1
|
Nandy A, Helderman RCM, Thapa S, Peck SH, Richards A, Jayapalan S, Narayani N, Czech MP, Rosen CJ, Rendina-Ruedy E. Enhanced fatty acid oxidation in osteoprogenitor cells provides protection from high-fat diet induced bone dysfunction. J Bone Miner Res 2025; 40:283-298. [PMID: 39657629 PMCID: PMC11789392 DOI: 10.1093/jbmr/zjae195] [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: 03/22/2024] [Revised: 11/14/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
Bone homeostasis within the skeletal system is predominantly maintained by bone formation and resorption, where formation of new bone involves maturation of stromal cells to mineral and matrix secreting mature osteoblasts, which requires cellular energy or adenosine triphosphate. Alterations in systemic metabolism can influence osteoblast function. In line with this, type 2 diabetes mellitus (T2DM), a common metabolic disorder is also associated with reduced bone formation and increased risk of fracture. Impairment in lipid metabolism is one of the key features associated with T2DM-related pathologies in multiple tissues. Therefore, we tested the hypothesis that the reduced bone formation reported in obese murine models of impaired glucose tolerance is a function of disrupted lipid metabolism in osteoblasts. We first confirmed that mice fed a high-fat diet (HFD) have reduced bone microarchitecture along with lower bone formation rates. Interestingly, osteoblasts from obese mice harbor higher numbers of cytosolic lipid droplets along with decreased bioenergetic profiles compared to control cells. Further supporting this observation, bone cortex demonstrated higher total lipid content in HFD fed mice compared to control-fed mice. As a further proof of principle, we generated a novel murine model to conditionally delete Plin2 in osteoblast-progenitor cells using Prrx1-Cre, to enhance lipid droplet breakdown. Our data demonstrate that knocking down Plin2 in an osteoprogenitor specific manner protects from HFD induced osteoblast dysfunction. Furthermore, the mechanism of action involves enhanced osteoblast fatty acid oxidation. In conclusion, the current studies establish that HFD induced glucose intolerance leads to perturbations in osteoblast lipid metabolism, thus causing lower bone formation, which can be protected against by increasing fatty acid oxidation.
Collapse
Affiliation(s)
- Ananya Nandy
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Ron C M Helderman
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Frank H. Netter M.D. School of Medicine, Quinnipiac University, North Haven, CT 06518, United States
| | - Santosh Thapa
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Sun H Peck
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
- Department of Biomedical Engineering, Vanderbilt University School of Engineering, Nashville, TN 37232, United States
- Department of Veterans Affairs, Nashville Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN 37232, United States
| | - Alison Richards
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Shobana Jayapalan
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Nikita Narayani
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Clifford J Rosen
- Maine Health Institute for Research, Scarborough, ME 04074, United States
| | - Elizabeth Rendina-Ruedy
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, United States
| |
Collapse
|
2
|
Buchwald LM, Neess D, Hansen D, Doktor TK, Ramesh V, Steffensen LB, Blagoev B, Litchfield DW, Andresen BS, Ravnskjaer K, Færgeman NJ, Guerra B. Body weight control via protein kinase CK2: diet-induced obesity counteracted by pharmacological targeting. Metabolism 2025; 162:156060. [PMID: 39521118 DOI: 10.1016/j.metabol.2024.156060] [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: 09/02/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Protein kinase CK2 is a highly conserved enzyme implicated in the pathogenesis of various human illnesses including obesity. Despite compelling evidence for the involvement of this kinase in the pathophysiology of obesity, the molecular mechanisms by which CK2 might regulate fat metabolism are still poorly understood. METHODS AND RESULTS In this study, we aimed to elucidate the role of CK2 on lipid metabolism by employing both in vitro and in vivo approaches using mouse pre-adipocytes and a mouse model of diet-induced obesity. We show that pharmacological inhibition of CK2 by CX-4945 results in premature upregulation of p27KIP1 preventing the progression of cells into mature adipocytes by arresting their development at the intermediate phase of adipogenic differentiation. Consistent with this, we show that in vivo, CK2 regulates the expression levels and ERK-mediated phosphorylation of C/EBPβ, which is one of the earliest transcription factors responsive to adipogenic stimuli. Furthermore, we demonstrate the functional implication of CK2 in the expression of late markers of adipogenesis and factors regulating lipogenesis in liver and white adipose tissue. Finally, we show that while mice subjected to high-fat diet increased their body weight, those additionally treated with CX-4945 gained considerably less weight. NMR-based body composition analysis revealed that this is linked to significant differences in body fat mass. CONCLUSIONS Taken together, our study provides novel insights into the role of CK2 in fat metabolism in response to chronic lipid overload and confirms CK2 pharmacological targeting as a potentially powerful strategy for body weight control and/or the treatment of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Laura M Buchwald
