetes-like alterations in glucose metabolism. Here, we investigated metabolic features of amyloid precursor protein overexpressing APP23 female mice modeling AD upon long-term challenge with high-sucrose (HSD) or high-fat diet (HFD). Compared to wild type littermates (WT), APP23 females were less prone to mild HSD-induced and considerable HFD-induced glucose tolerance deterioration, despite unaltered glucose tolerance during normal-control diet. Indirect calorimetry revealed increased energy expenditure and hyperactivity in APP23 females. Dietary interventions, especially HFD, had weaker effects on lean and fat mass gain, steatosis and adipocyte hypertrophy of APP23 than WT mice, as shown by 1H-magnetic-resonance-spectroscopy, histological and biochemical analyses. Proteome analysis revealed differentially regulated expression of mitochondrial proteins in APP23 livers and brains. In conclusion, hyperactivity, increased metabolic rate, and global mitochondrial dysfunction potentially add up to the development of AD-related body weight changes in APP23 females, becoming especially evident during diet-induced metabolic challenge. These findings emphasize the importance of translating this metabolic phenotyping into human research to decode the metabolic component in AD pathogenesis.
Collapse
|
48
|
Sergi D, Luscombe-Marsh N, Heilbronn LK, Birch-Machin M, Naumovski N, Lionetti L, Proud CG, Abeywardena MY, O'Callaghan N. The Inhibition of Metabolic Inflammation by EPA Is Associated with Enhanced Mitochondrial Fusion and Insulin Signaling in Human Primary Myotubes. J Nutr 2021; 151:810-819. [PMID: 33561210 DOI: 10.1093/jn/nxaa430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/07/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Sustained fuel excess triggers low-grade inflammation that can drive mitochondrial dysfunction, a pivotal defect in the pathogenesis of insulin resistance in skeletal muscle. OBJECTIVES This study aimed to investigate whether inflammation in skeletal muscle can be prevented by EPA, and if this is associated with an improvement in mitochondrial fusion, membrane potential, and insulin signaling. METHODS Human primary myotubes were treated for 24 h with palmitic acid (PA, 500 μM) under hyperglycemic conditions (13 mM glucose), which represents nutrient overload, and in the presence or absence of EPA (100 μM). After the treatments, the expression of peroxisome proliferator-activated receptor γ coactivator 1-α (PPARGC1A) and IL6 was assessed by q-PCR. Western blot was used to measure the abundance of the inhibitor of NF-κB (IKBA), mitofusin-2 (MFN2), mitochondrial electron transport chain complex proteins, and insulin-dependent AKT (Ser473) and AKT substrate 160 (AS 160; Thr642) phosphorylation. Mitochondrial dynamics and membrane potential were evaluated using immunocytochemistry and the JC-1 (tetraethylbenzimidazolylcarbocyanine iodide) dye, respectively. Data were analyzed using 1-factor ANOVA followed by Tukey post hoc test. RESULTS Nutrient excess activated the proinflammatory NFκB signaling marked by a decrease in IKBA (40%; P < 0.05) and the upregulation of IL6 mRNA (12-fold; P < 0.001). It also promoted mitochondrial fragmentation (53%; P < 0.001). All these effects were counteracted by EPA. Furthermore, nutrient overload-induced drop in mitochondrial membrane potential (6%; P < 0.05) was prevented by EPA. Finally, EPA inhibited fuel surplus-induced impairment in insulin-mediated phosphorylation of AKT (235%; P < 0.01) and AS160 (49%; P < 0.05). CONCLUSIONS EPA inhibited NFκB signaling, which was associated with an attenuation of the deleterious effects of PA and hyperglycemia on both mitochondrial health and insulin signaling in human primary myotubes. Thus, EPA might preserve skeletal muscle metabolic health during sustained fuel excess but this requires confirmation in human clinical trials.
