1
|
Pan X, Xue G, Zhao M, Xiang Z, Liu D, Duan Z, Wang C. Resveratrol ameliorates high‑fat diet‑induced insulin resistance via the DDIT4/mTOR pathway in skeletal muscle. Biomed Rep 2025; 22:99. [PMID: 40297802 PMCID: PMC12035599 DOI: 10.3892/br.2025.1977] [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: 08/09/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Resveratrol (RSV) is a natural ingredient used in the treatment of diabetes mellitus. However, the antidiabetic mechanism of RSV is not clear. In the present study the antidiabetic mechanism of RSV was investigated using mice with high-fat diet (HFD)-induced insulin resistance (IR). C57BL/6J mice were divided into the following three groups: Control (CON), HFD, and HFD + RSV (RSV, 100 mg/kg body weight/day). Mice were administered RSV for 6 weeks; then biochemical and histological parameters, as well as gene and protein expression were detected. Compared with the CON group, the circulating levels of blood glucose, insulin, triglycerides, total cholesterol and high-density lipoprotein cholesterol, and area under the glucose curve were increased (P<0.05), the quantitative insulin sensitivity check index was decreased (P<0.05), and lipid accumulation in skeletal muscle was increased in the HFD group. RSV treatment was able to reverse this process and promote the IRS-1/PI3K/AKT/GLUT4 signaling pathway. Moreover, DNA damage-inducible transcript 4 (DDIT4) expression was upregulated, while the expression levels of mammalian target of rapamycin (mTOR) and p70 ribosomal protein S6 kinase were downregulated in the HFD + RSV group compared with the HFD group (P<0.05). Cell experiments inhibiting DDIT4 or activating mTOR also confirmed the role of these pathways. In summary, RSV ameliorated IR and glucose as well as lipid metabolism, and promoted the IRS-1/PI3K/AKT/GLUT4 signaling pathway through the DDIT4/mTOR signaling pathway in mice with HFD-induced IR.
Collapse
Affiliation(s)
- Xinyan Pan
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Gangqiang Xue
- Department of Pharmaceutic Preparation, The Fourth Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Ming Zhao
- Clinical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Ziping Xiang
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
- Graduate School, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Dian Liu
- Graduate School, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Zesen Duan
- Graduate School, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Chao Wang
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
2
|
Cao JJ, Gregoire BR, Bussan DD. Incorporation of Whole or Milled Dry Edible Beans into a High-Fat Diet Improves Bone Structure in Obese Mice. J Nutr 2025; 155:1442-1451. [PMID: 40089109 DOI: 10.1016/j.tjnut.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Pulses, a healthy diet component, have many bone-beneficial nutrients. OBJECTIVES This study investigated whether incorporation of dried pulses to a high-fat diet improves bone metabolism in obese mice. METHODS Male C57BL/6 mice at 4-wk-old were randomly assigned to 4 diet groups (n = 22-24/group) for 12 wk: a normal-fat (NF; 4.1 kcal/g and 16% energy as fat), a HF (4.9 kcal/g and 48% energy as fat), or a HF containing either whole beans (HFWB) or milled bean flour (HFMB) at 15% wt/wt. Diets containing beans had similar total energy, protein, and fiber content as the HF without beans. Bone structure and related biomarkers were measured. Data were analyzed using a 1-way analysis of variance followed by Tukey-Kramer post hoc contrasts. RESULTS Mice fed the HFWB or HFMB but not the HF had higher lean mass than those fed the NF (P < 0.05, 9.1% and 8.7%, for HFWB and HFMB, respectively). Mice fed the HF, HFWB, or HFMB had 23%, 14%, and 12% lower tibial bone volume/total volume (P < 0.05), respectively, than those on the NF. Mice fed the HF but not the HFWB or HFMB had 9.2% lower bone volume/total volume in the second lumbar vertebrae than those fed the NF (P < 0.05). Pooled HF with bean groups had 18%, 14%, 27%, 17%, and 15% greater body weight, fat mass, plasma concentrations of tartrate-resistant acid phosphatase, C-terminal telopeptide of type 1 collagen, blood glucose than the HF alone, respectively (P < 0.05). Milled bean flour had a greater impact on body weight, fat mass, tibial structural model index, circulating leptin, and glucose than whole bean relative to the HF alone. CONCLUSIONS These data indicate that the incorporation of dry edible beans into a HF mitigates but does not fully prevent bone deterioration in obese mice.
Collapse
Affiliation(s)
- Jay J Cao
- Grand Forks Human Nutrition Research Center, USDA, Agricultural Research Service, Grand Forks, ND, United States.
