101
|
Vacurova E, Trnovska J, Svoboda P, Skop V, Novosadova V, Reguera DP, Petrezselyová S, Piavaux B, Endaya B, Spoutil F, Zudova D, Stursa J, Melcova M, Bielcikova Z, Werner L, Prochazka J, Sedlacek R, Huttl M, Hubackova SS, Haluzik M, Neuzil J. Mitochondrially targeted tamoxifen alleviates markers of obesity and type 2 diabetes mellitus in mice. Nat Commun 2022; 13:1866. [PMID: 35387987 PMCID: PMC8987092 DOI: 10.1038/s41467-022-29486-z] [Citation(s) in RCA: 10] [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: 08/25/2020] [Accepted: 03/17/2022] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes mellitus represents a major health problem with increasing prevalence worldwide. Limited efficacy of current therapies has prompted a search for novel therapeutic options. Here we show that treatment of pre-diabetic mice with mitochondrially targeted tamoxifen, a potential anti-cancer agent with senolytic activity, improves glucose tolerance and reduces body weight with most pronounced reduction of visceral adipose tissue due to reduced food intake, suppressed adipogenesis and elimination of senescent cells. Glucose-lowering effect of mitochondrially targeted tamoxifen is linked to improvement of type 2 diabetes mellitus-related hormones profile and is accompanied by reduced lipid accumulation in liver. Lower senescent cell burden in various tissues, as well as its inhibitory effect on pre-adipocyte differentiation, results in lower level of circulating inflammatory mediators that typically enhance metabolic dysfunction. Targeting senescence with mitochodrially targeted tamoxifen thus represents an approach to the treatment of type 2 diabetes mellitus and its related comorbidities, promising a complex impact on senescence-related pathologies in aging population of patients with type 2 diabetes mellitus with potential translation into the clinic.
Collapse
Affiliation(s)
- Eliska Vacurova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Jaroslava Trnovska
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Svoboda
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Vojtech Skop
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, 20892, USA
| | - Vendula Novosadova
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - David Pajuelo Reguera
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Silvia Petrezselyová
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Benoit Piavaux
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Berwini Endaya
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Frantisek Spoutil
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Dagmar Zudova
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jan Stursa
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Magdalena Melcova
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Prague, Czech Republic
| | | | - Lukas Werner
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jan Prochazka
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Radislav Sedlacek
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Martina Huttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Martin Haluzik
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic.
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD, Australia.
| |
Collapse
|
102
|
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
|
103
|
Rodrigo N, Chen H, Pollock CA, Glastras SJ. Preconception weight loss improves fertility and maternal outcomes in obese mice. J Endocrinol 2022; 253:27-38. [PMID: 35080198 DOI: 10.1530/joe-21-0399] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/26/2022] [Indexed: 11/08/2022]
Abstract
Women with obesity have higher incidences of infertility, with longer time to conception and increased risk of pregnancy complications compared to women with normal body weight. There is a lack of evidence demonstrating the benefit of preconception maternal weight loss on fertility and pregnancy outcomes. We aimed to determine if preconception weight loss, either with diet modification or glucose-like peptide 1 receptor agonist liraglutide, improves maternal weight, fertility, and pregnancy outcomes. C57BL/6 female mice were fed either a high-fat diet (HFD) or chow for 8 weeks. HFD-fed dams were administered liraglutide (0.3 mg/kg, s.c., for 4 weeks) or switched to chow to induce weight loss. Prior to mating, liraglutide was ceased and mice continued on HFD. Mice in the 'diet switch' group continued on chow. Pregnancy rates were recorded. Maternal anthropometry and glucose tolerance were measured before and after the intervention and at late gestation. Offspring outcomes were assessed. Liraglutide or diet switch led to weight reduction, improved insulin resistance (P< 0.001), and enhanced fertility, particularly in the liraglutide group (P< 0.005). Liraglutide-treated mice had significantly higher gestational weight gain (GWG) compared to the diet switch group (P< 0.05), with similar weight and glucose tolerance in late gestation to HFD mice. In contrast, diet switch maintained similar weight and glucose tolerance in late gestation to control mice. Pre-pregnancy weight intervention with liraglutide was effective at restoring fertility. Diet modification also improved fertility and avoided catch up weight gain in pregnancy. Liraglutide may be a therapeutic strategy for weight loss to prepare for pregnancy. However, our study provides caution about the potential for excessive GWG without diet intervention in pregnancy.
Collapse
Affiliation(s)
- Natassia Rodrigo
- Renal Research Laboratory, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia
- North Precinct, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Diabetes, Endocrinology and Metabolism, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Department of Diabetes and Endocrinology, Nepean Hospital, Sydney, New South Wales, Australia
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Carol A Pollock
- Renal Research Laboratory, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia
- North Precinct, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Sarah J Glastras
- Renal Research Laboratory, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia
- North Precinct, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Diabetes, Endocrinology and Metabolism, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
104
|
Systems-level analysis of insulin action in mouse strains provides insight into tissue- and pathway-specific interactions that drive insulin resistance. Cell Metab 2022; 34:227-239.e6. [PMID: 35021042 DOI: 10.1016/j.cmet.2021.12.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/13/2021] [Accepted: 12/10/2021] [Indexed: 02/08/2023]
Abstract
Skeletal muscle and adipose tissue insulin resistance are major drivers of metabolic disease. To uncover pathways involved in insulin resistance, specifically in these tissues, we leveraged the metabolic diversity of different dietary exposures and discrete inbred mouse strains. This revealed that muscle insulin resistance was driven by gene-by-environment interactions and was strongly correlated with hyperinsulinemia and decreased levels of ten key glycolytic enzymes. Remarkably, there was no relationship between muscle and adipose tissue insulin action. Adipocyte size profoundly varied across strains and diets, and this was strongly correlated with adipose tissue insulin resistance. The A/J strain, in particular, exhibited marked adipocyte insulin resistance and hypertrophy despite robust muscle insulin responsiveness, challenging the role of adipocyte hypertrophy per se in systemic insulin resistance. These data demonstrate that muscle and adipose tissue insulin resistance can occur independently and underscore the need for tissue-specific interrogation to understand metabolic disease.
Collapse
|
105
|
Rai V, Moellmer R, Agrawal DK. Clinically relevant experimental rodent models of diabetic foot ulcer. Mol Cell Biochem 2022; 477:1239-1247. [PMID: 35089527 DOI: 10.1007/s11010-022-04372-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/23/2022] [Indexed: 12/19/2022]
Abstract
Chronic wounds are a substantial clinical problem in diabetes and nearly 6% of diabetics suffer from foot disease including ulceration, infection, and tissue necrosis. Wound healing in diabetes is impaired and delayed and is augmented by diabetic complications. Wound healing involves complex cellular, molecular, and biochemical processes and animal models are the most suitable prototype to investigate and understand the underlying pathological changes in the process of wound healing. Animal models are also useful in evaluating the safety and efficacy of newer therapeutic agents and improving the clinical approaches for human patients with chronic ulcers. The wound healing strategies get more complicated in the presence of diabetes and its associated complication. Despite the advancement in methods of wound healing, the healing of the chronic diabetic foot ulcer (DFU) remains an important clinical problem resulting in costly and prolonged treatment and poses a risk for major amputation. Saying that it is important to elucidate the newer therapeutic targets and strategies via an in-depth understanding of the complicated cascade of the chronic DFU. A major challenge in translating lab findings to clinics is the lack of an optimal preclinical model capable of properly recapitulating human wounds. Both small and large animal models of wound healing involving rodents, rabbits, and pigs have been discussed. Mouse and rats as small animal models and pig as large animal models have been discussed in association with the diabetic wound but there are advantages and limitations for each model. In this review, we critically reviewed the pros and cons of experimental models of diabetic wound healing with a focus on type II diabetes rodent models.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| | - Rebecca Moellmer
- Western University College of Podiatric Medicine, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| |
Collapse
|
106
|
Mechanism of the switch from NO to H 2O 2 in endothelium-dependent vasodilation in diabetes. Basic Res Cardiol 2022; 117:2. [PMID: 35024970 PMCID: PMC8886611 DOI: 10.1007/s00395-022-00910-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023]
Abstract
Coronary microvascular dysfunction is prevalent among people with diabetes and is correlated with cardiac mortality. Compromised endothelial-dependent dilation (EDD) is an early event in the progression of diabetes, but its mechanisms remain incompletely understood. Nitric oxide (NO) is the major endothelium-dependent vasodilatory metabolite in the healthy coronary circulation, but this switches to hydrogen peroxide (H2O2) in coronary artery disease (CAD) patients. Because diabetes is a significant risk factor for CAD, we hypothesized that a similar NO-to-H2O2 switch would occur in diabetes. Vasodilation was measured ex vivo in isolated coronary arteries from wild type (WT) and microRNA-21 (miR-21) null mice on a chow or high-fat/high-sugar diet, and B6.BKS(D)-Leprdb/J (db/db) mice using myography. Myocardial blood flow (MBF), blood pressure, and heart rate were measured in vivo using contrast echocardiography and a solid-state pressure sensor catheter. RNA from coronary arteries, endothelial cells, and cardiac tissues was analyzed via quantitative real-time PCR for gene expression, and cardiac protein expression was assessed via western blot analyses. Superoxide was detected via electron paramagnetic resonance. (1) Ex vivo coronary EDD and in vivo MBF were impaired in diabetic mice. (2) Nω-Nitro-L-arginine methyl ester, an NO synthase inhibitor (L-NAME), inhibited ex vivo coronary EDD and in vivo MBF in WT. In contrast, polyethylene glycol-catalase, an H2O2 scavenger (Peg-Cat), inhibited diabetic mouse EDD ex vivo and MBF in vivo. (3) miR-21 was upregulated in diabetic mouse endothelial cells, and the deficiency of miR-21 prevented the NO-to-H2O2 switch and ameliorated diabetic mouse vasodilation impairments. (4) Diabetic mice displayed increased serum NO and H2O2, upregulated mRNA expression of Sod1, Sod2, iNos, and Cav1, and downregulated Pgc-1α in coronary arteries, but the deficiency of miR-21 reversed these changes. (5) miR-21-deficient mice exhibited increased cardiac PGC-1α, PPARα and eNOS protein and reduced endothelial superoxide. (6) Inhibition of PGC-1α changed the mRNA expression of genes regulated by miR-21, and overexpression of PGC-1α decreased the expression of miR-21 in high (25.5 mM) glucose treated coronary endothelial cells. Diabetic mice exhibit a NO-to-H2O2 switch in the mediator of coronary EDD, which contributes to microvascular dysfunction and is mediated by miR-21. This study represents the first mouse model recapitulating the NO-to-H2O2 switch seen in CAD patients in diabetes.
Collapse
|
107
|
Tordoff MG, Ellis HT. Obesity in C57BL/6J mice fed diets differing in carbohydrate and fat but not energy content. Physiol Behav 2022; 243:113644. [PMID: 34767835 PMCID: PMC8667181 DOI: 10.1016/j.physbeh.2021.113644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 01/03/2023]
Abstract
To investigate the contributions of carbohydrate and fat to obesity we measured the body weight, body composition and food intake of adult C57BL/6J mice fed ad libitum with various combinations of two semisynthetic diets that differed in carbohydrate and fat but not in protein, micronutrient or energy content. In Experiment 1, involving male mice, body weights were similar in groups fed diets comprised of (by energy) 20% protein, 75% carbohydrate and 5% fat (C75-F5) or 20% protein, 5% carbohydrate and 75% fat (C5-F75). However, mice fed a 50:50 composite mixture of the C75-F5 and C5-F75 diets (i.e., a C40-F40 diet) became substantially more obese. Mice that could choose between the C75-F5 and C5-F75 diets ate equal amounts of each diet and gained almost as much weight as did the group fed C40-F40 diet. Mice switched every day between the C75-F5 and C5-F75 diets gained no more weight than did those fed either diet exclusively. In Experiment 2, male and female mice were fed chow or one of 8 isocaloric diets that differed parametrically in carbohydrate and fat content. Groups fed diets in the middle of the range (i.e., C35-F45 or C45-F35) weighed significantly more and were significantly fatter than were those fed diets with more extreme proportions of carbohydrate and fat (e.g., C75-F5, C5-F75), an effect that was more pronounced in males than females. In Experiment 3 and 4, male mice fed versions of the C40-F40 formulation gained more weight than did those fed the C75-F5 or C5-F75 formulations irrespective of whether the carbohydrate was predominantly sucrose or predominantly starch, or whether the fat was vegetable shortening, corn oil, palm oil or canola oil; the type of carbohydrate or fat had little or no impact on body weight. In all four experiments, energy intakes differed among the diet groups but could not account for the differences in body weight. These results demonstrate that the proportion of carbohydrate and fat in the diet influences body weight independently of energy content, and that the type of carbohydrate or fat has little impact on body weight. Consuming carbohydrate and fat simultaneously or in close temporal proximity exacerbates obesity.