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Ditte Neess
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Daniel Hansen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Thomas K Doktor
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Vignesh Ramesh
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Lasse B Steffensen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | - Brage S Andresen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Kim Ravnskjaer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Barbara Guerra
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
3
|
Uppuganti S, Creecy A, Fernandes D, Garrett K, Donovan K, Ahmed R, Voziyan P, Rendina-Ruedy E, Nyman JS. Bone Fragility in High Fat Diet-induced Obesity is Partially Independent of Type 2 Diabetes in Mice. Calcif Tissue Int 2024; 115:298-314. [PMID: 39012489 PMCID: PMC11333511 DOI: 10.1007/s00223-024-01252-x] [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: 04/10/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024]
Abstract
Obesity and type 2 diabetes (T2D) are risk factors for fragility fractures. It is unknown whether this elevated risk is due to a diet favoring obesity or the diabetes that often occurs with obesity. Therefore, we hypothesized that the fracture resistance of bone is lower in mice fed with a high fat diet (45% kcal; HFD) than in mice that fed on a similar, control diet (10% kcal; LFD), regardless of whether the mice developed overt T2D. Sixteen-week-old, male NON/ShiLtJ mice (resistant to T2D) and age-matched, male NONcNZO10/LtJ (prone to T2D) received a control LFD or HFD for 21 weeks. HFD increased the bodyweight to a greater extent in the ShiLtJ mice compared to the NZO10 mice, while blood glucose levels were significantly higher in NZO10 than in ShiLtJ mice. As such, the glycated hemoglobin A1c (HbA1c) levels exceeded 10% in NZO10 mice, but it remained below 6% in ShiLtJ mice. Diet did not affect HbA1c. HFD lowered trabecular number and bone volume fraction of the distal femur metaphysis (micro-computed tomography or μCT) in both strains. For the femur mid-diaphysis, HFD significantly reduced the yield moment (mechanical testing by three-point bending) in both strains but did not affect cross-sectional bone area, cortical thickness, nor cortical tissue mineral density (μCT). Furthermore, the effect of diet on yield moment was independent of the structural resistance of the femur mid-diaphysis suggesting a negative effect of HFD on characteristics of the bone matrix. However, neither Raman spectroscopy nor assays of advanced glycation end-products identified how HFD affected the matrix. HFD also lowered the resistance of cortical bone to crack growth in only the diabetic NZO10 mice (fracture toughness testing of other femur), while HFD reduced the ultimate force of the L6 vertebra in both strains (compression testing). In conclusion, the HFD-related decrease in bone strength can occur in mice resistant and prone to diabetes indicating that a diet high in fat deleteriously affects bone without necessarily causing hyperglycemia.
Collapse
Affiliation(s)
- Sasidhar Uppuganti
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Medical Center East, South Tower, 1215 21st Ave. S., Suite 4200, Nashville, TN, 37232, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, 2215B Garland Ave., Nashville, TN, 37212, USA
| | - Amy Creecy
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 550 N. University Blvd, Indianapolis, IN, 46202, USA
| | - Daniel Fernandes
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN, 37232, USA
| | - Kate Garrett
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Medical Center East, South Tower, 1215 21st Ave. S., Suite 4200, Nashville, TN, 37232, USA
| | - Kara Donovan
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN, 37232, USA
| | - Rafay Ahmed
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Medical Center East, South Tower, 1215 21st Ave. S., Suite 4200, Nashville, TN, 37232, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, 2215B Garland Ave., Nashville, TN, 37212, USA
| | - Paul Voziyan
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Medical Center East, South Tower, 1215 21st Ave. S., Suite 4200, Nashville, TN, 37232, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, 2215B Garland Ave., Nashville, TN, 37212, USA
| | - Elizabeth Rendina-Ruedy
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, 2215B Garland Ave., Nashville, TN, 37212, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, 2215 Garland Ave., Nashville, TN, 37232, USA
| | - Jeffry S Nyman
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Medical Center East, South Tower, 1215 21st Ave. S., Suite 4200, Nashville, TN, 37232, USA.
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, 2215B Garland Ave., Nashville, TN, 37212, USA.
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN, 37232, USA.
- United States Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24th Ave. S., Nashville, TN, 37212, USA.