Collapse
Affiliation(s)
- Domenico Sergi
- Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Natalie Luscombe-Marsh
- Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Leonie K Heilbronn
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Nutrition, Diabetes & Metabolism, Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Mark Birch-Machin
- Dermatological Sciences, Translational and Clinical Research Institute, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nenad Naumovski
- Faculty of Health, University of Canberra, Canberra, ACT, Australia
| | - Lilla' Lionetti
- Department of Chemistry and Biology "A. Zambelli," University of Salerno, Fisciano, Italy
| | - Christopher G Proud
- Nutrition, Diabetes & Metabolism, Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Mahinda Y Abeywardena
- Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, Australia
| | - Nathan O'Callaghan
- Nutrition and Health Program, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Adelaide, SA, Australia
| |
Collapse
|
49
|
Darr J, Tomar A, Lassi M, Gerlini R, Berti L, Hering A, Scheid F, Hrabě de Angelis M, Witting M, Teperino R. iTAG-RNA Isolates Cell-Specific Transcriptional Responses to Environmental Stimuli and Identifies an RNA-Based Endocrine Axis. Cell Rep 2021; 30:3183-3194.e4. [PMID: 32130917 DOI: 10.1016/j.celrep.2020.02.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/06/2019] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
Biofluids contain various circulating cell-free RNAs (ccfRNAs). The composition of these ccfRNAs varies among biofluids. They constitute tantalizing biomarker candidates for several pathologies and have been demonstrated to be mediators of cellular communication. Little is known about their function in physiological and developmental settings, and most works are limited to in vitro studies. Here, we develop iTAG-RNA, a method for the unbiased tagging of RNA transcripts in mice in vivo. We use iTAG-RNA to isolate hepatocytes and kidney proximal epithelial cell-specific transcriptional responses to a dietary challenge without interfering with the tissue architecture and to identify multiple hepatocyte-secreted ccfRNAs in plasma. We also identify specific transfer of liver-derived ccfRNAs to adipose tissue and skeletal muscle, where they likely constitute a buffering system to maintain lipid homeostasis under acute high-fat-diet feeding. Our findings directly demonstrate in vivo transfer of RNAs between tissues and highlight its implications for endocrine signaling and homeostasis.
Collapse
Affiliation(s)
- Jonatan Darr
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Archana Tomar
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Maximilian Lassi
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Raffaele Gerlini
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lucia Berti
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Annette Hering
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Fabienne Scheid
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Experimental Genetics, Faculty of Life and Food Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Michael Witting
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Oberschleißheim, Germany; Chair of Analytical Food Chemistry, Technische Universität München, Freising, Germany.
| | - Raffaele Teperino
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
50
|
Clarkson-Townsend DA, Douglass AJ, Singh A, Allen RS, Uwaifo IN, Pardue MT. Impacts of high fat diet on ocular outcomes in rodent models of visual disease. Exp Eye Res 2021; 204:108440. [PMID: 33444582 PMCID: PMC7946735 DOI: 10.1016/j.exer.2021.108440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
High fat diets (HFD) have been utilized in rodent models of visual disease for over 50 years to model the effects of lipids, metabolic dysfunction, and diet-induced obesity on vision and ocular health. HFD treatment can recapitulate the pathologies of some of the leading causes of blindness, such as age-related macular degeneration (AMD) and diabetic retinopathy (DR) in rodent models of visual disease. However, there are many important factors to consider when using and interpreting these models. To synthesize our current understanding of the importance of lipid signaling, metabolism, and inflammation in HFD-driven visual disease processes, we systematically review the use of HFD in mouse and rat models of visual disease. The resulting literature is grouped into three clusters: models that solely focus on HFD treatment, models of diabetes that utilize both HFD and streptozotocin (STZ), and models of AMD that utilize both HFD and genetic models and/or other exposures. Our findings show that HFD profoundly affects vision, retinal function, many different ocular tissues, and multiple cell types through a variety of mechanisms. We delineate how HFD affects the cornea, lens, uvea, vitreous humor, retina, retinal pigmented epithelium (RPE), and Bruch's membrane (BM). Furthermore, we highlight how HFD impairs several retinal cell types, including glia (microglia), retinal ganglion cells, bipolar cells, photoreceptors, and vascular support cells (endothelial cells and pericytes). However, there are a number of gaps, limitations, and biases in the current literature. We highlight these gaps and discuss experimental design to help guide future studies. Very little is known about how HFD impacts the lens, ciliary bodies, and specific neuronal populations, such as rods, cones, bipolar cells, amacrine cells, and retinal ganglion cells. Additionally, sex bias is an important limitation in the current literature, with few HFD studies utilizing female rodents. Future studies should use ingredient-matched control diets (IMCD), include both sexes in experiments to evaluate sex-specific outcomes, conduct longitudinal metabolic and visual measurements, and capture acute outcomes. In conclusion, HFD is a systemic exposure with profound systemic effects, and rodent models are invaluable in understanding the impacts on visual and ocular disease.
Collapse
Affiliation(s)
- Danielle A Clarkson-Townsend
- Gangarosa Department of Environmental Health, Emory University, Atlanta, GA, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA
| | - Amber J Douglass
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA
| | - Anayesha Singh
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA; Emory Center for Ethics, Emory University, Atlanta, GA, USA
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ivie N Uwaifo
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA; Department of Neuroscience, Emory University, Atlanta, GA, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|