Collapse
|
108
|
Resveratrol ameliorates diabetic kidney injury by reducing lipotoxicity and modulates expression of components of the junctional adhesion molecule-like/sirtuin 1 lipid metabolism pathway. Eur J Pharmacol 2022; 918:174776. [DOI: 10.1016/j.ejphar.2022.174776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 12/25/2022]
|
109
|
Martinez C, Maschio DA, de Fontes CC, Vanzela EC, Benfato ID, Gazarini ML, Carneiro EM, de Oliveira CA, Collares-Buzato CB, de F. Carvalho CP. EARLY DECREASE IN CX36 IS ASSOCIATED WITH INCREASED CELL ADHESION MOLECULES (CAMs) JUNCTIONAL CONTENT IN MOUSE PANCREATIC ISLETS AFTER SHORT-TERM HIGH-FAT DIET FEEDING. Ann Anat 2022; 241:151891. [DOI: 10.1016/j.aanat.2022.151891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 11/29/2022]
|
110
|
Aggarwal R, Peng Z, Zeng N, Silva J, He L, Chen J, Debebe A, Tu T, Alba M, Chen CY, Stiles EX, Hong H, Stiles BL. Chronic Exposure to Palmitic Acid Down-Regulates AKT in Beta-Cells through Activation of mTOR. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:130-145. [PMID: 34619135 PMCID: PMC8759041 DOI: 10.1016/j.ajpath.2021.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 01/03/2023]
Abstract
High circulating lipids occurring in obese individuals and insulin-resistant patients are considered a contributing factor to type 2 diabetes. Exposure to high lipid concentration is proposed to both protect and damage beta-cells under different circumstances. Here, by feeding mice a high-fat diet (HFD) for 2 weeks to up to 14 months, the study showed that HFD initially causes the beta-cells to expand in population, whereas long-term exposure to HFD is associated with failure of beta-cells and the inability of animals to respond to glucose challenge. To prevent the failure of beta-cells and the development of type 2 diabetes, the molecular mechanisms that underlie this biphasic response of beta-cells to lipid exposure were explored. Using palmitic acid (PA) in cultured beta-cells and islets, the study demonstrated that chronic exposure to lipids leads to reduced viability and inhibition of cell cycle progression concurrent with down-regulation of a pro-growth/survival kinase AKT, independent of glucose. This AKT down-regulation by PA is correlated with the induction of mTOR/S6K activity. Inhibiting mTOR activity with rapamycin induced Raptor and restored AKT activity, allowing beta-cells to gain proliferation capacity that was lost after HFD exposure. In summary, a novel mechanism in which lipid exposure may cause the dipole effects on beta-cell growth was elucidated, where mTOR acts as a lipid sensor. These mechanisms can be novel targets for future therapeutic developments.
Collapse
Affiliation(s)
- Richa Aggarwal
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Zhechu Peng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Ni Zeng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Joshua Silva
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Lina He
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Jingyu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Anketse Debebe
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Taojian Tu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Mario Alba
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Chien-Yu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Eileen X. Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Handan Hong
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Bangyan L. Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California,Address correspondence to Bangyan L. Stiles, Ph.D., Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033.
| |
Collapse
|
111
|
Wang D, Jiang DM, Yu RR, Zhang LL, Liu YZ, Chen JX, Chen HC, Liu YP. The Effect of Aerobic Exercise on the Oxidative Capacity of Skeletal Muscle Mitochondria in Mice with Impaired Glucose Tolerance. J Diabetes Res 2022; 2022:3780156. [PMID: 35712028 PMCID: PMC9197611 DOI: 10.1155/2022/3780156] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/05/2022] [Accepted: 05/23/2022] [Indexed: 12/03/2022] Open
Abstract
METHODS Male C57BL/6J mice were randomly divided into six different experimental groups (8 animals/group): (1) normal group (NOR), (2) normal control group (NC), (3) normal + exercise group (NE), (4) IGT group (IGT), (5) IGT control group (IC), and (6) IGT+ exercise group (IE).The exercise group received aerobic exercise for 8 weeks. After the intervention, a blood glucose meter was used to detect the level of glucose tolerance in the mouse's abdominal cavity; a biochemical kit was used to detect serum lipid metabolism indicators, malondialdehyde, and superoxide dismutase levels; the ELISA method was used to detect serum insulin and mouse gastrocnemius homogenate LDH, PDH, SDH, and CCO levels. Western blot method was used to detect the protein expression levels of NOX4, PGC-1α, and Mfn2 in the gastrocnemius muscle of mice. RESULTS (1) Mice with high-fat diet for 30 weeks showed impaired glucose tolerance, insulin resistance, and lipid metabolism disorders. The level of LDH, PDH, SDH, and CCO in the gastrocnemius homogenate of mice was reduced. The expressions of NOX4 protein were significantly upregulated, while the expressions of PGC-1α and Mfn2 proteins were significantly downregulated. (2) 8-week aerobic exercise improved the disorders of glucose and lipid metabolism in IGT mice and increased homogenized LDH, PDH, SDH, and CCO levels, and the expressions of NOX4, PGC-1α, and Mfn2 proteins in the gastrocnemius muscle of mice were reversed. It is speculated that aerobic exercise can accelerate energy metabolism. CONCLUSION (1) C57BL/6 mice were fed high fat for 30 weeks and successfully constructed a mouse model of reduced diabetes; the mice with reduced diabetes have impaired glucose tolerance, insulin resistance, and lipid metabolism disorders; (2) 8 weeks of aerobic exercise improve glucose tolerance, reduce glucose tolerance in mice, reduce insulin resistance, improve lipid metabolism disorders, and reduce oxidative stress; (3) 8-week aerobic exercise reduces skeletal muscle NOX4 expression and increases glucose tolerance; reduces the expression of LDH, PDH, SDH, and CCO in mouse skeletal muscle; increases the expression level of mitochondrial fusion protein 2 and PGC-1α; improves glucose tolerance; reduces energy metabolism of mouse skeletal muscle; reduces oxidative stress; and reduces insulin resistance. It is speculated that aerobic exercise can accelerate energy metabolism. This process may involve two aspects: firstly, increase the expression level of oxidative metabolism enzymes and promote the tricarboxylic acid cycle; secondly, increase the expression of Mfn2 and accelerate mitochondria fission or fusion to regulate energy metabolism, thereby reducing oxidative stress and insulin resistance.
Collapse
Affiliation(s)
- Dan Wang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
- Key Laboratory of Kinesiological Evaluation General Administration of Sport of China, Fujian Province, China
| | - Dong-Mou Jiang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
- Key Laboratory of Kinesiological Evaluation General Administration of Sport of China, Fujian Province, China
| | - Rong-Rong Yu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
- Key Laboratory of Kinesiological Evaluation General Administration of Sport of China, Fujian Province, China
| | - Lin-Lin Zhang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
- Key Laboratory of Kinesiological Evaluation General Administration of Sport of China, Fujian Province, China
| | - Yan-Zhong Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
- Key Laboratory of Kinesiological Evaluation General Administration of Sport of China, Fujian Province, China
| | - Jia-Xin Chen
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
- Key Laboratory of Kinesiological Evaluation General Administration of Sport of China, Fujian Province, China
| | - Hai-Chun Chen
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
- Key Laboratory of Kinesiological Evaluation General Administration of Sport of China, Fujian Province, China
| | - Yi-Ping Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
- Key Laboratory of Kinesiological Evaluation General Administration of Sport of China, Fujian Province, China
| |
Collapse
|
112
|
Endothelial ADAM17 Expression in the Progression of Kidney Injury in an Obese Mouse Model of Pre-Diabetes. Int J Mol Sci 2021; 23:ijms23010221. [PMID: 35008648 PMCID: PMC8745741 DOI: 10.3390/ijms23010221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Disintegrin and metalloproteinase domain 17 (ADAM17) activates inflammatory and fibrotic processes through the shedding of various molecules such as Tumor Necrosis Factor-α (TNF-α) or Transforming Growht Factor-α (TGF-α). There is a well-recognised link between TNF-α, obesity, inflammation, and diabetes. In physiological situations, ADAM17 is expressed mainly in the distal tubular cell while, in renal damage, its expression increases throughout the kidney including the endothelium. The aim of this study was to characterize, for the first time, an experimental mouse model fed a high-fat diet (HFD) with a specific deletion of Adam17 in endothelial cells and to analyse the effects on different renal structures. Endothelial Adam17 knockout male mice and their controls were fed a high-fat diet, to induce obesity, or standard rodent chow, for 22 weeks. Glucose tolerance, urinary albumin-to-creatinine ratio, renal histology, macrophage infiltration, and galectin-3 levels were evaluated. Results showed that obese mice presented higher blood glucose levels, dysregulated glucose homeostasis, and higher body weight compared to control mice. In addition, obese wild-type mice presented an increased albumin-to-creatinine ratio; greater glomerular size and mesangial matrix expansion; and tubular fibrosis with increased galectin-3 expression. Adam17 deletion decreased the albumin-to-creatinine ratio, glomerular mesangial index, and tubular galectin-3 expression. Moreover, macrophage infiltration in the glomeruli of obese Adam17 knockout mice was reduced as compared to obese wild-type mice. In conclusion, the expression of ADAM17 in endothelial cells impacted renal inflammation, modulating the renal function and histology in an obese pre-diabetic mouse model.
Collapse
|
113
|
Pandeya PR, Lamichhane R, Lamichhane G, Lee KH, Lee HK, Rhee SJ, Jung HJ. 18KHT01, a Potent Anti-Obesity Polyherbal Formulation. Front Pharmacol 2021; 12:807081. [PMID: 34975503 PMCID: PMC8719591 DOI: 10.3389/fphar.2021.807081] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/02/2021] [Indexed: 01/11/2023] Open
Abstract
Obesity is a life-threatening metabolic disorder necessitating urgent development of safe and effective therapy. Currently, limited such therapeutic measures are available for obesity. The present study was designed to develop a novel, safe and effective herbal therapy for the management of obesity. A polyherbal formulation (18KHT01) was developed by homogeneously mixing a specific proportion of crude Quercus acutissima (acorn jelly powder), Camellia sinensis (dry leaf buds), and Geranium thunbergii (dry aerial part) along with Citrus limon (fruit juice). Synergistic antioxidant, antiadipogenic, and anti-obesity activities were evaluated by in vitro as well as in vivo studies. In vitro experiments revealed strong synergistic antioxidant and anti-adipogenic activities of 18KHT01. Molecular assessment of 18KHT01 showed significant down-regulation of vital adipogenic factors such as PPARγ, C/EBPα, aP2, SREBP-1c, FAS, and LPL. Based on the results of the preliminary toxicity study, 75 and 150 mg/kg, twice daily doses of 18KHT01 were administered to evaluate anti-obesity activity in diet-induced obese (DIO) C57BL/6J mice model. The major obesity-related parameters such as body weight, weight gain, food efficiency ratio, as well as serum lipid profile were significantly reduced by 18KHT01 with potential synergism. Also, the high-fat diet-induced insulin resistance was suggestively alleviated by the formulation, and thus ameliorated fasting blood glucose. Histological evaluation of liver and white adipose tissue revealed that the significant reduction of fat depositions and thus reduction of these tissue weights. Synergy evaluation experiments exhibited that the 18KHT01 offered strong synergism by improving efficacy and reducing the toxicity of its ingredients. Overall results evidenced the 18KHT01 as a safe and potent anti-obesity herbal therapy.
Collapse
Affiliation(s)
- Prakash Raj Pandeya
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
- Bio-Safety Research Institute, Jeonbuk National University, Iksan, South Korea
| | - Ramakanta Lamichhane
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Gopal Lamichhane
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Kyung-Hee Lee
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| | - Hyeong Kyu Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Su-jin Rhee
- Department of Pharmacy, Wonkwang University, Iksan, South Korea
| | - Hyun-Ju Jung
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, South Korea
| |
Collapse
|
114
|
Cohen DJ, Giaccagli MM, Herzfeld JD, González LN, Cuasnicú PS, Da Ros VG. Metabolic syndrome and male fertility disorders: Is there a causal link? Rev Endocr Metab Disord 2021; 22:1057-1071. [PMID: 34037916 DOI: 10.1007/s11154-021-09659-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 12/12/2022]
Abstract
Infertility is a global health problem affecting 10-15% of couples in reproductive age. Recent studies have provided growing evidence supporting that lifestyle factors can affect male fertility through alterations in endocrine profiles, spermatogenesis and/or sperm function. One of these critical factors could be the change in the food intake behavior in modern societies that produces metabolic alterations. Regarding this, metabolic syndrome (MetS) prevalence has increased in epidemic in the last 40-50 years. Although MetS is associated with advanced age, changes in lifestyles have accelerated the appearance of symptoms in the reproductive age. We review herein the current understanding of the relationship between MetS and the male reproductive status. For this purpose, in this narrative review a comprehensive literature search was made in both animal models and men, allowing us to evaluate such relationship. This analysis showed a high variability in the reproductive phenotypes observed in patients and mice suffering MetS, including sperm parameters, fertility and offspring health. In view of this, we proposed that the reproductive effects, which are diverse and not robust, observed among MetS-affected males, might depend on additional factors not associated with the metabolic condition and contributed not only by the affected male but also by his partner. With this perspective, this review provides a more accurate insight of this syndrome critical for the identification of specific diagnostic indicators and treatment of MetS-induced fertility disorders.