| |
Collapse
|
4
|
Chu DT, Thi HV, Bui NL, Le NH. The effects of a diet with high fat content from lard on the health and adipose-markers' mRNA expression in mice. Sci Prog 2024; 107:368504241269431. [PMID: 39090965 PMCID: PMC11297511 DOI: 10.1177/00368504241269431] [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] [Indexed: 08/04/2024]
Abstract
Pork is one type of the most frequently consumed meat with about 30% globally. Thus, the questions regarding to the health effects of diet with high fat content from lard are raised. Here, we developed a model of mice fed with high fat (HF) from lard to investigate and have more insights on the effects of long-time feeding with HF on health. The results showed that 66 days on HF induced a significant gain in the body weight of mice, and this weight gain was associated to the deposits in the white fat, but not brown fat. The glucose tolerance, not insulin resistance, in mice was decreased by the HF diet, and this was accompanied with significantly higher blood levels of total cholesterol and triglycerides. Furthermore, the weight gains in mice fed with HF seemed to link to increased mRNA levels of adipose biomarkers in lipogenesis, including Acly and Acaca genes, in white fat tissues. Thus, our study shows that a diet with high fat from lard induced the increase in body weight, white fat depots' expansion, disruption of glucose tolerance, blood dyslipidemia, and seemed to start affecting the mRNA expression of some adipose biomarkers in a murine model.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Hue Vu Thi
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Nhat-Le Bui
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Ngoc-Hoan Le
- Faculty of Biology, Hanoi National University of Education, Hanoi, Vietnam
| |
Collapse
|
5
|
Teng CY, Kao NJ, Nguyen NTK, Lin CI, Cross TWL, Lin SH. Effects of xylo-oligosaccharide on gut microbiota, brain protein expression, and lipid profile induced by high-fat diet. J Nutr Biochem 2024; 129:109640. [PMID: 38583497 DOI: 10.1016/j.jnutbio.2024.109640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/09/2024]
Abstract
Midlife overweight and obesity are risk factors of cognitive decline and Alzheimer' s disease (AD) in late life. In addition to increasing risk of obesity and cognitive dysfunction, diets rich in fats also contributes to an imbalance of gut microbiota. Xylo-oligosaccharides (XOS) are a kind of prebiotic with several biological advantages, and can selectively promote the growth of beneficial microorganisms in the gut. To explore whether XOS can alleviate cognitive decline induced by high-fat diet (HFD) through improving gut microbiota composition, mice were fed with normal control or 60% HFD for 9 weeks to induce obesity. After that, mice were supplemented with XOS (30 g or 60 g/kg-diet) or without, respectively, for 12 weeks. The results showed that XOS inhibited weight gain, decreased epidydimal fat weight, and improved fasting blood sugar and blood lipids in mice. Additionally, XOS elevated spatial learning and memory function, decreased amyloid plaques accumulation, increased brain-derived neurotrophic factor levels, and improved neuroinflammation status in hippocampus. Changes in glycerolipids metabolism-associated lipid compounds caused by HFD in hippocampus were reversed after XOS intervention. On the other hand, after XOS intervention, increase in immune-mediated bacteria, Faecalibacterium was observed. In conclusion, XOS improved gut dysbiosis and ameliorated spatial learning and memory dysfunction caused by HFD by decreasing cognitive decline-associated biomarkers and changing lipid composition in hippocampus.
Collapse
Affiliation(s)
- Chu-Yun Teng
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Ning-Jo Kao
- Department of Nutrition and Health Sciences, Kainan University, Taoyuan, Taiwan
| | - Ngan Thi Kim Nguyen
- Program of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ching-I Lin
- Department of Nutrition and Health Sciences, Chang-Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tzu-Wen L Cross
- Departmen of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| | - Shyh-Hsiang Lin
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan; School of Food Safety, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
6
|
Fiecke C, Simsek S, Sharma AK, Gallaher DD. Effect of red wheat, aleurone, and testa layers on colon cancer biomarkers, nitrosative stress, and gut microbiome composition in rats. Food Funct 2023; 14:9617-9634. [PMID: 37814914 DOI: 10.1039/d3fo03438k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
We previously found greater reduction of colon cancer (CC) biomarkers for red wheat compared to white wheat regardless of refinement state. In the present study we examined whether the phenolic-rich aleurone and testa layers are drivers of chemoprevention by red wheat and their influence on gut microbiota composition using a 1,2-dimethylhydrazine-induced CC rat model. Rats were fed a low-fat diet (16% of energy as fat), high-fat diet (50% of energy as fat), or high-fat diet containing whole red wheat, refined red wheat, refined white wheat, or aleurone- or testa-enriched fractions for 12 weeks. Morphological markers (aberrant crypt foci, ACF) were assessed after methylene blue staining and biochemical markers (3-nitrotyrosine [3-NT], Dclk1) by immunohistochemical determination of staining positivity within aberrant crypts. Gut microbiota composition was evaluated from 16S rRNA gene sequencing of DNA extracted from cecal contents. Relative to the high-fat diet, the whole and refined red wheat, refined white wheat, and testa-enriched fraction decreased ACF, while only the refined red wheat and aleurone-enriched fraction decreased 3-NT. No significant differences were observed for Dclk1. An increase in microbial diversity was observed for the aleurone-enriched fraction (ACE index) and whole red wheat (Inverse Simpson Index). The diet groups significantly modified overall microbiome composition, including altered abundances of Lactobacillus, Mucispirillum, Phascolarctobacterium, and Blautia coccoides. These results suggest that red wheat may reduce CC risk through modifications to the gut microbiota and nitrosative stress, which may be due, in part, to the influence of dietary fiber and the phenolic-rich aleurone layer.