| | - Brian R Gregoire
- Grand Forks Human Nutrition Research Center, USDA, Agricultural Research Service, Grand Forks, ND, United States
| | - Derek D Bussan
- Grand Forks Human Nutrition Research Center, USDA, Agricultural Research Service, Grand Forks, ND, United States
| |
Collapse
|
3
|
Chubanava S, Karavaeva I, Ehrlich AM, Justicia RM, Basse AL, Kulik I, Dalbram E, Ahwazi D, Heaselgrave SR, Trošt K, Stocks B, Hodek O, Rodrigues RN, Havelund JF, Schlabs FL, Larsen S, Yonamine CY, Henriquez-Olguín C, Giustarini D, Rossi R, Gerhart-Hines Z, Moritz T, Zierath JR, Sakamoto K, Jensen TE, Færgeman NJ, Lavery GG, Deshmukh AS, Treebak JT. NAD depletion in skeletal muscle does not compromise muscle function or accelerate aging. Cell Metab 2025:S1550-4131(25)00212-8. [PMID: 40311622 DOI: 10.1016/j.cmet.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/27/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025]
Abstract
Nicotinamide adenine dinucleotide (NAD) is a ubiquitous electron carrier essential for energy metabolism and post-translational modification of numerous regulatory proteins. Dysregulations of NAD metabolism are widely regarded as detrimental to health, with NAD depletion commonly implicated in aging. However, the extent to which cellular NAD concentration can decline without adverse consequences remains unclear. To investigate this, we generated a mouse model in which nicotinamide phosphoribosyltransferase (NAMPT)-mediated NAD+ biosynthesis was disrupted in adult skeletal muscle. The intervention resulted in an 85% reduction in muscle NAD+ abundance while maintaining tissue integrity and functionality, as demonstrated by preserved muscle morphology, contractility, and exercise tolerance. This absence of functional impairments was further supported by intact mitochondrial respiratory capacity and unaltered muscle transcriptomic and proteomic profiles. Furthermore, lifelong NAD depletion did not accelerate muscle aging or impair whole-body metabolism. Collectively, these findings suggest that NAD depletion does not contribute to age-related decline in skeletal muscle function.
Collapse
Affiliation(s)
- Sabina Chubanava
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Iuliia Karavaeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Roger M Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Astrid L Basse
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ivan Kulik
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Munich, Germany
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Danial Ahwazi
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Samuel R Heaselgrave
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Kajetan Trošt
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ondřej Hodek
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Raissa N Rodrigues
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper F Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Farina L Schlabs
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steen Larsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Caio Y Yonamine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carlos Henriquez-Olguín
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Center for Exercise Physiology and Metabolism, Department of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Daniela Giustarini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Section of Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas E Jensen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Park E, Jeon H, Oh KI, Jeong J, Kim DW, Jin HS, Jeong SY. Coactosin-like F-actin binding protein (Cotl1) plays a key role in adipocyte differentiation and obesity. Commun Biol 2025; 8:628. [PMID: 40246959 PMCID: PMC12006365 DOI: 10.1038/s42003-025-08062-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Actin dynamics, mediated by various actin-binding proteins, plays an important role in adipocyte differentiation. We investigated the role of coactosin-like F-actin binding protein (Cotl1) in adipocyte differentiation in vitro and in vivo. Cotl1 expression level was increased during adipocyte differentiation in mouse 3T3-L1 cells and primary cultured adipose-derived stem cells (ADSCs) and during weight gain in adipose tissues. However, Cotl1 deficient in 3T3-L1 and ADSCs inhibited adipocyte differentiation, and Cotl1-/- mice displayed resistance to high-fat diet (HFD)-induced weight gain, hepatic steatosis and adipocyte enlargement compared to HFD-fed wild type (WT) mice. Ingenuity Pathway Analysis of RNA-sequencing in adipose tissues of HFD-WT and HFD-Cotl1-/- mice predicted complicated relationships between Cotl1, differentiation of adipocytes, obesity and organization of actin cytoskeleton. Particularly, peroxisome proliferator-activated receptor gamma (Pparg) emerged as a central player, with Cotl1 influencing Pparg expression, consequently regulating adipocyte differentiation. These findings suggest Cotl1 as a pivotal regulator of terminal adipocyte differentiation by modulating adipogenic genes.
Collapse
Affiliation(s)
- Eunkuk Park
- Department of Medical Genetics, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
- Department of Biological Research Laboratory, Jeonbuk Institute for Food-Bioindustry, Jeonju, Republic of Korea
| | - Hyoju Jeon
- Department of Medical Genetics, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Kang-Il Oh
- Department of Medical Genetics, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Junhwan Jeong
- Department of Medical Genetics, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Do-Wan Kim
- Department of Medical Genetics, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Hyun-Seok Jin
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan, Republic of Korea.
| | - Seon-Yong Jeong
- Department of Medical Genetics, Ajou University School of Medicine, Suwon, Republic of Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
5
|
La Merrill MA, Smith MT, McHale CM, Heindel JJ, Atlas E, Cave MC, Collier D, Guyton KZ, Koliwad S, Nadal A, Rhodes CJ, Sargis RM, Zeise L, Blumberg B. Consensus on the key characteristics of metabolism disruptors. Nat Rev Endocrinol 2025; 21:245-261. [PMID: 39613954 PMCID: PMC11916920 DOI: 10.1038/s41574-024-01059-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 12/01/2024]
Abstract
Metabolism-disrupting agents (MDAs) are chemical, infectious or physical agents that increase the risk of metabolic disorders. Examples include pharmaceuticals, such as antidepressants, and environmental agents, such as bisphenol A. Various types of studies can provide evidence to identify MDAs, yet a systematic method is needed to integrate these data to help to identify such hazards. Inspired by work to improve hazard identification of carcinogens using key characteristics (KCs), we developed 12 KCs of MDAs based on our knowledge of processes underlying metabolic diseases and the effects of their causal agents: (1) alters function of the endocrine pancreas; (2) impairs function of adipose tissue; (3) alters nervous system control of metabolic function; (4) promotes insulin resistance; (5) disrupts metabolic signalling pathways; (6) alters development and fate of metabolic cell types; (7) alters energy homeostasis; (8) causes inappropriate nutrient handling and partitioning; (9) promotes chronic inflammation and immune dysregulation in metabolic tissues; (10) disrupts gastrointestinal tract function; (11) induces cellular stress pathways; and (12) disrupts circadian rhythms. In this Consensus Statement, we present the logic that revealed the KCs of MDAs and highlight evidence that supports the identification of KCs. We use chemical, infectious and physical agents as examples to illustrate how the KCs can be used to organize and use mechanistic data to help to identify MDAs.