Collapse
Affiliation(s)
- Débora Juana Cohen
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - María Milagros Giaccagli
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Jael Dafne Herzfeld
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Lucas Nicolás González
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Patricia Sara Cuasnicú
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Vanina Gabriela Da Ros
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
115
|
Arnone D, Vallier M, Hergalant S, Chabot C, Ndiaye NC, Moulin D, Aignatoaei AM, Alberto JM, Louis H, Boulard O, Mayeur C, Dreumont N, Peuker K, Strigli A, Zeissig S, Hansmannel F, Chamaillard M, Kökten T, Peyrin-Biroulet L. Long-Term Overconsumption of Fat and Sugar Causes a Partially Reversible Pre-inflammatory Bowel Disease State. Front Nutr 2021; 8:758518. [PMID: 34869528 PMCID: PMC8637418 DOI: 10.3389/fnut.2021.758518] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022] Open
Abstract
Nutrition appears to be an important environmental factor involved in the onset of inflammatory bowel diseases (IBD) through yet poorly understood biological mechanisms. Most studies focused on fat content in high caloric diets, while refined sugars represent up to 40% of caloric intake within industrialized countries and contribute to the growing epidemics of inflammatory diseases. Herein we aim to better understand the impact of a high-fat-high-sucrose diet on intestinal homeostasis in healthy conditions and the subsequent colitis risk. We investigated the early events and the potential reversibility of high caloric diet-induced damage in mice before experimental colitis. C57BL/6 mice were fed with a high-fat or high-fat high-sucrose or control diet before experimental colitis. In healthy mice, a high-fat high-sucrose diet induces a pre-IBD state characterized by gut microbiota dysbiosis with a total depletion of bacteria belonging to Barnesiella that is associated with subclinical endoscopic lesions. An overall down-regulation of the colonic transcriptome converged with broadly decreased immune cell populations in the mesenteric lymph nodes leading to the inability to respond to tissue injury. Such in-vivo effects on microbiome and transcriptome were partially restored when returning to normal chow. Long-term consumption of diet enriched in sucrose and fat predisposes mice to colitis. This enhanced risk is preceded by gut microbiota dysbiosis and transcriptional reprogramming of colonic genes related to IBD. Importantly, diet-induced transcriptome and microbiome disturbances are partially reversible after switching back to normal chow with persistent sequelae that may contribute to IBD predisposition in the general population.
Collapse
Affiliation(s)
- Djésia Arnone
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Marie Vallier
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University and Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Sébastien Hergalant
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Caroline Chabot
- CHRU-Nancy, Pediatric Hepato-Gastroenterology and Nutrition Unit, Department of Child Medicine and Clinical Genetics, Inserm U1256, Université de Lorraine, Nancy, France
| | - Ndeye Coumba Ndiaye
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - David Moulin
- IMoPA, UMR7365 CNRS-Université de Lorraine, CHRU de Nancy, Contrat d'interface, Nancy, France
| | | | - Jean-Marc Alberto
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Huguette Louis
- Department Inserm UMRS_1116 DCAC, Université de Lorraine, Nancy, France
- Cytometry Core Facility, UMS2008 IBSLor (CNRS-Université de Lorraine-INSERM), Campus Brabois-Santé, Nancy, France
| | - Olivier Boulard
- Laboratory of Cell Physiology, INSERM U1003, University of Lille, Lille, France
| | - Camille Mayeur
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Natacha Dreumont
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Kenneth Peuker
- Center for Regenerative Therapies, Technische Universität (TU) Dresden, Dresden, Germany
- Department of Medicine I, University Medical Center Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Anne Strigli
- Center for Regenerative Therapies, Technische Universität (TU) Dresden, Dresden, Germany
- Department of Medicine I, University Medical Center Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Sebastian Zeissig
- Center for Regenerative Therapies, Technische Universität (TU) Dresden, Dresden, Germany
- Department of Medicine I, University Medical Center Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Franck Hansmannel
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | | | - Tunay Kökten
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Laurent Peyrin-Biroulet
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
- Department of Gastroenterology, CHRU-Nancy, Université de Lorraine, Nancy, France
| |
Collapse
|
116
|
Novel GPR120 Agonists with Improved Pharmacokinetic Profiles for the Treatment of Type 2 Diabetes. Molecules 2021; 26:molecules26226907. [PMID: 34833999 PMCID: PMC8624523 DOI: 10.3390/molecules26226907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/16/2022] Open
Abstract
GPR120 is a promising target for the treatment of type 2 diabetes (T2DM), which is activated by free fatty acids (FFAs) and stimulates the release of glucagon-like peptide-1(GLP-1). GLP-1, as an incretin, can enhance glucose-dependent secretion of insulin from pancreatic beta cells and reduce blood glucose. In this study, a series of novel GPR120 agonists were designed and synthesized to improve the stability and hydrophilicity of the phenylpropanoic acid GPR120 agonist TUG-891. Compound 11b showed excellent GPR120 agonistic activity and pharmacokinetic properties, and could reduce the blood glucose of normal mice in a dose-dependent manner. In addition, no hypoglycemic side effects were observed even at a dose of 100 mg/kg. Moreover, 11b showed good anti-hyperglycemic effects in diet-induced obese (DIO) mice. Molecular simulation illustrated that compound 11b could enter the active site of GPR120 and interact with ARG99. Taken together, the results indicate that compound 11b might be a promising drug candidate for the treatment of T2DM.
Collapse
|
117
|
Hulme KD, Noye EC, Short KR, Labzin LI. Dysregulated Inflammation During Obesity: Driving Disease Severity in Influenza Virus and SARS-CoV-2 Infections. Front Immunol 2021; 12:770066. [PMID: 34777390 PMCID: PMC8581451 DOI: 10.3389/fimmu.2021.770066] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Acute inflammation is a critical host defense response during viral infection. When dysregulated, inflammation drives immunopathology and tissue damage. Excessive, damaging inflammation is a hallmark of both pandemic influenza A virus (IAV) infections and Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infections. Chronic, low-grade inflammation is also a feature of obesity. In recent years, obesity has been recognized as a growing pandemic with significant mortality and associated costs. Obesity is also an independent risk factor for increased disease severity and death during both IAV and SARS-CoV-2 infection. This review focuses on the effect of obesity on the inflammatory response in the context of viral respiratory infections and how this leads to increased viral pathology. Here, we will review the fundamentals of inflammation, how it is initiated in IAV and SARS-CoV-2 infection and its link to disease severity. We will examine how obesity drives chronic inflammation and trained immunity and how these impact the immune response to IAV and SARS-CoV-2. Finally, we review both medical and non-medical interventions for obesity, how they impact on the inflammatory response and how they could be used to prevent disease severity in obese patients. As projections of global obesity numbers show no sign of slowing down, future pandemic preparedness will require us to consider the metabolic health of the population. Furthermore, if weight-loss alone is insufficient to reduce the risk of increased respiratory virus-related mortality, closer attention must be paid to a patient’s history of health, and new therapeutic options identified.
Collapse
Affiliation(s)
- Katina D Hulme
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ellesandra C Noye
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Larisa I Labzin
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
118
|
Iglesias-Carres L, Neilson AP. Utilizing preclinical models of genetic diversity to improve translation of phytochemical activities from rodents to humans and inform personalized nutrition. Food Funct 2021; 12:11077-11105. [PMID: 34672309 DOI: 10.1039/d1fo02782d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mouse models are an essential tool in different areas of research, including nutrition and phytochemical research. Traditional inbred mouse models have allowed the discovery of therapeutical targets and mechanisms of action and expanded our knowledge of health and disease. However, these models lack the genetic variability typically found in human populations, which hinders the translatability of the results found in mice to humans. The development of genetically diverse mouse models, such as the collaborative cross (CC) or the diversity outbred (DO) models, has been a useful tool to overcome this obstacle in many fields, such as cancer, immunology and toxicology. However, these tools have not yet been widely adopted in the field of phytochemical research. As demonstrated in other disciplines, use of CC and DO models has the potential to provide invaluable insights for translation of phytochemicals from rodents to humans, which are desperately needed given the challenges and numerous failed clinical trials in this field. These models may prove informative for personalized use of phytochemicals in humans, including: predicting interindividual variability in phytochemical bioavailability and efficacy, identifying genetic loci or genes governing response to phytochemicals, identifying phytochemical mechanisms of action and therapeutic targets, and understanding the impact of genetic variability on individual response to phytochemicals. Such insights would prove invaluable for personalized implementation of phytochemicals in humans. This review will focus on the current work performed with genetically diverse mouse populations, and the research opportunities and advantages that these models can offer to phytochemical research.
Collapse
Affiliation(s)
- Lisard Iglesias-Carres
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA.
| | - Andrew P Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA.
| |
Collapse
|
119
|
Short-term high-fat feeding exacerbates degeneration in retinitis pigmentosa by promoting retinal oxidative stress and inflammation. Proc Natl Acad Sci U S A 2021; 118:2100566118. [PMID: 34667124 DOI: 10.1073/pnas.2100566118] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2021] [Indexed: 12/22/2022] Open
Abstract
A high-fat diet (HFD) can induce hyperglycemia and metabolic syndromes that, in turn, can trigger visual impairment. To evaluate the acute effects of HFD feeding on retinal degeneration, we assessed retinal function and morphology, inflammatory state, oxidative stress, and gut microbiome in dystrophic retinal degeneration 10 (rd10) mice, a model of retinitis pigmentosa, fed an HFD for 2 to 3 wk. Short-term HFD feeding impaired retinal responsiveness and visual acuity and enhanced photoreceptor degeneration, microglial cell activation, and Müller cell gliosis. HFD consumption also triggered the expression of inflammatory and oxidative markers in rd10 retinas. Finally, an HFD caused gut microbiome dysbiosis, increasing the abundance of potentially proinflammatory bacteria. Thus, HFD feeding drives the pathological processes of retinal degeneration by promoting oxidative stress and activating inflammatory-related pathways. Our findings suggest that consumption of an HFD could accelerate the progression of the disease in patients with retinal degenerative disorders.
Collapse
|
120
|
Yau B, Naghiloo S, Diaz-Vegas A, Carr AV, Van Gerwen J, Needham EJ, Jevon D, Chen SY, Hoehn KL, Brandon AE, Macia L, Cooney GJ, Shortreed MR, Smith LM, Keller MP, Thorn P, Larance M, James DE, Humphrey SJ, Kebede MA. Proteomic pathways to metabolic disease and type 2 diabetes in the pancreatic islet. iScience 2021; 24:103099. [PMID: 34622154 PMCID: PMC8479695 DOI: 10.1016/j.isci.2021.103099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/09/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic islets are essential for maintaining physiological blood glucose levels, and declining islet function is a hallmark of type 2 diabetes. We employ mass spectrometry-based proteomics to systematically analyze islets from 9 genetic or diet-induced mouse models representing a broad cross-section of metabolic health. Quantifying the islet proteome to a depth of >11,500 proteins, this study represents the most detailed analysis of mouse islet proteins to date. Our data highlight that the majority of islet proteins are expressed in all strains and diets, but more than half of the proteins vary in expression levels, principally due to genetics. Associating these varied protein expression levels on an individual animal basis with individual phenotypic measures reveals islet mitochondrial function as a major positive indicator of metabolic health regardless of strain. This compendium of strain-specific and dietary changes to mouse islet proteomes represents a comprehensive resource for basic and translational islet cell biology.
Collapse
Affiliation(s)
- Belinda Yau
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Sheyda Naghiloo
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Alexis Diaz-Vegas
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Austin V. Carr
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Julian Van Gerwen
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Elise J. Needham
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Dillon Jevon
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Sing-Young Chen
- Department of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2052, Australia
| | - Kyle L. Hoehn
- Department of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2052, Australia
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Amanda E. Brandon
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Laurance Macia
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Gregory J. Cooney
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | | | - Lloyd M. Smith
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark P. Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Peter Thorn
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Mark Larance
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - David E. James
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Sean J. Humphrey
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Melkam A. Kebede
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| |
Collapse
|
121
|
Doridot L, Hannou SA, Krawczyk SA, Tong W, Kim MS, McElroy GS, Fowler AJ, Astapova II, Herman MA. A Systems Approach Dissociates Fructose-Induced Liver Triglyceride from Hypertriglyceridemia and Hyperinsulinemia in Male Mice. Nutrients 2021; 13:3642. [PMID: 34684643 PMCID: PMC8540719 DOI: 10.3390/nu13103642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/01/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
The metabolic syndrome (MetS), defined as the co-occurrence of disorders including obesity, dyslipidemia, insulin resistance, and hepatic steatosis, has become increasingly prevalent in the world over recent decades. Dietary and other environmental factors interacting with genetic predisposition are likely contributors to this epidemic. Among the involved dietary factors, excessive fructose consumption may be a key contributor. When fructose is consumed in large amounts, it can quickly produce many of the features of MetS both in humans and mice. The mechanisms by which fructose contributes to metabolic disease and its potential interactions with genetic factors in these processes remain uncertain. Here, we generated a small F2 genetic cohort of male mice derived from crossing fructose-sensitive and -resistant mouse strains to investigate the interrelationships between fructose-induced metabolic phenotypes and to identify hepatic transcriptional pathways that associate with these phenotypes. Our analysis indicates that the hepatic transcriptional pathways associated with fructose-induced hypertriglyceridemia and hyperinsulinemia are distinct from those that associate with fructose-mediated changes in body weight and liver triglyceride. These results suggest that multiple independent mechanisms and pathways may contribute to different aspects of fructose-induced metabolic disease.