Collapse
Affiliation(s)
- Chelsey Fiecke
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, 55108, USA.
| | - Senay Simsek
- North Dakota State University, Department of Plant Sciences, Cereal Science Graduate Program, Fargo, ND, 58105, USA
| | - Ashok Kumar Sharma
- Department of Animal Science, University of Minnesota, St. Paul, MN, 55108, USA
| | - Daniel D Gallaher
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, 55108, USA.
| |
Collapse
|
7
|
Wali JA, Ni D, Facey HJW, Dodgson T, Pulpitel TJ, Senior AM, Raubenheimer D, Macia L, Simpson SJ. Determining the metabolic effects of dietary fat, sugars and fat-sugar interaction using nutritional geometry in a dietary challenge study with male mice. Nat Commun 2023; 14:4409. [PMID: 37479702 PMCID: PMC10362033 DOI: 10.1038/s41467-023-40039-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 07/10/2023] [Indexed: 07/23/2023] Open
Abstract
The metabolic effects of sugars and fat lie at the heart of the "carbohydrate vs fat" debate on the global obesity epidemic. Here, we use nutritional geometry to systematically investigate the interaction between dietary fat and the major monosaccharides, fructose and glucose, and their impact on body composition and metabolic health. Male mice (n = 245) are maintained on one of 18 isocaloric diets for 18-19 weeks and their metabolic status is assessed through in vivo procedures and by in vitro assays involving harvested tissue samples. We find that in the setting of low and medium dietary fat content, a 50:50 mixture of fructose and glucose (similar to high-fructose corn syrup) is more obesogenic and metabolically adverse than when either monosaccharide is consumed alone. With increasing dietary fat content, the effects of dietary sugar composition on metabolic status become less pronounced. Moreover, higher fat intake is more harmful for glucose tolerance and insulin sensitivity irrespective of the sugar mix consumed. The type of fat consumed (soy oil vs lard) does not modify these outcomes. Our work shows that both dietary fat and sugars can lead to adverse metabolic outcomes, depending on the dietary context. This study shows how the principles of the two seemingly conflicting models of obesity (the "energy balance model" and the "carbohydrate insulin model") can be valid, and it will help in progressing towards a unified model of obesity. The main limitations of this study include the use of male mice of a single strain, and not testing the metabolic effects of fructose intake via sugary drinks, which are strongly linked to human obesity.
Collapse
Affiliation(s)
- Jibran A Wali
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia.
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Harrison J W Facey
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Tim Dodgson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Tamara J Pulpitel
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Cytometry, The University of Sydney, Sydney, NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Kanashiro-Takeuchi RM, Takeuchi LM, Dulce RA, Kazmierczak K, Balkan W, Cai R, Sha W, Schally AV, Hare JM. Efficacy of a growth hormone-releasing hormone agonist in a murine model of cardiometabolic heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol 2023; 324:H739-H750. [PMID: 36897749 PMCID: PMC10151038 DOI: 10.1152/ajpheart.00601.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) represents a major unmet medical need owing to its diverse pathophysiology and lack of effective therapies. Potent synthetic, agonists (MR-356 and MR-409) of growth hormone-releasing hormone (GHRH) improve the phenotype of models of HF with reduced ejection fraction (HFrEF) and in cardiorenal models of HFpEF. Endogenous GHRH exhibits a broad range of regulatory influences in the cardiovascular (CV) system and aging and plays a role in several cardiometabolic conditions including obesity and diabetes. Whether agonists of GHRH can improve the phenotype of cardiometabolic HFpEF remains untested and unknown. Here we tested the hypothesis that MR-356 can mitigate/reverse the cardiometabolic HFpEF phenotype. C57BL6N mice received a high-fat diet (HFD) plus the nitric oxide synthase inhibitor (l-NAME) for 9 wk. After 5 wk of HFD + l-NAME regimen, animals were randomized to receive daily injections of MR-356 or placebo during a 4-wk period. Control animals received no HFD + l-NAME or agonist treatment. Our results showed the unique potential of MR-356 to treat several HFpEF-like features including cardiac hypertrophy, fibrosis, capillary rarefaction, and pulmonary congestion. MR-356 improved cardiac performance by improving diastolic function, global longitudinal strain (GLS), and exercise capacity. Importantly, the increased expression of cardiac pro-brain natriuretic peptide (pro-BNP), inducible nitric oxide synthase (iNOS), and vascular endothelial growth factor-A (VEGF-A) was restored to normal levels suggesting that MR-356 reduced myocardial stress associated with metabolic inflammation in HFpEF. Thus, agonists of GHRH may be an effective therapeutic strategy for the treatment of cardiometabolic HFpEF phenotype.NEW & NOTEWORTHY This randomized study used rigorous hemodynamic tools to test the efficacy of a synthetic GHRH agonist to improve cardiac performance in a cardiometabolic HFpEF. Daily injection of the GHRH agonist, MR-356, reduced the HFpEF-like effects as evidenced by improved diastolic dysfunction, reduced cardiac hypertrophy, fibrosis, and pulmonary congestion. Notably, end-diastolic pressure and end-diastolic pressure-volume relationship were reset to control levels. Moreover, treatment with MR-356 increased exercise capacity and reduced myocardial stress associated with metabolic inflammation in HFpEF.