Collapse
Affiliation(s)
- Michele A La Merrill
- Department of Environmental Toxicology, University of California, Davis, CA, USA.
| | - Martyn T Smith
- School of Public Health, University of California, Berkeley, CA, USA
| | - Cliona M McHale
- School of Public Health, University of California, Berkeley, CA, USA
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Environmental Health Sciences, Bozeman, MT, USA
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Matthew C Cave
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
| | - David Collier
- Department of Pediatrics, East Carolina University, Greenville, NC, USA
| | - Kathryn Z Guyton
- Board on Environmental Studies and Toxicology, National Academies of Sciences, Engineering, and Medicine, Washington, DC, USA
| | - Suneil Koliwad
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Angel Nadal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), CIBERDEM, Miguel Hernandez University of Elche, Elche, Spain
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Robert M Sargis
- Division of Endocrinology, Diabetes and Metabolism, The University of Illinois at Chicago, Chicago, IL, USA
| | - Lauren Zeise
- Office of the Director, Office of Environmental Health Hazard Assessment of the California Environmental Protection Agency, Sacramento, CA, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
6
|
Cai M, Lai W, Chen H, Cao D, Zhang B, Wang F, Xian M, Wang S. Puerarin Targets HIF-1α to Modulate Hypoxia-Related Sphingolipid Metabolism in Diabetic Hepatopathy via the SPTLC2/Ceramide Pathway. Pharmaceuticals (Basel) 2025; 18:398. [PMID: 40143173 PMCID: PMC11945571 DOI: 10.3390/ph18030398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Background and Objectives: Diabetic hepatopathy, characterized by hepatic hypoxia and metabolic dysregulation, has a rising global incidence and prevalence, with limited effective treatments. Hepatic hypoxia activates hypoxia-inducible factor-1 alpha (HIF-1α), regulating sphingolipid metabolism and elevating ceramide, a key factor in insulin resistance. Puerarin (Pue), a flavonoid derived from Pueraria lobata, exhibits therapeutic effects in diabetes, but its effects on hypoxia-related hepatic metabolism are unclear. This study investigates Pue's mechanisms in modulating hepatic metabolism, focusing on HIF-1α and sphingolipid metabolism. Methods: Using bioinformatics and molecular docking, HIF-1α was identified as a key target in diabetic liver disease, confirmed via drug affinity responsive target stability. In vitro experiments utilized insulin-resistant HepG2 cells to assess glucose intake and HIF-1α expression. In vivo, type 2 diabetes mellitus (T2DM) was induced in mice using a high-fat diet and streptozotocin injections. Pue administration was evaluated for its effects on fasting blood glucose, oral glucose tolerance, and hepatoprotective effects. Liver metabolomics and qPCR/Western blot analyses were conducted to assess metabolic pathways. Results: Pue increased glucose uptake in HepG2 cells and bound HIF-1α. Pue reduced HIF-1α expression in HepG2 cells, an effect attenuated by the HIF-1α stabilizer DMOG. Pue improved fasting blood glucose, oral glucose tolerance, and hepatoprotective effects in T2DM mice, which DMOG reversed. Metabolomics revealed that Pue modulates sphingolipid metabolism, decreasing ceramide content. qPCR and Western blot results confirmed that Pue dramatically decreases HIF-1α and SPTLC2 expression. Conclusions: Pue improves diabetic hepatopathy by reducing ceramide expression through the HIF-1α/SPTLC2 pathway, offering a novel therapeutic strategy for diabetes management.