Collapse
Affiliation(s)
- Ludivine Doridot
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (L.D.); (S.A.K.); (M.-S.K.); (G.S.M.); (A.J.F.)
| | - Sarah A. Hannou
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; (S.A.H.); (W.T.); (I.I.A.)
| | - Sarah A. Krawczyk
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (L.D.); (S.A.K.); (M.-S.K.); (G.S.M.); (A.J.F.)
| | - Wenxin Tong
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; (S.A.H.); (W.T.); (I.I.A.)
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27705, USA
| | - Mi-Sung Kim
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (L.D.); (S.A.K.); (M.-S.K.); (G.S.M.); (A.J.F.)
| | - Gregory S. McElroy
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (L.D.); (S.A.K.); (M.-S.K.); (G.S.M.); (A.J.F.)
| | - Alan J. Fowler
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (L.D.); (S.A.K.); (M.-S.K.); (G.S.M.); (A.J.F.)
| | - Inna I. Astapova
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; (S.A.H.); (W.T.); (I.I.A.)
| | - Mark A. Herman
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; (S.A.H.); (W.T.); (I.I.A.)
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27705, USA
- Division of Endocrinology and Metabolism and Nutrition, Duke University, Durham, NC 27710, USA
| |
Collapse
|
122
|
Templin AT, Schmidt C, Hogan MF, Esser N, Kitsis RN, Hull RL, Zraika S, Kahn SE. Loss of apoptosis repressor with caspase recruitment domain (ARC) worsens high fat diet-induced hyperglycemia in mice. J Endocrinol 2021; 251:125-135. [PMID: 34382577 PMCID: PMC8651217 DOI: 10.1530/joe-20-0612] [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: 07/20/2021] [Accepted: 08/11/2021] [Indexed: 11/08/2022]
Abstract
Apoptosis repressor with caspase recruitment domain (ARC) is an endogenous inhibitor of cell death signaling that is expressed in insulin-producing β cells. ARC has been shown to reduce β-cell death in response to diabetogenic stimuli in vitro, but its role in maintaining glucose homeostasis in vivo has not been fully established. Here we examined whether loss of ARC in FVB background mice exacerbates high fat diet (HFD)-induced hyperglycemia in vivo over 24 weeks. Prior to commencing 24-week HFD, ARC-/- mice had lower body weight than wild type (WT) mice. This body weight difference was maintained until the end of the study and was associated with decreased epididymal and inguinal adipose tissue mass in ARC-/- mice. Non-fasting plasma glucose was not different between ARC-/- and WT mice prior to HFD feeding, and ARC-/- mice displayed a greater increase in plasma glucose over the first 4 weeks of HFD. Plasma glucose remained elevated in ARC-/- mice after 16 weeks of HFD feeding, at which time it had returned to baseline in WT mice. Following 24 weeks of HFD, non-fasting plasma glucose in ARC-/- mice returned to baseline and was not different from WT mice. At this final time point, no differences were observed between genotypes in plasma glucose or insulin under fasted conditions or following intravenous glucose administration. However, HFD-fed ARC-/- mice exhibited significantly decreased β-cell area compared to WT mice. Thus, ARC deficiency delays, but does not prevent, metabolic adaptation to HFD feeding in mice, worsening transient HFD-induced hyperglycemia.
Collapse
Affiliation(s)
- Andrew T. Templin
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Christine Schmidt
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Meghan F. Hogan
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Nathalie Esser
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Richard N. Kitsis
- Departments of Medicine and Cell Biology and Wilf Family
Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY,
USA
| | - Rebecca L. Hull
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Sakeneh Zraika
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| | - Steven E. Kahn
- Department of Medicine, Division of Metabolism,
Endocrinology and Nutrition, Veteran Affairs Puget Sound Health Care System and
University of Washington, Seattle, WA, USA
| |
Collapse
|
123
|
The Hedgehog Signaling Pathway is Expressed in the Adult Mouse Hypothalamus and Modulated by Fasting. eNeuro 2021; 8:ENEURO.0276-21.2021. [PMID: 34535504 PMCID: PMC8482854 DOI: 10.1523/eneuro.0276-21.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/28/2021] [Accepted: 08/10/2021] [Indexed: 11/21/2022] Open
Abstract
The hedgehog signaling pathway is best known for its role in developmental patterning of the neural tube and limb bud. More recently, hedgehog signaling has been recognized for its roles in growth of adult tissues and maintenance of progenitor cell niches. However, the role of hedgehog signaling in fully differentiated cells like neurons in the adult brain is less clear. In mammals, coordination of hedgehog pathway activity relies on primary cilia and patients with ciliopathies such as Bardet–Biedl and Alström syndrome exhibit clinical features clearly attributable to errant hedgehog such as polydactyly. However, these ciliopathies also present with features not clearly associated with hedgehog signaling such as hyperphagia-associated obesity. How hedgehog signaling may contribute to feeding behavior is complex and unclear, but cilia are critical for proper energy homeostasis. Here, we provide a detailed analysis of the expression of core components of the hedgehog signaling pathway in the adult mouse hypothalamus with an emphasis on feeding centers. We show that hedgehog pathway genes continue to be expressed in differentiated neurons important for the regulation of feeding behavior. Furthermore, we demonstrate for the first time that pathway activity is regulated at the transcriptional level by fasting. These data suggest that hedgehog signaling is involved in the proper functioning of brain regions that regulate feeding behavior and that hedgehog pathway dysfunction may play a role in the obesity observed in certain ciliopathies.
Collapse
|
124
|
Kominato H, Takeda K, Mizutani K, Mikami R, Kido D, Buranasin P, Saito N, Takemura S, Nakagawa K, Nagasawa T, Iwata T. Metformin accelerates wound healing by Akt phosphorylation of gingival fibroblasts in insulin-resistant prediabetes mice. J Periodontol 2021; 93:256-268. [PMID: 34427916 DOI: 10.1002/jper.21-0362] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND This study aimed to investigate the effects of metformin on gingival wound healing in insulin-resistant prediabetes. METHODS C57BL/6J mice were fed normal diet (ND) or high-fat diet (HFD) for 10 weeks; half of the HFD mice were treated with metformin (HFD+ Met) for the last 2 weeks. Insulin and glucose tolerance tests were performed. The palatal gingiva (2.0 × 0.5 mm) was surgically removed adjacent to the maxillary molars. Post-surgical wound closure was histomorphometrically evaluated for 1 week. The mRNA expression of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS) in the tissue were quantified by real-time polymerase chain reaction. In vitro, the proliferation and migration of human gingival fibroblasts (HGFs) cultured under high-glucose or control conditions with/without metformin were analyzed. Akt phosphorylation and VEGF expression following the insulin stimulation were evaluated with/without metformin in high-glucose or control media. RESULTS HFD mice showed significantly higher plasma glucose levels and insulin resistance than ND mice. Gingival wound healing was delayed in HFD group compared with ND group but significantly improved in HFD + MET group. The decreased expression of VEGF and eNOS in HFD group was significantly elevated in the HFD + MET group. The proliferation and migration of HGFs were significantly impaired in high-glucose conditions compared with control; metformin treatment partially attenuated these effects. Metformin treatment significantly recovered the downregulated Akt phosphorylation and VEGF expression in high-glucose conditions. CONCLUSIONS Metformin improved delayed gingival wound healing in insulin-resistant prediabetes by accelerating HGFs proliferation and migration via Akt phosphorylation in insulin signaling pathway.
Collapse
Affiliation(s)
- Hiromi Kominato
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Kohei Takeda
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Koji Mizutani
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Risako Mikami
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Daisuke Kido
- Oral Diagnosis and General Dentistry, Dental Hospital, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Prima Buranasin
- Department of Conservative Dentistry and Prosthodontics, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| | - Natsumi Saito
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Shu Takemura
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Keita Nakagawa
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Toshiyuki Nagasawa
- Division of Advanced Clinical Education, Department of Integrated Dental Education, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-gun, Hokkaido, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
125
|
Systemic versus local adipokine expression differs in a combined obesity and osteoarthritis mouse model. Sci Rep 2021; 11:17001. [PMID: 34417537 PMCID: PMC8379250 DOI: 10.1038/s41598-021-96545-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/02/2021] [Indexed: 12/23/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage loss and reduced joint function. OA risk factors are age and obesity. Many adipokines are altered by obesity but also OA although systemic adipokine regulation in OA is not always clear. Therefore, metabolic effects of diet-induced obesity on OA development as well as the influence of obesity and OA progression on systemic vs. local adipokine expression in joints were compared. C57Bl/6-mice fed with HFD (high fat diet) or normal diet prior to destabilization of the medial meniscus (DMM) were sacrificed 4/6/8 weeks after surgery. Sera were evaluated for adiponectin, leptin, visfatin, cytokines. Liver grading and staging for non-alcoholic steatohepatitis (NASH) was performed and crown-like structures (CLS) in adipose tissue measured. OA progression was scored histologically. Adipokine-expressing cells and types were evaluated by immunohistochemistry. Time-dependent changes in DMM-progression were reflected by increased systemic adiponectin levels in DMM especially combined with HFD. While HFD increased serum leptin, DMM reduced systemic leptin significantly. OA scores correlated with bodyweight, leptin and hepatic scoring. Locally, increased numbers of adiponectin- and leptin-producing fibroblasts were observed in damaged menisci but visfatin was not changed. Local adipokine expression was independent from systemic levels, suggesting different mechanisms of action.
Collapse
|
126
|
Evaluation of Anti-Obesity Activity of an Herbal Formulation (F2) in DIO Mice Model and Validation of UPLC-DAD Method for Quality Control. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11167404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity is considered a chronic metabolic disorder that can be associated with multiple medical complications. Currently, there is no or limited curative therapy for obesity. This study focused on the assessment of anti-obesity activity and UPLC standardization of a polyherbal formulation (F2). An anti-obesity activity was investigated using the diet-induced obese (DIO) mice model, where obesity was developed in C57BL/6J mice by providing a high-fat diet (HFD) for five weeks without treating drugs. After the successful development of obesity, the obese mice were treated with F2 for seven weeks with continuing HFD feeding. The major obesity-related parameters such as body weight gain, food efficiency ratio, serum lipid profile, and white adipose tissue (WAT) mass were found to be significantly reduced in F2 treated obese mice. These results were supported by the down-regulation of specific adipogenic transcription factors (PPARγ, SREBP-1c, and ap2) in epididymal WAT. Histological evaluation of liver and WAT also revealed reduced fat deposition in the tissues by F2 compared to the HFD control group. The overall observations indicated that the F2 exhibited pronounced obesity-controlling activity through the inhibition of adipocyte differentiation and triglyceride accumulation in the tissues, and serum lipid depletion. In addition, F2 ameliorated obesity-induced insulin resistance. Furthermore, the UPLC-DAD method for quality control of F2 was validated and standardized using five reference compounds: astragalin, ellagic acid, fisetin, fustin, and sulfuretin.
Collapse
|
127
|
Srivastava A, Palaia T, Hall C, Stevenson M, Lee J, Ragolia L. Lipocalin-type Prostaglandin D2 Synthase appears to function as a Novel Adipokine Preventing Adipose Dysfunction in response to a High Fat Diet. Prostaglandins Other Lipid Mediat 2021; 157:106585. [PMID: 34371198 DOI: 10.1016/j.prostaglandins.2021.106585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/24/2021] [Accepted: 08/03/2021] [Indexed: 12/29/2022]
Abstract
Adipose dysfunction is the primary defect in obesity that contributes to the development of dyslipidemia, insulin resistance, cardiovascular diseases, type 2 diabetes, non-alcoholic fatty liver disease (NAFLD) and some cancers. Previously, we demonstrated the development of NAFLD in lipocalin-type prostaglandin D2 synthase (L-PGDS) knockout mice regardless of diet. In the present study, we examined the role of L-PGDS in adipose in response to a high fat diet. We observed decreased expression of L-PGDS in adipose tissue and concomitant lower plasma levels in a dietary model of obesity as well as in insulin resistant 3T3-L1 adipocytes. We show reduced adiponectin expression and phosphorylation of AMPK in white adipose tissue of L-PGDS KO mice after 14 weeks on a high fat diet as compared to control C57BL/6 mice. We also observe an increased fat content in L-PGDS KO mice as demonstrated by adipocyte hypertrophy and increased expression of lipogenenic genes. We confirmed our in vivo findings in in vitro 3T3-L1 adipocytes, using an enzymatic inhibitor of L-PGDS (AT56). Rosiglitazone treatment drastically increased L-PGDS expression in insulin resistant 3T3-L1 adipocytes and increased adiponectin expression and AMPK phosphorylation in AT56 treated 3T3-L1 adipocytes. We conclude that the absence of L-PGDS has a deleterious effect on adipose tissue functioning, which further reduces insulin sensitivity in adipose tissue. Consequently, we propose L-PGDS appears to function as a potential member of the adipokine secretome involved in the regulation of the obesity-associated metabolic syndrome.