Collapse
Affiliation(s)
- Rosemeire M Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Lauro M Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Raul A Dulce
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
- Division of Cardiology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Renzhi Cai
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, Florida, United States
| | - Wei Sha
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, Florida, United States
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, United States
| | - Andrew V Schally
- Division of Oncology, Department of Medicine and Endocrinology, University of Miami Miller School of Medicine, Miami, Florida, United States
- Division of Endocrinology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, Florida, United States
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, United States
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, United States
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States
- Division of Cardiology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States
| |
Collapse
|
9
|
Thanaruenin P, Sutthasupa S, Kanha N, Sangsuwan J. Antioxidation effect of alginate beads containing thyme, rosemary or geranium essential oils in lard and coconut oil. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.15884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | - Sutthira Sutthasupa
- Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
- Center of Excellence in Materials Science and Technology Chiang Mai University Chiang Mai 50200 Thailand
| | - Nattapong Kanha
- Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
| | - Jurmkwan Sangsuwan
- Faculty of Agro‐Industry Chiang Mai University Chiang Mai 50100 Thailand
- Center of Excellence in Materials Science and Technology Chiang Mai University Chiang Mai 50200 Thailand
| |
Collapse
|
10
|
Havula E, Ghazanfar S, Lamichane N, Francis D, Hasygar K, Liu Y, Alton LA, Johnstone J, Needham EJ, Pulpitel T, Clark T, Niranjan HN, Shang V, Tong V, Jiwnani N, Audia G, Alves AN, Sylow L, Mirth C, Neely GG, Yang J, Hietakangas V, Simpson SJ, Senior AM. Genetic variation of macronutrient tolerance in Drosophila melanogaster. Nat Commun 2022; 13:1637. [PMID: 35347148 PMCID: PMC8960806 DOI: 10.1038/s41467-022-29183-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 02/28/2022] [Indexed: 11/08/2022] Open
Abstract
Carbohydrates, proteins and lipids are essential nutrients to all animals; however, closely related species, populations, and individuals can display dramatic variation in diet. Here we explore the variation in macronutrient tolerance in Drosophila melanogaster using the Drosophila genetic reference panel, a collection of ~200 strains derived from a single natural population. Our study demonstrates that D. melanogaster, often considered a "dietary generalist", displays marked genetic variation in survival on different diets, notably on high-sugar diet. Our genetic analysis and functional validation identify several regulators of macronutrient tolerance, including CG10960/GLUT8, Pkn and Eip75B. We also demonstrate a role for the JNK pathway in sugar tolerance and de novo lipogenesis. Finally, we report a role for tailless, a conserved orphan nuclear hormone receptor, in regulating sugar metabolism via insulin-like peptide secretion and sugar-responsive CCHamide-2 expression. Our study provides support for the use of nutrigenomics in the development of personalized nutrition.
Collapse
Affiliation(s)
- E Havula
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - S Ghazanfar
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - N Lamichane
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - D Francis
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - K Hasygar
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Y Liu
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - L A Alton
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - J Johnstone
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - E J Needham
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - T Pulpitel
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - T Clark
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - H N Niranjan
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Shang
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Tong
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - N Jiwnani
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - G Audia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - A N Alves
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - L Sylow
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - C Mirth
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - G G Neely
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - J Yang
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Hietakangas
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - S J Simpson
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - A M Senior
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
11
|
Rendina-Ruedy E, Smith BJ. Common Dietary Modifications in Preclinical Models to Study Skeletal Health. Front Endocrinol (Lausanne) 2022; 13:932343. [PMID: 35909523 PMCID: PMC9329513 DOI: 10.3389/fendo.2022.932343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/27/2022] [Indexed: 12/03/2022] Open
Abstract
Bone is a highly dynamic tissue that undergoes continuous remodeling by bone resorbing osteoclasts and bone forming osteoblasts, a process regulated in large part by osteocytes. Dysregulation of these coupled catabolic and anabolic processes as in the case of menopause, type 2 diabetes mellitus, anorexia nervosa, and chronic kidney disease is known to increase fracture risk. Recent advances in the field of bone cell metabolism and bioenergetics have revealed that maintenance of the skeleton places a high energy demand on these cells involved in bone remodeling. These new insights highlight the reason that bone tissue is the beneficiary of a substantial proportion of cardiac output and post-prandial chylomicron remnants and requires a rich supply of nutrients. Studies designed for the specific purpose of investigating the impact of dietary modifications on bone homeostasis or that alter diet composition and food intake to produce the model can be found throughout the literature; however, confounding dietary factors are often overlooked in some of the preclinical models. This review will examine some of the common pre-clinical models used to study skeletal biology and its pathologies and the subsequent impact of various dietary factors on these model systems. Furthermore, the review will include how inadvertent effects of some of these dietary components can influence bone cell function and study outcomes.