Collapse
Affiliation(s)
- Mangui Cai
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wenxi Lai
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huien Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dongmin Cao
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Boyan Zhang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Feng Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Minghua Xian
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Traditional Chinese Medicine Resource Germplasm Bank Management Center, Yunfu 527300, China
| | - Shumei Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Traditional Chinese Medicine Resource Germplasm Bank Management Center, Yunfu 527300, China
| |
Collapse
|
7
|
Lee WD, Weilandt DR, Liang L, MacArthur MR, Jaiswal N, Ong O, Mann CG, Chu Q, Hunter CJ, Ryseck RP, Lu W, Oschmann AM, Cowan AJ, TeSlaa TA, Bartman CR, Jang C, Baur JA, Titchenell PM, Rabinowitz JD. Lactate homeostasis is maintained through regulation of glycolysis and lipolysis. Cell Metab 2025; 37:758-771.e8. [PMID: 39889702 PMCID: PMC11926601 DOI: 10.1016/j.cmet.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 09/19/2024] [Accepted: 12/17/2024] [Indexed: 02/03/2025]
Abstract
Lactate is among the highest flux circulating metabolites. It is made by glycolysis and cleared by both tricarboxylic acid (TCA) cycle oxidation and gluconeogenesis. Severe lactate elevations are life-threatening, and modest elevations predict future diabetes. How lactate homeostasis is maintained, however, remains poorly understood. Here, we identify, in mice, homeostatic circuits regulating lactate production and consumption. Insulin induces lactate production by upregulating glycolysis. We find that hyperlactatemia inhibits insulin-induced glycolysis, thereby suppressing excess lactate production. Unexpectedly, insulin also promotes lactate TCA cycle oxidation. The mechanism involves lowering circulating fatty acids, which compete with lactate for mitochondrial oxidation. Similarly, lactate can promote its own consumption by lowering circulating fatty acids via the adipocyte-expressed G-protein-coupled receptor hydroxycarboxylic acid receptor 1 (HCAR1). Quantitative modeling suggests that these mechanisms suffice to produce lactate homeostasis, with robustness to noise and perturbation of individual regulatory mechanisms. Thus, through regulation of glycolysis and lipolysis, lactate homeostasis is maintained.
Collapse
Affiliation(s)
- Won Dong Lee
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Daniel R Weilandt
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Lingfan Liang
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Natasha Jaiswal
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Olivia Ong
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Charlotte G Mann
- Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Qingwei Chu
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig J Hunter
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Rolf-Peter Ryseck
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Anna M Oschmann
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Alexis J Cowan
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Tara A TeSlaa
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Caroline R Bartman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
8
|
Sun Y, Li Y, Ding X, Xu P, Jing X, Cong H, Hu H, Yu B, Xu FJ. An NIR-responsive hydrogel loaded with polydeoxyribonucleotide nano-vectors for enhanced chronic wound healing. Biomaterials 2025; 314:122789. [PMID: 39260030 DOI: 10.1016/j.biomaterials.2024.122789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
Chronic diabetic wounds are difficult to treat due to imbalanced inflammatory responses, high blood glucose levels, and bacterial infections. Novel therapeutic approaches based on nucleic acid analogues have been proposed, with unique advantages in improving angiogenesis, increasing collagen synthesis, and exerting anti-inflammatory effects. However, the inherent electronegativity of nucleic acids makes them less susceptible to cellular uptake. In this paper, a kind of near infrared (NIR)-responsive nanocomposite hydrogel loaded with nucleic acid vectors was proposed for promoting wound healing. The redox system composed of molybdenum disulphide nanosheets (MoS2 NSs) initiated the copolymerization of quaternized chitosan containing double bonds and N-isopropylacrylamide (NIPAAm) to form the matrix. In addition, MoS2 NSs with photothermal conversion performance endow the nanocomposite hydrogel to have NIR-response property and act as physical crosslinking points in the matrix. Polydeoxyribonucleotides (PDRN), which have the effect of promoting wound healing, were made into nucleic acid vectors, and loaded into the NIR-responsive hydrogel. MoS2 NSs can convert NIR irradiation into heat, causing phase transitions of temperature-sensitive segments that trigger volume contraction of the hydrogel to extrude the nucleic acid vector. Promoting angiogenesis, slowing inflammation, and guiding tissue regeneration were demonstrated in the diabetic wound model treated with the NIR-responsive nanocomposite hydrogel.
Collapse
Affiliation(s)
- Yanzhen Sun
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Yao Li
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Xiaokang Ding
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Pan Xu
- Department of Respiration, Binzhou Medical University Hospital, Binzhou, 256500, China
| | - Xiaodong Jing
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China.
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China.
| | - Fu-Jian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| |
Collapse
|
9
|
Xu J, Zhang Y, Zhao S, Zhang J, Wang Y, Liu W, Ji K, Xu G, Wen P, Wei X, Mei S, Lu L, Yao Y, Liu F, Ma Y, You J, Gao J, Buse JB, Wang J, Gu Z. A bioinspired polymeric membrane-enclosed insulin crystal achieves long-term, self-regulated drug release for type 1 diabetes therapy. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01860-0. [PMID: 40011600 DOI: 10.1038/s41565-025-01860-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 01/08/2025] [Indexed: 02/28/2025]
Abstract
The nuclear envelope serves as a highly regulated gateway for macromolecule exchange between the nucleus and cytoplasm in eukaryotes. Here we have developed a cell nucleus-mimicking polymeric membrane-enclosed system for long and self-regulated therapy. A polymeric nano-membrane with nanopores is conformally synthesized in situ on the surface of each insulin crystal, ensuring sustained, adjustable and zero-order drug release kinetics. Glucose- and β-hydroxybutyrate-dually sensitive microdomains are integrated into the nano-membranes. Under a normal state, the microdomains are uncharged and the channel is narrow enough to block insulin outflow. Under hyperglycaemia and ketonaemia, microdomains convert the high glucose and β-hydroxybutyrate concentration signals to the negative electric potential of membranes, widening the nanopores with rapid insulin outflow. In type 1 diabetic mice and minipigs, this system can maintain normoglycaemia for longer than 1 month and 3 weeks, respectively, with validated glucose- and β-hydroxybutyrate-triggered insulin release. Such membrane-enclosed drug crystal/powder formulation provides a broad platform for long-acting controlled release.