Collapse
Affiliation(s)
- Ankita Srivastava
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States
| | - Thomas Palaia
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States; Department of Foundations of Medicine, NYU Long Island School of Medicine, 101 Mineola Blvd. Suite 4-003, Mineola, NY, 11501, United States
| | - Christopher Hall
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States
| | - Matthew Stevenson
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States
| | - Jenny Lee
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States
| | - Louis Ragolia
- Department of Biomedical research, NYU Langone Hospital, Long Island, United States; Department of Foundations of Medicine, NYU Long Island School of Medicine, 101 Mineola Blvd. Suite 4-003, Mineola, NY, 11501, United States.
| |
Collapse
|
128
|
Quiroz J, Yazdanyar A. Animal models of diabetic retinopathy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1272. [PMID: 34532409 PMCID: PMC8421981 DOI: 10.21037/atm-20-6737] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/23/2020] [Indexed: 12/16/2022]
Abstract
The retina is the posterior neuro-integrated layer of the eye that conducts impulses induced by light to the optic nerve for human vision. Diseases of the retina often leads to diminished vision and in some cases blindness. Diabetes mellitus (DM) is a worldwide public health issue and globally, there is an estimated 463 million people that are affected by DM and its consequences. Diabetic retinopathy (DR) is a blinding complication of chronic uncontrolled DM and is the most common cause of blindness in the United States between the ages 24-75. It is estimated that the global prevalence of DR will increase to 191.0 million by 2030, of those 56.3 million possessing vision-threatening diabetic retinopathy (VTDR). For the most part, current treatment modalities control the complications of DR without addressing the underlying pathophysiology of the disease. Therefore, there is an unmet need for new therapeutics that not only repair the damaged retinal tissue, but also reverse the course of DR. The key element in developing these treatments is expanding our basic knowledge by studying DR pathogenesis in animal models of proliferative and non-proliferative DR (PDR and NPDR). There are numerous models available for the research of both PDR and NPDR with substantial overlap. Animal models available include those with genetic backgrounds prone to hyperglycemic states, immunologic etiologies, or environmentally induced disease. In this review we aimed to comprehensively summarize the available animal models for DR while also providing insight to each model's ocular therapeutic potential for drug discovery.
Collapse
Affiliation(s)
- Jose Quiroz
- Medical Scientist Training Program, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Amirfarbod Yazdanyar
- Department of Ophthalmology and Visual Sciences, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
129
|
Barakat M, DiPietro LA, Chen L. Limited Treatment Options for Diabetic Wounds: Barriers to Clinical Translation Despite Therapeutic Success in Murine Models. Adv Wound Care (New Rochelle) 2021; 10:436-460. [PMID: 33050829 PMCID: PMC8236303 DOI: 10.1089/wound.2020.1254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/12/2020] [Indexed: 12/15/2022] Open
Abstract
Significance: Millions of people worldwide suffer from diabetes mellitus and its complications, including chronic diabetic wounds. To date, there are few widely successful clinical therapies specific to diabetic wounds beyond general wound care, despite the vast number of scientific discoveries in the pathogenesis of defective healing in diabetes. Recent Advances: In recent years, murine animal models of diabetes have enabled the investigation of many possible therapeutics for diabetic wound care. These include specific cell types, growth factors, cytokines, peptides, small molecules, plant extracts, microRNAs, extracellular vesicles, novel wound dressings, mechanical interventions, bioengineered materials, and more. Critical Issues: Despite many research discoveries, few have been translated from their success in murine models to clinical use in humans. This massive gap between bench discovery and bedside application begs the simple and critical question: what is still missing? The complexity and multiplicity of the diabetic wound makes it an immensely challenging therapeutic target, and this lopsided progress highlights the need for new methods to overcome the bench-to-bedside barrier. How can laboratory discoveries in animal models be effectively translated to novel clinical therapies for human patients? Future Directions: As research continues to decipher deficient healing in diabetes, new approaches and considerations are required to ensure that these discoveries can become translational, clinically usable therapies. Clinical progress requires the development of new, more accurate models of the human disease state, multifaceted investigations that address multiple critical components in wound repair, and more innovative research strategies that harness both the existing knowledge and the potential of new advances across disciplines.
Collapse
Affiliation(s)
- May Barakat
- Center for Wound Repair and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Luisa A. DiPietro
- Center for Wound Repair and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Lin Chen
- Center for Wound Repair and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
130
|
Garcia DI, Hurst KE, Bradshaw A, Janakiraman H, Wang C, Camp ER. High-Fat Diet Drives an Aggressive Pancreatic Cancer Phenotype. J Surg Res 2021; 264:163-172. [PMID: 33838401 DOI: 10.1016/j.jss.2020.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/31/2020] [Accepted: 10/13/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Emerging evidence indicates associations between high-fat diet (HFD), metabolic syndrome (MetS), and increased risk of pancreatic cancer. However, individual components of an HFD that increase cancer risk have not been isolated. In addition, a specific pattern of cytokine elevation by which MetS drives pancreatic tumor progression is not well described. We hypothesized that oleic acid (OA), a major component of HFD, would augment pancreatic neoplastic processes. METHODS An orthotopic pancreatic cancer model with Panc02 cells was used to compare the effect of low-fat diet to OA-based HFD on cancer progression. Tumors were quantitated, analyzed by immunohistochemistry. In addition, serum cytokine levels were quantitated. Proliferation, migration assays, and expression of epithelial-to-mesenchymal transition factors were evaluated on Panc02 and MiaPaCa-2 pancreatic cancer cells cultured in high concentrations of OA. RESULTS HFD tumor-bearing mice (n = 8) had an 18% weight increase (P < 0.001) and increased tumor burden (P < 0.05) compared with the low-fat diet tumor-bearing group (n = 6). HFD tumors had significantly increased angiogenesis (P < 0.001) and decreased apoptosis (P < 0.05). Serum of HFD mice demonstrated increased levels of glucagon and glucagon-like peptide-1. Two pancreatic cancer cell lines cultured in OA demonstrated significant increases in proliferation (P < 0.001) and a >2.5-fold increase in cell migration (P < 0.001) when treated with OA. Panc02 treated with OA had increased expression of epithelial-to-mesenchymal transition factors SNAI-1 (Snail) and Zeb-1(P < 0.01). CONCLUSIONS High-fat conditions in vitro and in vivo resulted in an aggressive pancreatic cancer phenotype. Our data support further investigations elucidating molecular pathways augmented by MetS conditions to identify novel therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Denise I Garcia
- Department of Surgery, Charleston, South Carolina; Department of Hollings Cancer Center, Charleston, South Carolina
| | - Katie E Hurst
- Department of Surgery, Charleston, South Carolina; Department of Hollings Cancer Center, Charleston, South Carolina
| | - Alexandra Bradshaw
- Department of Surgery, Charleston, South Carolina; Department of Hollings Cancer Center, Charleston, South Carolina
| | - Harinarayanan Janakiraman
- Department of Surgery, Charleston, South Carolina; Department of Hollings Cancer Center, Charleston, South Carolina
| | - Cindy Wang
- Department of Surgery, Charleston, South Carolina; Department of Hollings Cancer Center, Charleston, South Carolina
| | - E Ramsay Camp
- Department of Surgery, Charleston, South Carolina; Department of Hollings Cancer Center, Charleston, South Carolina; Department of Ralph H. Johnson VA Medical Center, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
131
|
Nash Y, Ganoth A, Borenstein-Auerbach N, Levy-Barazany H, Goldsmith G, Kopelevich A, Pozyuchenko K, Sakhneny L, Lazdon E, Blanga-Kanfi S, Alhadeff R, Benromano T, Landsman L, Tsfadia Y, Frenkel D. From virus to diabetes therapy: Characterization of a specific insulin-degrading enzyme inhibitor for diabetes treatment. FASEB J 2021; 35:e21374. [PMID: 33835493 DOI: 10.1096/fj.201901945r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022]
Abstract
Inhibition of insulin-degrading enzyme (IDE) is a possible target for treating diabetes. However, it has not yet evolved into a medical intervention, mainly because most developed inhibitors target the zinc in IDE's catalytic site, potentially causing toxicity to other essential metalloproteases. Since IDE is a cellular receptor for the varicella-zoster virus (VZV), we constructed a VZV-based inhibitor. We computationally characterized its interaction site with IDE showing that the peptide specifically binds inside IDE's central cavity, however, not in close proximity to the zinc ion. We confirmed the peptide's effective inhibition on IDE activity in vitro and showed its efficacy in ameliorating insulin-related defects in types 1 and 2 diabetes mouse models. In addition, we suggest that inhibition of IDE may ameliorate the pro-inflammatory profile of CD4+ T-cells toward insulin. Together, we propose a potential role of a designed VZV-derived peptide to serve as a selectively-targeted and as an efficient diabetes therapy.
Collapse
Affiliation(s)
- Yuval Nash
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Assaf Ganoth
- The Interdisciplinary Center (IDC), Herzliya, Israel.,Department of Physical Therapy, School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nofit Borenstein-Auerbach
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Hilit Levy-Barazany
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Guy Goldsmith
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adi Kopelevich
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Katia Pozyuchenko
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Lina Sakhneny
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ekaterina Lazdon
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shani Blanga-Kanfi
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Raphael Alhadeff
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Tali Benromano
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Limor Landsman
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yossi Tsfadia
- Department of Biochemistry and Molecular Biology, School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
132
|
Lactobacillus plantarum Reduces Low-Grade Inflammation and Glucose Levels in a Mouse Model of Chronic Stress and Diabetes. Infect Immun 2021; 89:e0061520. [PMID: 34001561 DOI: 10.1128/iai.00615-20] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study aimed to examine the effects of Lactobacillus plantarum, a lactic acid bacteria strain isolated from kimchi, on the development of low-grade inflammation and type 2 diabetes mellitus (T2DM) exacerbated by chronic stress. C57BL/6 mice were fed either a high-fat diet (HFD) and randomized into an HFD group or a group that was fed an HFD and subjected to chronic cold exposure-related stress (HFDS), or mice were fed a normal diet (ND) and randomized into an ND group or a group that was fed an ND and subjected to chronic cold exposure-related stress (NDS). Lactobacillus plantarum LRCC5310 (108, 1010 CFU) and LRCC5314 (108, 1010 CFU) as well as L. gasseri BNR17 (108 CFU), as a positive control, were administered orally twice every day to all the mice for 12 weeks. The expression of Glut4 and adiponectin, main glucose transporter-related genes, was upregulated in the LRCC5310- and LRCC5314-treated groups. Levels of serum proinflammatory cytokines (tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6]) and of mRNAs of proinflammatory genes (Tnf-α, Il-6, Ccl2, leptin) were elevated in HFDS mice. The expression of proinflammatory genes was downregulated in LRCC5310- and LRCC5314-treated groups; this was not the case for Tnf-α expression in HFDS mice. Levels of serum corticosterone and mRNA levels of stress-related genes (Npy, Y2r) were decreased in lactic acid bacteria (LAB)-fed groups, with only LRCC5314 downregulating Npy expression in HFDS mice. These results suggest that the LAB strains can normalize the expression of metabolic genes, inhibit inflammatory responses, and suppress stress in HFDS mice.
Collapse
|
133
|
Sharma S. High fat diet and its effects on cognitive health: alterations of neuronal and vascular components of brain. Physiol Behav 2021; 240:113528. [PMID: 34260890 DOI: 10.1016/j.physbeh.2021.113528] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 01/01/2023]
Abstract
It has been well recognized that intake of diets rich in saturated fats could result in development of metabolic disorders such as type 2 diabetes mellitus, obesity and cardiovascular diseases. Recent studies have suggested that intake of high fat diet (HFD) is also associated with cognitive dysfunction. Various preclinical studies have demonstrated the impact of short and long term HFD feeding on the biochemical and behavioural alterations. This review summarizes studies and the protocols used to assess the impacts of HFD feeding on cognitive performance in rodents. Further, it discuss the key mechanisms that are altered by HFD feeding, such as, insulin resistance, oxidative stress, neuro-inflammation, transcriptional dysregulation and loss of synaptic plasticity. Along with these, HFD feeding also alters the vascular components of brain such as loss of BBB integrity and reduced cerebral blood flow. It is highly possible that these factors are responsible for the development of cognitive deficits as a result of HFD feeding.