Collapse
Affiliation(s)
- Elizabeth Rendina-Ruedy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- *Correspondence: Elizabeth Rendina-Ruedy,
| | - Brenda J. Smith
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
12
|
Phattarateera S, Ausab R, Jemkuntod N, Wiriya-amornchai A. A local green composite study: the effect of edible oil on the morphological and mechanical properties of PBS/bentonite composite. JOURNAL OF POLYMER ENGINEERING 2021. [DOI: 10.1515/polyeng-2021-0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Composites of a biodegradable thermoplastic aliphatic polyester, polybutylene succinate (PBS), with bentonite were investigated for morphological and mechanical properties. The bentonite was modified with soybean oil (SBO) and lard oil (LO) (2:98 clay:oil % by weight) by mechanical stirring and ultrasonication. The PBS/modified bentonite composite was prepared by using an internal mixer and processed by compression molding. Under bentonite modification conditions, XRD and SEM showed that the bentonite layers were broken into small layers, and the d-spacing between the layers was increased by edible oil molecules. A small plate like structure of modified bentonite composite was observed by SEM micrograph, which revealed short and long layer silicate structure non-directionally throughout the matrix phase. The mechanical properties of PBS were reinforced by this structure. The tensile modulus and elongation at break seem to depend on its directional bentonite. Interestingly, considerable improvement in impact strength was observed at over 2 wt% of clay. The impact strengths of PBS, PBS/modified BTN with SBO composite, and PBS/modified BTN with LO composite were increased from 1 to 1.5 and 2 kJ/m2, respectively. Comparatively, using LO modified bentonite had a better performance for increased interlayer and resulted in higher impact strength of the composite than that of SBO composite. The results demonstrated that PBS/modified bentonite using edible oil could be a potential alternative low cost, eco-friendly material with superior impact properties useful for further applications.
Collapse
Affiliation(s)
- Supanut Phattarateera
- National Metal and Materials Technology Center, National Science and Technology Development Agency, Thailand Science Park , Pathumthani , Thailand
| | - Rudeerat Ausab
- Materials and Process Engineering Technology, Faculty of Engineering and Technology, King Mongkut’s University of Technology North Bangkok Rayong Campus , Bangkok , Thailand
| | - Neungruthai Jemkuntod
- Materials and Process Engineering Technology, Faculty of Engineering and Technology, King Mongkut’s University of Technology North Bangkok Rayong Campus , Bangkok , Thailand
| | - Atiwat Wiriya-amornchai
- Materials and Process Engineering Technology, Faculty of Engineering and Technology, King Mongkut’s University of Technology North Bangkok Rayong Campus , Bangkok , Thailand
| |
Collapse
|
13
|
Nickl B, Qadri F, Bader M. Anti-inflammatory role of Gpnmb in adipose tissue of mice. Sci Rep 2021; 11:19614. [PMID: 34608215 PMCID: PMC8490452 DOI: 10.1038/s41598-021-99090-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/20/2021] [Indexed: 12/31/2022] Open
Abstract
Obesity can cause a chronic, low-grade inflammation, which is a critical step in the development of type II diabetes and cardiovascular diseases. Inflammation is associated with the expression of glycoprotein nonmetastatic melanoma protein b (Gpnmb), which is mainly expressed by macrophages and dendritic cells. We generated a Gpnmb-knockout mouse line using Crispr-Cas9 to assess the role of Gpnmb in a diet-induced obesity. The absence of Gpnmb did not affect body weight gain and blood lipid parameters. While wildtype animals became obese but remained otherwise metabolically healthy, Gpnmb-knockout animals developed, in addition to obesity, symptoms of metabolic syndrome such as adipose tissue inflammation, insulin resistance and liver fibrosis. We observed a strong Gpnmb expression in adipose tissue macrophages in wildtype animals and a decreased expression of most macrophage-related genes independent of their inflammatory function. This was corroborated by in vitro data showing that Gpnmb was mostly expressed by reparative macrophages while only pro-inflammatory stimuli induced shedding of Gpnmb. The data suggest that Gpnmb is ameliorating adipose tissue inflammation independent of the polarization of macrophages. Taken together, the data suggest an immune-balancing function of Gpnmb that could delay the metabolic damage caused by the induction of obesity.