Collapse
Affiliation(s)
- Jianchang Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yang Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Sheng Zhao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Juan Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Wei Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Kangfan Ji
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Guangzheng Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Ping Wen
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xinwei Wei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Shaoqian Mei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Leihao Lu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yuejun Yao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Feng Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yufei Ma
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jiahuan You
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jianqing Gao
- Jinhua Institute of Zhejiang University, Jinhua, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - John B Buse
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
10
|
Unger CA, Hope MC, Kettering MC, Socia CE, Rice BC, Niamira DS, Cotham WE, Enos RT. The deuterated glucose insulin tolerance test: a new tool to delineate insulin-stimulated glucose uptake from suppression of endogenous glucose production. LIFE METABOLISM 2025; 4:loae036. [PMID: 39872987 PMCID: PMC11770813 DOI: 10.1093/lifemeta/loae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 01/30/2025]
Abstract
Graphical Abstract.
Collapse
Affiliation(s)
- Christian A Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29029, United States
| | - Marion C Hope
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29029, United States
| | - Michael Chase Kettering
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29029, United States
| | - Cassidy E Socia
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29029, United States
| | - Barton C Rice
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29029, United States
| | - Darya S Niamira
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29029, United States
| | - William E Cotham
- Department of Chemistry and Biochemistry, College of Arts and Science, University of South Carolina, Columbia, SC 29208, United States
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC 29029, United States
| |
Collapse
|
11
|
Ernesto CMS, Laura SSD, Obed PMI, David AGR, Eloy GGJ, Lilia GHA. LCN2 blockade mitigating metabolic dysregulation and redefining appetite control in type 2 diabetes. Metab Brain Dis 2025; 40:97. [PMID: 39808380 PMCID: PMC11732943 DOI: 10.1007/s11011-024-01454-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/28/2024] [Indexed: 01/16/2025]
Abstract
LCN2 has an osteokine important for appetite regulation; in type 2 diabetes (T2D) it is not known whether appetite regulation mediated by LCN2 in the brain is altered. In this work, we focus on exploring the role of blocking LCN2 in metabolic health and appetite regulation within the central nervous system of mice with T2D. MATERIAL AND METHODS 4-week-old male C57BL/6 mice were used, divided into four experimental groups: intact, T2D, TD2/anti-LCN2, and T2D/IgG as isotype control. T2D was induced by low doses of streptozotocin and a high-carbohydrate diet. LCN2 blockade was performed by intraperitoneal administration of a polyclonal anti-LCN2 antibody. We analyzed metabolic parameters, food intake, feeding patterns, and serum LCN2 and leptin concentrations. In another group of intact or T2D mice, we analyzed the effect of blocking LCN2 and recombinant LCN2 on food consumption in a fasting-refeeding test and, the expression of cFOS and LCN2 in brain sections, specifically in the hypothalamus, piriform cortex, visceral area, arcuate nucleus and caudate-putamen. RESULTS T2D caused an increase in serum LCN2, without alterations in Ad libitum feeding, but with changes in the feeding pattern associated with alterations in LCN2-cFOS signalling in hypothalamic and non-hypothalamic brain regions. Blocking LCN2 improved metabolic parameters, increased Ad libitum feeding, and restored the feeding pattern after fasting, which is associated with enhanced LCN2 signalling in the brain. CONCLUSIONS Blocking LCN2 restores metabolic health and normalizes the pattern of food consumption by normalizing LCN2 signalling in different brain regions.
Collapse
Affiliation(s)
- Cifuentes-Mendiola Saúl Ernesto
- Section of Osteimmunology and Oral Immunology, Laboratory of Dental Reseach. FES Iztacala, National Autonomous University of Mexico (UNAM), México, Mexico State, México
| | - Sólis-Suarez Diana Laura
- Section of Osteimmunology and Oral Immunology, Laboratory of Dental Reseach. FES Iztacala, National Autonomous University of Mexico (UNAM), México, Mexico State, México
- Postgraduate in Dentistry Science, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Pérez-Martínez Isaac Obed
- Section of Sensation Neurobiology and Oral Movements, Laboratory of Dental Reseach. FES Iztacalaestigación Odontológica, National Autonomous University of Mexico (UNAM), México State, México, México
| | - Andrade-González Rey David
- Section of Sensation Neurobiology and Oral Movements, Laboratory of Dental Reseach. FES Iztacalaestigación Odontológica, National Autonomous University of Mexico (UNAM), México State, México, México
| | - García-Gama Jahaziel Eloy
- Section of Osteimmunology and Oral Immunology, Laboratory of Dental Reseach. FES Iztacala, National Autonomous University of Mexico (UNAM), México, Mexico State, México
| | - García-Hernández Ana Lilia
- Section of Osteimmunology and Oral Immunology, Laboratory of Dental Reseach. FES Iztacala, National Autonomous University of Mexico (UNAM), México, Mexico State, México.