Collapse
Affiliation(s)
- Sorabh Sharma
- Division of Medical Sciences, University of Victoria, PO Box 1700 STN CSC, Victoria, BC, V8W2Y2, Canada.
| |
Collapse
|
134
|
Federico M, De la Fuente S, Palomeque J, Sheu SS. The role of mitochondria in metabolic disease: a special emphasis on heart dysfunction. J Physiol 2021; 599:3477-3493. [PMID: 33932959 PMCID: PMC8424986 DOI: 10.1113/jp279376] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/18/2021] [Indexed: 01/10/2023] Open
Abstract
Metabolic diseases (MetDs) embrace a series of pathologies characterized by abnormal body glucose usage. The known diseases included in this group are metabolic syndrome, prediabetes and diabetes mellitus types 1 and 2. All of them are chronic pathologies that present metabolic disturbances and are classified as multi-organ diseases. Cardiomyopathy has been extensively described in diabetic patients without overt macrovascular complications. The heart is severely damaged during the progression of the disease; in fact, diabetic cardiomyopathies are the main cause of death in MetDs. Insulin resistance, hyperglycaemia and increased free fatty acid metabolism promote cardiac damage through mitochondria. These organelles supply most of the energy that the heart needs to beat and to control essential cellular functions, including Ca2+ signalling modulation, reactive oxygen species production and apoptotic cell death regulation. Several aspects of common mitochondrial functions have been described as being altered in diabetic cardiomyopathies, including impaired energy metabolism, compromised mitochondrial dynamics, deficiencies in Ca2+ handling, increases in reactive oxygen species production, and a higher probability of mitochondrial permeability transition pore opening. Therefore, the mitochondrial role in MetD-mediated heart dysfunction has been studied extensively to identify potential therapeutic targets for improving cardiac performance. Herein we review the cardiac pathology in metabolic syndrome, prediabetes and diabetes mellitus, focusing on the role of mitochondrial dysfunctions.
Collapse
Affiliation(s)
- Marilen Federico
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Medicas, UNLP, La Plata, Argentina
| | - Sergio De la Fuente
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Medicas, UNLP, La Plata, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, CABA, Argentina
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| |
Collapse
|
135
|
Estrada-Meza J, Videlo J, Bron C, Saint-Béat C, Silva M, Duboeuf F, Peyruchaud O, Rajas F, Mithieux G, Gautier-Stein A. Tamoxifen Treatment in the Neonatal Period Affects Glucose Homeostasis in Adult Mice in a Sex-Dependent Manner. Endocrinology 2021; 162:6277101. [PMID: 33999998 DOI: 10.1210/endocr/bqab098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Indexed: 12/11/2022]
Abstract
Tamoxifen is a selective estrogen receptor modulator used to activate the CREERT2 recombinase, allowing tissue-specific and temporal control of the somatic mutagenesis to generate transgenic mice. Studies integrating development and metabolism require a genetic modification induced by a neonatal tamoxifen administration. Here, we investigate the effects of a neonatal tamoxifen administration on energy homeostasis in adult male and female C57BL/6J mice. C57BL/6J male and female mouse pups received a single injection of tamoxifen 1 day after birth (NTT) and were fed a high-fat/high-sucrose diet at 6 weeks of age. We measured weight, body composition, glucose and insulin tolerance, basal metabolism, and tibia length and weight in adult mice. The neonatal tamoxifen administration exerted long-term, sex-dependent effects on energy homeostasis. NTT female mice became overweight and developed impaired glucose control in comparison to vehicle-treated littermates. NTT females exhibited 60% increased fat mass, increased food intake, decreased physical activity and energy expenditure, impaired glucose and insulin tolerance, and fasting hyperglycemia and hyperinsulinemia. In contrast, NTT male mice exhibited a modest amelioration of glucose and insulin tolerance and long-term decreased lean mass linked to decreased bone weight. These results suggest that the neonatal tamoxifen administration exerted a marked and sex-dependent influence on adult energy homeostasis and bone weight and must therefore be used with caution for the development of transgenic mouse models regarding studies on energy homeostasis and bone biology.
Collapse
Affiliation(s)
- Judith Estrada-Meza
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Jasmine Videlo
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Clara Bron
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Cécile Saint-Béat
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Marine Silva
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - François Duboeuf
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1033, Lyon, France
| | - Olivier Peyruchaud
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1033, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | | |
Collapse
|
136
|
The Use of Natural Compounds as a Strategy to Counteract Oxidative Stress in Animal Models of Diabetes Mellitus. Int J Mol Sci 2021; 22:ijms22137009. [PMID: 34209800 PMCID: PMC8268811 DOI: 10.3390/ijms22137009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease characterised by insulin deficiency, resulting in hyperglycaemia, a characteristic symptom of type 2 diabetes mellitus (DM2). DM substantially affects numerous metabolic pathways, resulting in β-cell dysfunction, insulin resistance, abnormal blood glucose levels, impaired lipid metabolism, inflammatory processes, and excessive oxidative stress. Oxidative stress can affect the body’s normal physiological function and cause numerous cellular and molecular changes, such as mitochondrial dysfunction. Animal models are useful for exploring the cellular and molecular mechanisms of DM and improving novel therapeutics for their safe use in human beings. Due to their health benefits, there is significant interest in a wide range of natural compounds that can act as naturally occurring anti-diabetic compounds. Due to rodent models’ relatively similar physiology to humans and ease of handling and housing, they are widely used as pre-clinical models for studying several metabolic disorders. In this review, we analyse the currently available rodent animal models of DM and their advantages and disadvantages and highlight the potential anti-oxidative effects of natural compounds and their mechanisms of action.
Collapse
|
137
|
Bruce CR, Hamley S, Ang T, Howlett KF, Shaw CS, Kowalski GM. Translating glucose tolerance data from mice to humans: Insights from stable isotope labelled glucose tolerance tests. Mol Metab 2021; 53:101281. [PMID: 34175474 PMCID: PMC8313600 DOI: 10.1016/j.molmet.2021.101281] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 11/29/2022] Open
Abstract
Objective The glucose tolerance test (GTT) is widely used in human and animal biomedical and pharmaceutical research. Despite its prevalent use, particularly in mouse metabolic phenotyping, to the best of our knowledge we are not aware of any studies that have attempted to qualitatively compare the metabolic events during a GTT in mice with those performed in humans. Methods Stable isotope labelled oral glucose tolerance tests (siOGTTs; [6,6-2H2]glucose) were performed in both human and mouse cohorts to provide greater resolution into postprandial glucose kinetics. The siOGTT allows for the partitioning of circulating glucose into that derived from exogenous and endogenous sources. Young adults spanning the spectrum of normal glucose tolerance (n = 221), impaired fasting (n = 14), and impaired glucose tolerance (n = 19) underwent a 75g siOGTT, whereas a 50 mg siOGTT was performed on chow (n = 43) and high-fat high-sucrose fed C57Bl6 male mice (n = 46). Results During the siOGTT in humans, there is a long period (>3hr) of glucose absorption and, accordingly, a large, sustained insulin response and robust suppression of lipolysis and endogenous glucose production (EGP), even in the presence of glucose intolerance. In contrast, mice appear to be highly reliant on glucose effectiveness to clear exogenous glucose and experience only modest, transient insulin responses with little, if any, suppression of EGP. In addition to the impaired stimulation of glucose uptake, mice with the worst glucose tolerance appear to have a paradoxical and persistent rise in EGP during the OGTT, likely related to handling stress. Conclusions The metabolic response to the OGTT in mice and humans is highly divergent. The potential reasons for these differences and their impact on the interpretation of mouse glucose tolerance data and their translation to humans are discussed. We compared the mechanisms governing glucose handling in humans and mice. Humans and mice underwent stable isotope labelled oral glucose tolerance tests. Metabolic responses between humans and mice were highly divergent. Unlike humans, most mice exhibit little EGP suppression or insulin response.
Collapse
Affiliation(s)
- Clinton R Bruce
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Waurn Ponds, Victoria, 3216, Australia
| | - Steven Hamley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Waurn Ponds, Victoria, 3216, Australia
| | - Teddy Ang
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Waurn Ponds, Victoria, 3216, Australia
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Waurn Ponds, Victoria, 3216, Australia
| | - Christopher S Shaw
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Waurn Ponds, Victoria, 3216, Australia
| | - Greg M Kowalski
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Waurn Ponds, Victoria, 3216, Australia; Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Waurn Ponds, Victoria, 3216, Australia.
| |
Collapse
|
138
|
Shin CH, Kim KH, Jeeva S, Kang SM. Towards Goals to Refine Prophylactic and Therapeutic Strategies Against COVID-19 Linked to Aging and Metabolic Syndrome. Cells 2021; 10:1412. [PMID: 34204163 PMCID: PMC8227274 DOI: 10.3390/cells10061412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) gave rise to the coronavirus disease 2019 (COVID-19) pandemic. A strong correlation has been demonstrated between worse COVID-19 outcomes, aging, and metabolic syndrome (MetS), which is primarily derived from obesity-induced systemic chronic low-grade inflammation with numerous complications, including type 2 diabetes mellitus (T2DM). The majority of COVID-19 deaths occurs in people over the age of 65. Individuals with MetS are inclined to manifest adverse disease consequences and mortality from COVID-19. In this review, we examine the prevalence and molecular mechanisms underlying enhanced risk of COVID-19 in elderly people and individuals with MetS. Subsequently, we discuss current progresses in treating COVID-19, including the development of new COVID-19 vaccines and antivirals, towards goals to elaborate prophylactic and therapeutic treatment options in this vulnerable population.
Collapse
Affiliation(s)
- Chong-Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (S.J.)
| | | | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (S.J.)
| |
Collapse
|
139
|
Ross DS, Yeh TH, King S, Mathers J, Rybchyn MS, Neist E, Cameron M, Tacey A, Girgis CM, Levinger I, Mason RS, Brennan-Speranza TC. Distinct Effects of a High Fat Diet on Bone in Skeletally Mature and Developing Male C57BL/6J Mice. Nutrients 2021; 13:nu13051666. [PMID: 34068953 PMCID: PMC8157111 DOI: 10.3390/nu13051666] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 01/01/2023] Open
Abstract
Increased risks of skeletal fractures are common in patients with impaired glucose handling and type 2 diabetes mellitus (T2DM). The pathogenesis of skeletal fragility in these patients remains ill-defined as patients present with normal to high bone mineral density. With increasing cases of glucose intolerance and T2DM it is imperative that we develop an accurate rodent model for further investigation. We hypothesized that a high fat diet (60%) administered to developing male C57BL/6J mice that had not reached skeletal maturity would over represent bone microarchitectural implications, and that skeletally mature mice would better represent adult-onset glucose intolerance and the pre-diabetes phenotype. Two groups of developing (8 week) and mature (12 week) male C57BL/6J mice were placed onto either a normal chow (NC) or high fat diet (HFD) for 10 weeks. Oral glucose tolerance tests were performed throughout the study period. Long bones were excised and analysed for ex vivo biomechanical testing, micro-computed tomography, 2D histomorphometry and gene/protein expression analyses. The HFD increased fasting blood glucose and significantly reduced glucose tolerance in both age groups by week 7 of the diets. The HFD reduced biomechanical strength, both cortical and trabecular indices in the developing mice, but only affected cortical outcomes in the mature mice. Similar results were reflected in the 2D histomorphometry. Tibial gene expression revealed decreased bone formation in the HFD mice of both age groups, i.e., decreased osteocalcin expression and increased sclerostin RNA expression. In the mature mice only, while the HFD led to a non-significant reduction in runt-related transcription factor 2 (Runx2) RNA expression, this decrease became significant at the protein level in the femora. Our mature HFD mouse model more accurately represents late-onset impaired glucose tolerance/pre-T2DM cases in humans and can be used to uncover potential insights into reduced bone formation as a mechanism of skeletal fragility in these patients.
Collapse
Affiliation(s)
- Dean S. Ross
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
| | - Tzu-Hsuan Yeh
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
| | - Shalinie King
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
- Faculty of Medicine and Health, School of Dentistry, University of Sydney, Sydney 2006, Australia
| | - Julia Mathers
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
| | - Mark S. Rybchyn
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
| | - Elysia Neist
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
| | - Melissa Cameron
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
| | - Alexander Tacey
- Institute for Health and Sport (IHES), Victoria University, Melbourne 3011, Australia; (A.T.); (I.L.)
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans 3021, Australia
| | - Christian M. Girgis
- Department of Diabetes and Endocrinology, Westmead Hospital, Sydney 2145, Australia;
- Department of Endocrinology, Royal North Shore Hospital, Sydney 2065, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Melbourne 3011, Australia; (A.T.); (I.L.)