Collapse
Affiliation(s)
- Bernadette Nickl
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany
| | - Fatimunnisa Qadri
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125, Berlin, Germany. .,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany. .,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany. .,Charité University Medicine, 10117, Berlin, Germany. .,Institute for Biology, University of Lübeck, 23538, Lübeck, Germany.
| |
Collapse
|
14
|
Antraco VJ, Hirata BKS, de Jesus Simão J, Cruz MM, da Silva VS, da Cunha de Sá RDC, Abdala FM, Armelin-Correa L, Alonso-Vale MIC. Omega-3 Polyunsaturated Fatty Acids Prevent Nonalcoholic Steatohepatitis (NASH) and Stimulate Adipogenesis. Nutrients 2021; 13:nu13020622. [PMID: 33671850 PMCID: PMC7918199 DOI: 10.3390/nu13020622] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/01/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
The increasing impact of obesity on global human health intensifies the importance of studies focusing on agents interfering with the metabolism and remodeling not only of the white adipose tissue (WAT) but also of the liver. In the present study, we have addressed the impact of n-3 PUFA in adipose cells' proliferation and adipogenesis, as well as in the hepatic lipid profile and morphology. Mice were induced to obesity by the consumption of a high-fat diet (HFD) for 16 weeks. At the 9th week, the treatment with fish oil (FO) was initiated and maintained until the end of the period. The FO treatment reduced the animals' body mass, plasma lipids, glucose, plasma transaminases, liver mass, triacylglycerol, and cholesterol liver content when compared to animals consuming only HFD. FO also decreased the inguinal (ing) WAT mass, reduced adipocyte volume, increased adipose cellularity (hyperplasia), and increased the proliferation of adipose-derived stromal cells (AdSCs) which corroborates the increment in the proliferation of 3T3-L1 pre-adipocytes or AdSCs treated in vitro with n-3 PUFA. After submitting the in vitro treated (n-3 PUFA) cells, 3T3-L1 and AdSCs, to an adipogenic cocktail, there was an increase in the mRNA expression of adipogenic transcriptional factors and other late adipocyte markers, as well as an increase in lipid accumulation when compared to not treated cells. Finally, the expression of browning-related genes was also higher in the n-3 PUFA treated group. We conclude that n-3 PUFA exerts an attenuating effect on body mass, dyslipidemia, and hepatic steatosis induced by HFD. FO treatment led to decreasing adiposity and adipocyte hypertrophy in ingWAT while increasing hyperplasia. Data suggest that FO treatment might induce recruitment (by increased proliferation and differentiation) of new adipocytes (white and/or beige) to the ingWAT, which is fundamental for the healthy expansion of WAT.
Collapse
|
15
|
Effects of different diets used in diet-induced obesity models on insulin resistance and vascular dysfunction in C57BL/6 mice. Sci Rep 2019; 9:19556. [PMID: 31862918 PMCID: PMC6925252 DOI: 10.1038/s41598-019-55987-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/22/2019] [Indexed: 11/11/2022] Open
Abstract
The aim of the present study was to compare different diets used to induce obesity in a head-to-head manner with a focus on insulin resistance and vascular dysfunction. Male C57BL/6J mice were put on standard chow diet (SCD), normal-fat diet (NFD), cafeteria diet (CAF) or high-fat diet (HFD) for 12 weeks starting at the age of 6 weeks. Both CAF and HFD led to obesity (weight gain of 179% and 194%, respectively), glucose intolerance and insulin resistance to a comparable extent. In aortas containing perivascular adipose tissue (PVAT), acetylcholine-induced vasodilation was best in the NFD group and worst in the CAF group. Reduced phosphorylation of endothelial nitric oxide synthase at serine 1177 was observed in both CAF and HFD groups. Plasma coagulation activity was highest in the HFD group and lowest in the SCD group. Even the NFD group had significantly higher coagulation activity than the SCD group. In conclusions, CAF and HFD are both reliable mouse diets in inducing visceral obesity, glucose intolerance and insulin resistance. CAF is more effective than HFD in causing PVAT dysfunction and vascular dysfunction, whereas hypercoagulability was mostly evident in the HFD group. Coagulation activity was higher in NFD than NCD group.
Collapse
|
16
|
Lüersen K, Röder T, Rimbach G. Drosophila melanogaster in nutrition research-the importance of standardizing experimental diets. GENES AND NUTRITION 2019; 14:3. [PMID: 30766617 PMCID: PMC6359822 DOI: 10.1186/s12263-019-0627-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
Abstract
The fruit fly Drosophila melanogaster has been increasingly recognized as an important model organism in nutrition research. In order to conduct nutritional studies in fruit flies, special attention should be given to the composition of the experimental diets. Besides complex diets, which are often based on maize, yeast, sucrose, and agar, Drosophila can be also fed chemically defined diets. These so-called holidic diets are standardized in terms of their macro- and micronutrient composition although the quantitative nutrient requirements of flies have yet not been fully established and warrant further investigations. For instance, only few studies address the fatty acid, vitamin, mineral, and trace element requirements of fruit flies. D. melanogaster may be also of interest in the field of nutritional medicine. Diet-induced diabetes and obesity models have been established, and in this context, often, the so-called high-fat and high-sugar diets are fed. However, the composition of these diets is not sufficiently defined and varies between studies. A consensus within the scientific community needs to be reached to standardize the exact composition of experimental complex and holidic diets for D. melanogaster in nutrition research. Since D. melanogaster is an established valuable model system for numerous human diseases, standardized diets are also a prerequisite to conduct diet-disease interaction studies. We suggest that a comprehensive approach, which combines deep phenotyping with disease-related Drosophila models under defined dietary conditions, might lead to the foundation of a so-called fly clinic.