| |
Collapse
|
12
|
Moro C, Magnan C. Revisited guidelines for metabolic tolerance tests in mice. Lab Anim (NY) 2025; 54:16-23. [PMID: 39587363 PMCID: PMC11695259 DOI: 10.1038/s41684-024-01473-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024]
Abstract
Preclinical mouse models are extensively used in biomedical research to gain insight into disease mechanisms and to test new drug treatments. Glucose and insulin tolerance tests are simple experimental tests frequently used worldwide to assess glucose metabolism in mice. Various guidelines and methodological considerations have been published to help researchers standardize procedures and optimize research outcomes. Yet, there is still important experimental heterogeneity in the way these simple procedures are performed, with no real consensus on what the best practices are to achieve high-quality research and reproducible results. Here we critically examine several published guidelines and recent technical reports on how to perform these metabolic tests in laboratory mice and discuss the influence of various confounding factors on test results. We hope this work will help scientists establish more consensual guidelines for maximizing the relevance and clinical translation of studies using mouse models in metabolic research.
Collapse
Affiliation(s)
- Cedric Moro
- Institute of Metabolic and Cardiovascular Diseases, INSERM, Team MetaDiab, Paul Sabatier University, UMR1297, Toulouse, France.
| | - Christophe Magnan
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France
| |
Collapse
|
13
|
Laughlin M, McIndoe R, Adams SH, Araiza R, Ayala JE, Kennedy L, Lanoue L, Lantier L, Macy J, Malabanan E, McGuinness OP, Perry R, Port D, Qi N, Elias CF, Shulman GI, Wasserman DH, Lloyd KCK. The mouse metabolic phenotyping center (MMPC) live consortium: an NIH resource for in vivo characterization of mouse models of diabetes and obesity. Mamm Genome 2024; 35:485-496. [PMID: 39191872 PMCID: PMC11522164 DOI: 10.1007/s00335-024-10067-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
The Mouse Metabolic Phenotyping Center (MMPC)Live Program was established in 2023 by the National Institute for Diabetes, Digestive and Kidney Diseases (NIDDK) at the National Institutes of Health (NIH) to advance biomedical research by providing the scientific community with standardized, high quality phenotyping services for mouse models of diabetes and obesity. Emerging as the next iteration of the MMPC Program which served the biomedical research community for 20 years (2001-2021), MMPCLive is designed as an outwardly-facing consortium of service cores that collaborate to provide reduced-cost consultation and metabolic, physiologic, and behavioral phenotyping tests on live mice for U.S. biomedical researchers. Four MMPCLive Centers located at universities around the country perform complex and often unique procedures in vivo on a fee for service basis, typically on mice shipped from the client or directly from a repository or vendor. Current areas of expertise include energy balance and body composition, insulin action and secretion, whole body carbohydrate and lipid metabolism, cardiovascular and renal function, food intake and behavior, microbiome and xenometabolism, and metabolic pathway kinetics. Additionally, an opportunity arose to reduce barriers to access and expand the diversity of the biomedical research workforce by establishing the VIBRANT Program. Directed at researchers historically underrepresented in the biomedical sciences, VIBRANT-eligible investigators have access to testing services, travel and career development awards, expert advice and experimental design consultation, and short internships to learn test technologies. Data derived from experiments run by the Centers belongs to the researchers submitting mice for testing which can be made publicly available and accessible from the MMPCLive database following publication. In addition to services, MMPCLive staff provide expertise and advice to researchers, develop and refine test protocols, engage in outreach activities, publish scientific and technical papers, and conduct educational workshops and training sessions to aid researchers in unraveling the heterogeneity of diabetes and obesity.
Collapse
Affiliation(s)
- Maren Laughlin
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, USA
| | - Richard McIndoe
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, USA
| | - Sean H Adams
- Department of Surgery, School of Medicine, University of California Davis, Davis, USA
- Center for Alimentary and Metabolic Science, School of Medicine, University of California Davis, Davis, USA
| | - Renee Araiza
- Mouse Biology Program, University of California Davis, Davis, USA
| | | | - Lucy Kennedy
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, USA
| | - Louise Lanoue
- Mouse Biology Program, University of California Davis, Davis, USA
| | | | - James Macy
- Department of Comparative Medicine, Yale School of Medicine, New Haven, USA
| | | | | | - Rachel Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Daniel Port
- Mouse Biology Program, University of California Davis, Davis, USA
| | - Nathan Qi
- Department of Molecular & Integrated Physiology, University of Michigan, Ann Arbor, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, USA
| | - Carol F Elias
- Department of Molecular & Integrated Physiology, University of Michigan, Ann Arbor, USA
- Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, USA
| | | | - K C Kent Lloyd
- Department of Surgery, School of Medicine, University of California Davis, Davis, USA.