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans 3021, Australia
| | - Rebecca S. Mason
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
| | - Tara C. Brennan-Speranza
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney 2006, Australia; (D.S.R.); (T.-H.Y.); (S.K.); (J.M.); (M.S.R.); (E.N.); (M.C.); (R.S.M.)
- Faculty of Medicine and Health, School of Public Health, University of Sydney, Sydney 2006, Australia
- Correspondence: ; Tel.: +61-2-9351-4099
| |
Collapse
|
140
|
Cugno C, Kizhakayil D, Calzone R, Rahman SM, Halade GV, Rahman MM. Omega-3 fatty acid-rich fish oil supplementation prevents rosiglitazone-induced osteopenia in aging C57BL/6 mice and in vitro studies. Sci Rep 2021; 11:10364. [PMID: 33990655 PMCID: PMC8121944 DOI: 10.1038/s41598-021-89827-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Rosiglitazone is an effective insulin-sensitizer, however associated with bone loss mainly due to increased bone resorption and bone marrow adiposity. We investigated the effect of the co-administration of fish oil rich in omega-3 fatty acids (FAs) on rosiglitazone-induced bone loss in C57BL/6 mice and the mechanisms underlying potential preventive effect. Mice fed the iso-caloric diet supplemented with fish oil exhibited significantly higher levels of bone density in different regions compared to the other groups. In the same cohort of mice, reduced activity of COX-2, enhanced activity of alkaline phosphatase, lower levels of cathepsin k, PPAR-γ, and pro-inflammatory cytokines, and a higher level of anti-inflammatory cytokines were observed. Moreover, fish oil restored rosiglitazone-induced down-regulation of osteoblast differentiation and up-regulation of adipocyte differentiation in C3H10T1/2 cells and inhibited the up-regulation of osteoclast differentiation of RANKL-treated RAW264.7 cells. We finally tested our hypothesis on human Mesenchymal Stromal Cells differentiated to osteocytes and adipocytes confirming the beneficial effect of docosahexaenoic acid (DHA) omega-3 FA during treatment with rosiglitazone, through the down-regulation of adipogenic genes, such as adipsin and FABP4 along the PPARγ/FABP4 axis, and reducing the capability of osteocytes to switch toward adipogenesis. Fish oil may prevent rosiglitazone-induced bone loss by inhibiting inflammation, osteoclastogenesis, and adipogenesis and by enhancing osteogenesis in the bone microenvironment.
Collapse
Affiliation(s)
- Chiara Cugno
- Advanced Cell Therapy Core, Sidra Medicine, Doha, Qatar
| | | | - Rita Calzone
- Advanced Cell Therapy Core, Sidra Medicine, Doha, Qatar
| | - Shaikh Mizanoor Rahman
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, The University of South Florida Health, Tampa, FL, USA
| | - Md M Rahman
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar.
| |
Collapse
|
141
|
Siino V, Jensen P, James P, Vasto S, Amato A, Mulè F, Accardi G, Larsen MR. Obesogenic Diets Cause Alterations on Proteins and Theirs Post-Translational Modifications in Mouse Brains. Nutr Metab Insights 2021; 14:11786388211012405. [PMID: 34017182 PMCID: PMC8114309 DOI: 10.1177/11786388211012405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/13/2021] [Indexed: 11/17/2022] Open
Abstract
Obesity constitutes a major global health threat and is associated with a variety of diseases ranging from metabolic and cardiovascular disease, cancer to neurodegeneration. The hallmarks of neurodegeneration include oxidative stress, proteasome impairment, mitochondrial dysfunction and accumulation of abnormal protein aggregates as well as metabolic alterations. As an example, in post-mortem brain of patients with Alzheimer’s disease (AD), several studies have reported reduction of insulin, insulin-like growth factor 1 and insulin receptor and an increase in tau protein and glycogen-synthase kinase-3β compared to healthy controls suggesting an impairment of metabolism in the AD patient’s brain. Given these lines of evidence, in the present study we investigated brains of mice treated with 2 obesogenic diets, high-fat diet (HFD) and high-glycaemic diet (HGD), compared to mice fed with a standard diet (SD) employing a quantitative mass spectrometry-based approach. Moreover, post-translational modified proteins (phosphorylated and N-linked glycosylated) were studied. The aim of the study was to identify proteins present in the brain that are changing their expression based on the diet given to the mice. We believed that some of these changes would highlight pathways and molecular mechanisms that could link obesity to brain impairment. The results showed in this study suggest that, together with cytoskeletal proteins, mitochondria and metabolic proteins are changing their post-translational status in brains of obese mice. Specifically, proteins involved in metabolic pathways and in mitochondrial functions are mainly downregulated in mice fed with obesogenic diets compared to SD. These changes suggest a reduced metabolism and a lower activity of mitochondria in obese mice. Some of these proteins, such as PGM1 and MCT1 have been shown to be involved in brain impairment as well. These results might shed light on the well-studied correlation between obesity and brain damage. The results presented here are in agreement with previous findings and aim to open new perspectives on the connection between diet-induced obesity and brain impairment.
Collapse
Affiliation(s)
| | - Pia Jensen
- Department of Biochemistry and Molecular Biology, PR Group, University of Southern Denmark, Odense, Denmark
| | - Peter James
- Department of Immunotechnology, Lund University, Sweden.,Turku Centre for Biotechnology and Åbo Academy University, Turku, Finland
| | - Sonya Vasto
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy.,Institute of Biomedicine and Molecular Immunology 'Alberto Monroy' CNR, Palermo, Italy
| | - Antonella Amato
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Flavia Mulè
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Giulia Accardi
- Department of Biopathology and Medical biotechnologies Pathobiology and Medical Biotechnologies, University of Palermo, Palermo, Italy
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, PR Group, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
142
|
Hitchen B, Norwood K, Gault VA, Leslie JC. Behavioural evaluation of mouse models of type 2 diabetes. LEARNING AND MOTIVATION 2021. [DOI: 10.1016/j.lmot.2021.101730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
143
|
Buckley TN, Omotola O, Archer LA, Rostron CR, Kamineni EP, Llanora JD, Chalfant JM, Lei F, Slade E, Pendergast JS. High-fat feeding disrupts daily eating behavior rhythms in obesity-prone but not in obesity-resistant male inbred mouse strains. Am J Physiol Regul Integr Comp Physiol 2021; 320:R619-R629. [PMID: 33626995 PMCID: PMC8163612 DOI: 10.1152/ajpregu.00150.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/18/2021] [Accepted: 02/21/2021] [Indexed: 12/16/2022]
Abstract
Abnormal meal timing, like skipping breakfast and late-night snacking, is associated with obesity in humans. Disruption of daily eating rhythms also contributes to obesity in mice. When fed a high-fat diet, male C57BL/6J mice have disrupted eating behavior rhythms and they become obese. In contrast to obesity-prone C57BL/6J mice, some inbred strains of mice are resistant to high-fat diet-induced obesity. In this study, we sought to determine whether there are distinct effects of high-fat feeding on daily eating behavior rhythms in obesity-prone and obesity-resistant male mice. Male obesity-prone (C57BL/6J and 129X1/SvJ) and obesity-resistant (SWR/J and BALB/cJ) mice were fed low-fat diet or high-fat diet for 6 wk. Consistent with previous studies, obesity-prone male mice gained more weight and adiposity during high-fat diet feeding than obesity-resistant male mice. The amplitude of the daily rhythm of eating behavior was markedly attenuated in male obesity-prone mice fed high-fat diet, but not in obesity-resistant males. In contrast, high-fat feeding did not differentially affect locomotor activity rhythms in obesity-prone and obesity-resistant male mice. Together, these data suggest that regulation of the daily rhythm of eating may underlie the propensity to develop diet-induced obesity in male mice.
Collapse
Affiliation(s)
| | | | - Luke A Archer
- Department of Biology, University of Kentucky, Lexington, Kentucky
| | | | - Ellora P Kamineni
- Department of Biology, University of Kentucky, Lexington, Kentucky
- Math Science Technology Center, Paul Laurence Dunbar High School, Lexington, Kentucky
| | - Josie D Llanora
- Department of Biology, University of Kentucky, Lexington, Kentucky
| | | | - Feitong Lei
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky
| | - Emily Slade
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky
| | - Julie S Pendergast
- Department of Biology, University of Kentucky, Lexington, Kentucky
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, Kentucky
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
144
|
Gulnaz A, Nadeem J, Han JH, Lew LC, Son JD, Park YH, Rather IA, Hor YY. Lactobacillus Sps in Reducing the Risk of Diabetes in High-Fat Diet-Induced Diabetic Mice by Modulating the Gut Microbiome and Inhibiting Key Digestive Enzymes Associated with Diabetes. BIOLOGY 2021; 10:biology10040348. [PMID: 33924088 PMCID: PMC8074288 DOI: 10.3390/biology10040348] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/08/2021] [Accepted: 04/17/2021] [Indexed: 12/31/2022]
Abstract
Simple Summary Type 2 diabetes (T2D) is increasingly spreading across the globe. The disease is linked to a disruption of gut microbiome. Probiotics are essential gut microbiota modulators proven to restore microbiota changes, thereby conferring health to its host. This study aimed to use probiotics (lactobacilli) and their metabolites as natural anti-diabetic therapy through the modulation of gut microbiota and inhibit diabetes-causing enzymes. Lactobacillus-treated high-fat diet mice showed lower blood glucose levels and body weight. Interestingly, our study also proved that the lactobacilli altered gut microbiota composition by suppressing opportunistic bacteria that are highly associated with metabolic diseases. Our findings substantiate the use of probiotics as natural anti-diabetic therapeutics. Abstract Obesity caused by a high-fat diet (HFD) affects gut microbiota linked to the risk of type-2 diabetes (T2D). This study evaluates live cells and ethanolic extract (SEL) of Lactobacillus sakei Probio65 and Lactobacillus plantarum Probio-093 as natural anti-diabetic compounds. In-vitro anti-diabetic effects were determined based on the inhibition of α-glucosidase and α-amylase enzymes. The SEL of Probio65 and Probio-093 significantly retarded α-glucosidase and α-amylase enzymes (p < 0.05). Live Probio65 and Probio-093 inhibited α-glucosidase and α-amylase, respectively (p < 0.05). In mice fed with a 45% kcal high-fat diet (HFD), the SEL and live cells of both strains reduced body weight significantly compared to HFD control (p < 0.05). Probio-093 also improved blood glucose level compared to control (p < 0.05). The gut microbiota modulatory effects of lactobacilli on HFD-induced diabetic mice were analyzed with qPCR method. The SEL and live cells of both strains reduced phyla Deferribacteres compared to HFD control (p < 0.05). The SEL and live cells of Probio-093 promoted more Actinobacteria (phyla), Bifidobacterium, and Prevotella (genus) compared to control (p < 0.05). Both strains exerted metabolic-modulatory effects, with strain Probio-093 showing more prominent alteration in gut microbiota, substantiating the role of probiotics in gut microbiome modulations and anti-diabetic effect. Both lactobacilli are potential candidates to lessen obesity-linked T2D.
Collapse
Affiliation(s)
- Aneela Gulnaz
- Department of Biotechnology, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Korea; (A.G.); (J.N.); (J.-H.H.); (Y.-H.P.)
| | - Jawad Nadeem
- Department of Biotechnology, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Korea; (A.G.); (J.N.); (J.-H.H.); (Y.-H.P.)
| | - Jong-Hun Han
- Department of Biotechnology, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Korea; (A.G.); (J.N.); (J.-H.H.); (Y.-H.P.)
| | - Lee-Ching Lew
- Probionic Corp. Jeonbuk Institute for Food-Bioindustry, 111-18, Wonjangdong-gil, Deokjin-gu, Jeonju-si, Jeollabuk-do 38541, Korea;
| | - Jae-Dong Son
- Department of Veterinary Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju-si, Gyeongsangnam-do 52828, Korea;
| | - Yong-Ha Park
- Department of Biotechnology, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Korea; (A.G.); (J.N.); (J.-H.H.); (Y.-H.P.)
- Probionic Corp. Jeonbuk Institute for Food-Bioindustry, 111-18, Wonjangdong-gil, Deokjin-gu, Jeonju-si, Jeollabuk-do 38541, Korea;
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: or (I.A.R.); (Y.-Y.H.)
| | - Yan-Yan Hor
- Department of Biotechnology, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Korea; (A.G.); (J.N.); (J.-H.H.); (Y.-H.P.)