Collapse
Affiliation(s)
- Kai Lüersen
- 1Institute of Human Nutrition and Food Science, University of Kiel, 24118 Kiel, Germany
| | - Thomas Röder
- 2Department of Molecular Physiology, Institute of Zoology, Kiel University, Kiel, Germany.,3Airway Research Center North, German Center for Lung Research (DZL), Kiel, Germany
| | - Gerald Rimbach
- 1Institute of Human Nutrition and Food Science, University of Kiel, 24118 Kiel, Germany
| |
Collapse
|
17
|
Probiotic Lactobacillus paracasei HII01 protects rats against obese-insulin resistance-induced kidney injury and impaired renal organic anion transporter 3 function. Clin Sci (Lond) 2018; 132:1545-1563. [DOI: 10.1042/cs20180148] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 07/04/2018] [Accepted: 07/05/2018] [Indexed: 01/24/2023]
Abstract
The relationship between gut dysbiosis and obesity is currently acknowledged to be a health topic which causes low-grade systemic inflammation and insulin resistance and may damage the kidney. Organic anion transporter 3 (Oat3) has been shown as a transporter responsible for renal handling of gut microbiota products which are involved in the progression of metabolic disorder. The present study investigated the effect of probiotic supplementation on kidney function, renal Oat3 function, inflammation, endoplasmic reticulum (ER) stress, and apoptosis in obese, insulin-resistant rats. After 12 weeks of being provided with either a normal or a high-fat diet (HF), rats were divided into normal diet (ND); ND treated with probiotics (NDL); HF; and HF treated with probiotic (HFL). Lactobacillus paracasei HII01 1 × 108 colony forming unit (CFU)/ml was administered to the rats daily by oral gavage for 12 weeks. Obese rats showed significant increases in serum lipopolysaccharide (LPS), plasma lipid profiles, and insulin resistance. Renal Oat 3 function was decreased along with kidney dysfunction in HF-fed rats. Obese rats also demonstrated the increases in inflammation, ER stress, apoptosis, and gluconeogenesis in the kidneys. These alterations were improved by Lactobacillus paracasei HII01 treatment. In conclusion, probiotic supplementation alleviated kidney inflammation, ER stress, and apoptosis, leading to improved kidney function and renal Oat3 function in obese rats. These benefits involve the attenuation of hyperlipidemia, systemic inflammation, and insulin resistance. The present study also suggested the idea of remote sensing and signaling system between gut and kidney by which probiotic might facilitate renal handling of gut microbiota products through the improvement of Oat3 function.
Collapse
|
18
|
Pellizzon MA, Ricci MR. Effects of Rodent Diet Choice and Fiber Type on Data Interpretation of Gut Microbiome and Metabolic Disease Research. ACTA ACUST UNITED AC 2018; 77:e55. [PMID: 30028909 DOI: 10.1002/cptx.55] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Poor diet reporting and improperly controlling laboratory animal diet continues to reduce our ability to interpret data effectively in animal studies. In order to make the best use of our resources and improve research transparency, proper reporting methods that include a diet design are essential to improving our understanding of the links between gut health and metabolic disease onset. This unit will focus on the importance of diet choice in laboratory animal studies, specifically as it relates to gut health, microbiome, and metabolic disease development. The two most commonly used diet types, grain-based (GB) diets, and purified ingredient diets, will each be described, with particular emphasis on their differences in dietary fiber. A further description of how these diet types and fiber can affect gut morphology and microbiota will be provided as well as how purified ingredient diets may be improved upon. © 2018 by John Wiley & Sons, Inc.
Collapse
|
19
|
Pellizzon MA, Ricci MR. The common use of improper control diets in diet-induced metabolic disease research confounds data interpretation: the fiber factor. Nutr Metab (Lond) 2018; 15:3. [PMID: 29371873 PMCID: PMC5769545 DOI: 10.1186/s12986-018-0243-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/10/2018] [Indexed: 02/06/2023] Open
Abstract
Diets used to induce metabolic disease are generally high in fat and refined carbohydrates and importantly, are usually made with refined, purified ingredients. However, researchers will often use a low fat grain-based (GB) diet containing unrefined ingredients as the control diet. Such a comparison between two completely different diet types makes it impossible to draw conclusions regarding the phenotypic differences driven by diet. While many compositional differences can account for this, one major difference that could have the greatest impact between GB and purified diets is the fiber content, both in terms of the level and composition. We will review recent data showing how fiber differences between GB diets and purified diets can significantly influence gut health and microbiota, which itself can affect metabolic disease development. Researchers need to consider the control diet carefully in order to make the best use of precious experimental resources.
Collapse
Affiliation(s)
| | - Matthew R Ricci
- Research Diets, Inc, 20 Jules Lane, New Brunswick, NJ 08901 USA
| |
Collapse
|