- Mouse Biology Program, University of California Davis, Davis, USA.
| |
Collapse
|
14
|
Hesketh RL, Lewis DY, Brindle KM. Optimisation of Animal Handing and Timing of 2-deoxy-2-[ 18F]fluoro-D-glucose PET Tumour Imaging in Mice. Mol Imaging Biol 2024; 26:965-976. [PMID: 39528890 PMCID: PMC11634969 DOI: 10.1007/s11307-024-01956-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE In humans, 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) tumour-to-background contrast continues to increase long after a typical uptake period of 45 - 60 min. Similar studies have not been performed in mice and the static imaging time point for most studies is arbitrarily set at 30 - 60 min post-injection of [18F]FDG. Ideally, static PET imaging should be performed after the initial period of rapid uptake but this period has not been defined in mice, with previous dynamic studies in mice being limited to 60 min. This study aimed to define the kinetics of [18F]FDG biodistribution over periods of 3 - 4 h in different murine tumour models, both subcutaneous and autochthonous, and to further refine fasting and warming protocols used prior to imaging. PROCEDURES Dynamic [18F]FDG PET-CT scans lasting 3 or 4 h were performed with C57BL/6 J and Balb/c nude mice bearing subcutaneous EL4 murine T-cell lymphoma and Colo205 human colorectal tumours, respectively, and with transgenic Eμ-Myc lymphoma mice. Prior to [18F]FDG injection, four combinations of different animal handling conditions were used: warming for 1 h at 31 °C; maintenance at room temperature (20 - 24 °C), fasting for 6 - 10 h and a fed state. RESULTS Tumour mean standardised uptake value (SUVmean) peaked at 147 ± 48 min post injection in subcutaneous tumours and 74 ± 31 min in autochthonous Eμ-Myc lymphomas. The tumour-to-blood ratio (TBR) peaked at 171 ± 57 and 83 ± 33 min in subcutaneous and autochthonous Eμ-Myc tumours, respectively. Fasting increased tumour [18F]FDG uptake and suppressed myocardial uptake in EL4 tumour-bearing mice. There was a good correlation between tumour SUVmean and Ki calculated using an input function (IDIF) derived from the inferior vena cava. CONCLUSIONS Delayed static [18F]FDG-PET imaging (> 60 min) in both autochthonous and subcutaneous tumours in improved tumour-to-background contrast and increased reproducibility.
Collapse
Affiliation(s)
- Richard L Hesketh
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK.
- Centre for Medical Imaging, University College London, Charles Bell House, 43-45 Foley Street, London, W1W 7TY, UK.
| | - David Y Lewis
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| |
Collapse
|
15
|
Gautam J, Aggarwal H, Kumari D, Gupta SK, Kumar Y, Dikshit M. A methionine-choline-deficient diet induces nonalcoholic steatohepatitis and alters the lipidome, metabolome, and gut microbiome profile in the C57BL/6J mouse. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159545. [PMID: 39089643 DOI: 10.1016/j.bbalip.2024.159545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
The methionine-choline-deficient (MCD) diet-induced non-alcoholic steatohepatitis (NASH) in mice is a well-established model. Our study aims to elucidate the factors influencing liver pathology in the MCD mouse model by examining physiological, biochemical, and molecular changes using histology, molecular techniques, and OMICS approaches (lipidomics, metabolomics, and metagenomics). Male C57BL/6J mice were fed a standard chow diet, a methionine-choline-sufficient (MCS) diet, or an MCD diet for 10 weeks. The MCD diet resulted in reduced body weight and fat mass, along with decreased plasma triglyceride, cholesterol, glucose, and insulin levels. However, it notably induced steatosis, inflammation, and alterations in gene expression associated with lipogenesis, inflammation, fibrosis, and the synthesis of apolipoproteins, sphingolipids, ceramides, and carboxylesterases. Lipid analysis revealed significant changes in plasma and tissues: most ceramide non-hydroxy-sphingosine lipids significantly decreased in the liver and plasma but increased in the adipose tissue of MCD diet-fed animals. Oxidized glycerophospholipids mostly increased in the liver but decreased in the adipose tissue of the MCD diet-fed group. The gut microbiome of the MCD diet-fed group showed an increase in Firmicutes and a decrease in Bacteroidetes and Actinobacteria. Metabolomic profiling demonstrated that the MCD diet significantly altered amino acid biosynthesis, metabolism, and nucleic acid metabolism pathways in plasma, liver, fecal, and cecal samples. LC-MS data indicated higher total plasma bile acid intensity and reduced fecal glycohyodeoxycholic acid intensity in the MCD diet group. This study demonstrates that although the MCD diet induces hepatic steatosis, the mechanisms underlying NASH in this model differ from those in human NASH pathology.