- Correspondence: or (I.A.R.); (Y.-Y.H.)
| |
Collapse
|
145
|
RYGB Is More Effective than VSG at Protecting Mice from Prolonged High-Fat Diet Exposure: An Occasion to Roll Up Our Sleeves? Obes Surg 2021; 31:3227-3241. [PMID: 33856636 DOI: 10.1007/s11695-021-05389-8] [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: 12/24/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Understanding the effects of Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) on adipose tissue physiology is important for the treatment of obesity-related metabolic disorders. By using robust mouse models of bariatric surgery that closely resemble those performed in humans, we can compare the effects of RYGB and VSG on adipose physiology in the absence of post-operative confounds such as diet and lifestyle changes. MATERIALS AND METHODS RYGB and VSG were compared using a diet-induced mouse model of obesity. High-fat diet (HFD) was administered post-operatively and changes to white and brown adipose tissue were evaluated, along with alterations to weight, glucose homeostasis, dyslipidemia, and insulin sensitivity. RESULTS After prolonged exposure to high-fat diet post-operatively, RYGB was effective in achieving sustained weight loss, while VSG unexpectedly accelerated weight gain rates. The resolution of obesity-related comorbidities such as glucose and insulin intolerance, dyslipidemia, and insulin sensitivity was improved after RYGB, but not for VSG. In RYGB, there were improvements to the function and health of white adipose tissue, enhanced brown adipose metabolism, and the browning of subcutaneous white adipose tissue, with no comparable changes seen for these factors after VSG. Some markers of systemic inflammation improved after both RYGB and VSG. CONCLUSION There are significantly different effects between RYGB and VSG when HFD is administered post-operatively and robust mouse models of bariatric surgery are used. RYGB resulted in lasting physiological and metabolic changes but VSG showed little difference from that of its sham-operated, DIO counterpart.
Collapse
|
146
|
Salehpour A, Rezaei M, Khoradmehr A, Tahamtani Y, Tamadon A. Which Hyperglycemic Model of Zebrafish ( Danio rerio) Suites My Type 2 Diabetes Mellitus Research? A Scoring System for Available Methods. Front Cell Dev Biol 2021; 9:652061. [PMID: 33791308 PMCID: PMC8005598 DOI: 10.3389/fcell.2021.652061] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
Despite extensive studies on type 2 diabetes mellitus (T2DM), there is no definitive cure, drug, or prevention. Therefore, for developing new therapeutics, proper study models of T2DM is necessary to conduct further preclinical researches. Diabetes has been induced in animals using chemical, genetic, hormonal, antibody, viral, and surgical methods or a combination of them. Beside different approaches of diabetes induction, different animal species have been suggested. Although more than 85% of articles have proposed rat (genus Rattus) as the proper model for diabetes induction, zebrafish (Danio rerio) models of diabetes are being used more frequently in diabetes related studies. In this systematic review, we compare different aspects of available methods of inducing hyperglycemia referred as T2DM in zebrafish by utilizing a scoring system. Evaluating 26 approved models of T2DM in zebrafish, this scoring system may help researchers to compare different T2DM zebrafish models and select the best one regarding their own research theme. Eventually, glyoxalase1 (glo1-/-) knockout model of hyperglycemia achieved the highest score. In addition to assessment of hyperglycemic induction methods in zebrafish, eight most commonly proposed diabetic induction approval methods are suggested to help researchers confirm their subsequent proposed models.
Collapse
Affiliation(s)
- Aria Salehpour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bandar Bushehr, Iran
| | - Mohammad Rezaei
- Department of Diabetes, Obesity and Metabolism, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bandar Bushehr, Iran
| | - Yaser Tahamtani
- Department of Diabetes, Obesity and Metabolism, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Amin Tamadon
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bandar Bushehr, Iran
- Center of Marine Experimental and Comparative Medicine, The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bandar Bushehr, Iran
| |
Collapse
|
147
|
Inglis A, Ubungen R, Farooq S, Mata P, Thiam J, Saleh S, Shibin S, Al-Mohanna FA, Collison KS. Strain-based and sex-biased differences in adrenal and pancreatic gene expression between KK/HlJ and C57BL/6 J mice. BMC Genomics 2021; 22:180. [PMID: 33711921 PMCID: PMC7953684 DOI: 10.1186/s12864-021-07495-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/26/2021] [Indexed: 11/15/2022] Open
Abstract
Background The ever-increasing prevalence of diabetes and associated comorbidities serves to highlight the necessity of biologically relevant small-animal models to investigate its etiology, pathology and treatment. Although the C57BL/6 J model is amongst the most widely used mouse model due to its susceptibility to diet-induced obesity (DIO), there are a number of limitations namely [1] that unambiguous fasting hyperglycemia can only be achieved via dietary manipulation and/or chemical ablation of the pancreatic beta cells. [2] Heterogeneity in the obesogenic effects of hypercaloric feeding has been noted, together with sex-dependent differences, with males being more responsive. The KK mouse strain has been used to study aspects of the metabolic syndrome and prediabetes. We recently conducted a study which characterized the differences in male and female glucocentric parameters between the KK/HlJ and C57BL/6 J strains as well as diabetes-related behavioral differences (Inglis et al. 2019). In the present study, we further characterize these models by examining strain- and sex-dependent differences in pancreatic and adrenal gene expression using Affymetrix microarray together with endocrine-associated serum analysis. Results In addition to strain-associated differences in insulin tolerance, we found significant elevations in KK/HlJ mouse serum leptin, insulin and aldosterone. Additionally, glucagon and corticosterone were elevated in female mice of both strains. Using 2-factor ANOVA and a significance level set at 0.05, we identified 10,269 pancreatic and 10,338 adrenal genes with an intensity cut-off of ≥2.0 for all 4 experimental groups. In the pancreas, gene expression upregulated in the KK/HlJ strain related to increased insulin secretory granule biofunction and pancreatic hyperplasia, whereas ontology of upregulated adrenal differentially expressed genes (DEGs) related to cell signaling and neurotransmission. We established a network of functionally related DEGs commonly upregulated in both endocrine tissues of KK/HlJ mice which included the genes coding for endocrine secretory vesicle biogenesis and regulation: PCSK2, PCSK1N, SCG5, PTPRN, CHGB and APLP1. We also identified genes with sex-biased expression common to both strains and tissues including the paternally expressed imprint gene neuronatin. Conclusion Our novel results have further characterized the commonalities and diversities of pancreatic and adrenal gene expression between the KK/HlJ and C57BL/6 J strains as well as differences in serum markers of endocrine physiology. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07495-4.
Collapse
Affiliation(s)
- Angela Inglis
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia
| | - Rosario Ubungen
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia
| | - Sarah Farooq
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia
| | - Princess Mata
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia
| | - Jennifer Thiam
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia
| | - Soad Saleh
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia
| | - Sherin Shibin
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia
| | - Futwan A Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia
| | - Kate S Collison
- Department of Cell Biology, King Faisal Specialist Hospital & Research Centre, PO BOX 3354, Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
148
|
Casimiro I, Stull ND, Tersey SA, Mirmira RG. Phenotypic sexual dimorphism in response to dietary fat manipulation in C57BL/6J mice. J Diabetes Complications 2021; 35:107795. [PMID: 33308894 PMCID: PMC7856196 DOI: 10.1016/j.jdiacomp.2020.107795] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/17/2020] [Accepted: 11/07/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Obesity and the metabolic syndrome are increasingly prevalent in society and their complications and response to treatment exhibit sexual dimorphism. Mouse models of high fat diet-induced obesity are commonly used for both mechanistic and therapeutic studies of metabolic disease and diabetes. However, the inclusion of female mammals in obesity research has not been a common practice, and has resulted in a paucity of data regarding the effect of sex on metabolic parameters and its applicability to humans. METHODS Here we analyzed male and female C57BL/6 J mice beginning at 4 weeks of age that were placed on a low-fat diet (LFD, 10% calories from fat), a Western Diet (WD, 45% calories from fat), or a high fat diet (HFD, 60% calories from fat). Assessments of body composition, glucose homeostasis, insulin production, and energy metabolism, as well as histological analyses of pancreata were performed. RESULTS Both male and female C57BL/6 J mice had similar increases in total percent body weight gain with both WD and HFD compared to LFD, however, male mice gained weight earlier upon HFD or WD feeding compared to female mice. Male mice maintained their caloric food intake while reducing their locomotor activity with either WD or HFD compared to LFD, whereas female mice increased their caloric food intake with WD feeding. Locomotor activity of female mice did not significantly change upon WD or HFD feeding, yet female mice exhibited increased energy expenditure compared to WD or HFD fed male mice. Glucose tolerance tests performed at 4, 12 and 20 weeks of dietary intervention revealed impaired glucose tolerance that was worse in male mice compared to females. Furthermore, male mice exhibited an increase in pancreatic β cell area as well as reduced insulin sensitivity after HFD feeding compared to WD or LFD, whereas female mice did not. CONCLUSIONS Male and female C57BL/6 J mice exhibited strikingly different responses in weight, food consumption, locomotor activity, energy expenditure and β cell adaptation upon dietary manipulation, with the latter exhibiting less striking phenotypic changes. We conclude that the nature of these responses emphasizes the need to contextualize studies of obesity pathophysiology and treatment with respect to sex.
Collapse
Affiliation(s)
- Isabel Casimiro
- Department of Medicine, Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL 60637, United States of America
| | - Natalie D Stull
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, United States of America
| | - Sarah A Tersey
- Department of Medicine, Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL 60637, United States of America.
| | - Raghavendra G Mirmira
- Department of Medicine, Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
149
|
Park W, Kim J, Choi S, Kim T, Park M, Kim S, You JC, Kim JH, Ha KS, Lee JH, Kwon YG, Kim YM. Human plasminogen-derived N-acetyl-Arg-Leu-Tyr-Glu antagonizes VEGFR-2 to prevent blood-retinal barrier breakdown in diabetic mice. Biomed Pharmacother 2021; 134:111110. [PMID: 33338749 DOI: 10.1016/j.biopha.2020.111110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/25/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Targeting the vascular endothelial growth factor (VEGF)/its receptor-2 (VEGFR-2) system has become a mainstay of treatment for many human diseases, including retinal diseases. We examined the therapeutic effect of recently developed N-acetylated Arg-Leu-Tyr-Glu (Ac-RLYE), a human plasminogen kringle-5 domain-derived VEGFR-2 antagonists, on the pathogenesis of diabetic retinopathy. Ac-RLYE inhibited VEGF-A-mediated VEGFR-2 activation and endothelial nitric oxide synthase (eNOS)-derived NO production in the retinas of diabetic mice. In addition, Ac-RLYE prevented the disruption of adherens and tight junctions and vascular leakage by inhibiting S-nitrosylation of β-catenin and tyrosine nitration of p190RhoGAP in the retinal vasculature of diabetic mice. Peptide treatment preserved the pericyte coverage of retinal capillaries by upregulating angiopoietin-2. These results suggest that Ac-RLYE potentially prevents blood-retinal barrier breakdown and vascular leakage by antagonizing VEGFR-2; Ac-RLYE can be used as a potential therapeutic drug for the treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Wonjin Park
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Joohwan Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Seunghwan Choi
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Taesam Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Minsik Park
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Suji Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Ji-Chang You
- National Research Laboratory for Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul National University, Seoul, 03080, Republic of Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Jeong-Hyung Lee
- Department of Biochemistry, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea; Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea; Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-Do, 24341, Republic of Korea.
| |
Collapse
|
150
|
Coker CR, Keller BN, Arnold AC, Silberman Y. Impact of High Fat Diet and Ethanol Consumption on Neurocircuitry Regulating Emotional Processing and Metabolic Function. Front Behav Neurosci 2021; 14:601111. [PMID: 33574742 PMCID: PMC7870708 DOI: 10.3389/fnbeh.2020.601111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/28/2020] [Indexed: 01/12/2023] Open
Abstract
The prevalence of psychiatry disorders such as anxiety and depression has steadily increased in recent years in the United States. This increased risk for anxiety and depression is associated with excess weight gain, which is often due to over-consumption of western diets that are typically high in fat, as well as with binge eating disorders, which often overlap with overweight and obesity outcomes. This finding suggests that diet, particularly diets high in fat, may have important consequences on the neurocircuitry regulating emotional processing as well as metabolic functions. Depression and anxiety disorders are also often comorbid with alcohol and substance use disorders. It is well-characterized that many of the neurocircuits that become dysregulated by overconsumption of high fat foods are also involved in drug and alcohol use disorders, suggesting overlapping central dysfunction may be involved. Emerging preclinical data suggest that high fat diets may be an important contributor to increased susceptibility of binge drug and ethanol intake in animal models, suggesting diet could be an important aspect in the etiology of substance use disorders. Neuroinflammation in pivotal brain regions modulating metabolic function, food intake, and binge-like behaviors, such as the hypothalamus, mesolimbic dopamine circuits, and amygdala, may be a critical link between diet, ethanol, metabolic dysfunction, and neuropsychiatric conditions. This brief review will provide an overview of behavioral and physiological changes elicited by both diets high in fat and ethanol consumption, as well as some of their potential effects on neurocircuitry regulating emotional processing and metabolic function.
Collapse
Affiliation(s)
- Caitlin R. Coker
- Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC, United States
| | - Bailey N. Keller
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Amy C. Arnold
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Yuval Silberman
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|