Collapse
Affiliation(s)
- Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Hobby Aggarwal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| | - Madhu Dikshit
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| |
Collapse
|
16
|
Alqudah A, Qnais E, Alqudah M, Gammoh O, Wedyan M, Abdalla SS. Isorhamnetin as a potential therapeutic agent for diabetes mellitus through PGK1/AKT activation. Arch Physiol Biochem 2024; 130:866-876. [PMID: 38445617 DOI: 10.1080/13813455.2024.2323947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/13/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
CONTEXT Type 2 Diabetes Mellitus (T2D) is a significant health concern worldwide, necessitating novel therapeutic approaches beyond conventional treatments. OBJECTIVE To assess isorhamnetin's potential in improving insulin sensitivity and mitigating T2D characteristics through oxidative and glycative stress modulation. MATERIALS AND METHODS T2D was induced in mice with a high-fat diet and streptozotocin injections. Isorhamnetin was administered at 10 mg/kg for 12 weeks. HepG2 cells were used to examine in vitro effects on stress markers and insulin sensitivity. Molecular effects on the PGK1 and AKT signalling pathway were also analyzed. RESULTS The administration of isorhamnetin significantly impacted both in vivo and in vitro models. In HepG2 cells, oxidative and glycative stresses were markedly reduced, indicating a direct effect of isorhamnetin on cellular stress pathways, which are implicated in the deterioration of insulin sensitivity. Specifically, treated cells showed a notable decrease in markers of oxidative stress, such as malondialdehyde, and advanced glycation end products, highlighting isorhamnetin's antioxidant and antiglycative properties. In vivo, isorhamnetin-treated mice exhibited substantially lower fasting glucose levels compared to untreated T2D mice, suggesting a strong hypoglycemic effect. Moreover, these mice showed improved insulin responsiveness, evidenced by enhanced glucose tolerance and insulin tolerance tests. The molecular investigation revealed that isorhamnetin activated PGK1, leading to the activation of the AKT signalling pathway, crucial for promoting glucose uptake and reducing insulin resistance. This molecular action underscores the potential mechanism through which isorhamnetin exerts its beneficial effects in T2D management. DISCUSSION The study underscores isorhamnetin's multifaceted role in T2D management, emphasizing its impact on oxidative and glycative stress reduction and molecular pathways critical for insulin sensitivity. CONCLUSION Isorhamnetin presents a promising avenue for T2D treatment, offering a novel approach to enhancing insulin sensitivity and managing glucose levels through the modulation of key molecular pathways. Further research is needed to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammed Alqudah
- Physiology Department, School of Medicine and Biomedical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Mohammed Wedyan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Shtaywy S Abdalla
- Department of Biological Sciences, Faculty of Science, University of Jordan, Amman, Jordan
| |
Collapse
|
17
|
Han Y, Quan Z, Tian M, Wang R, Guo D, Zhang D, Liu L. BACE1 inhibition via miR-6838-5p overexpression inhibits insulin resistance and the immune response in HFD-induced obesity in mice model. Immunopharmacol Immunotoxicol 2024:1-11. [PMID: 39604020 DOI: 10.1080/08923973.2024.2430668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/10/2024] [Indexed: 11/29/2024]
Abstract
CONTEXT Obesity is a chronic inflammatory disorder, which promotes the progression of metabolic disorders. MicroRNA (miR)-6838-5p is dysregulated and participates in the progression of several disorder models. OBJECTIVE To explore the role and mechanism of miR-6838-5p in insulin resistance. METHODS Mice were fed with high-fat diet (HFD) to construct an obesity animal model. The role of miR-6838-5p was evaluated by insulin tolerance test (ITT), glucose tolerance test (GTT), homeostasis model assessment of insulin resistance (HOMA-IR) analysis, enzyme-linked immunosorbent assay (ELISA) and western blot assays. The potential target of miR-6838-5p was screened through the starBase online website and confirmed by the luciferase assay. RESULTS HFD supply induced a prominent increase in the body weight, white adipose tissue (WAT) weight, the area under the curve (AUC) of GTT and ITT, HOMA-IR, the serum level of insulin and the serum concentrations and relative protein levels of interleukin (IL)-1β, IL-6 and monocyte chemoattractant protein-1 (MCP-1) accompanied with reduced levels of IL-10 in mice. The level of miR-6838-5p was reduced in HFD-fed mice. Upregulation of miR-6838-5p partly reversed the above-mentioned indicators. Moreover, miR-6838-5p directly targeted to β-site amyloid precursor protein cleaving enzyme1 (BACE1) and negatively regulated the BACE1 expression. Downregulation of BACE1 improved insulin sensitivity and inflammatory mediators release involving in AKT/GSK3β signaling pathway in HFD-fed mice. Besides, overexpression of BACE1 counteracted the depressant role of miR-6838-5p overexpression in insulin resistance and inflammatory factors release in HFD-fed mice. CONCLUSION MiR-6838-5p/BACE1 axis regulated insulin resistance and inflammatory factors release in HFD-fed mice.
Collapse
Affiliation(s)
- Yubo Han
- The First Department of Cardiovascular, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhenhua Quan
- The First Department of Cardiovascular, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Miao Tian
- Experimental Training Center, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ruinan Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Donghao Guo
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Dandan Zhang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Li Liu
- The First Department of Cardiovascular, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
18
|
Dijk FJ, van Dijk M, Roberts J, van Helvoort A, Furber MJW. Pea and soy fortified with leucine stimulates muscle protein synthesis comparable to whey in a murine ageing model. Eur J Nutr 2024; 64:12. [PMID: 39567431 PMCID: PMC11579064 DOI: 10.1007/s00394-024-03506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024]
Abstract
PURPOSE To meet the global dietary protein demands, a trend towards plant-based protein (PBP) sources to replace animal-derived protein is currently ongoing. However, PBPs may not have the same anabolic capacity to stimulate muscle protein synthesis (MPS) as dairy proteins. For vulnerable populations with specific medical needs, it is especially important to validate the anabolic properties of PBPs. In this study, a blend of pea and soy protein isolate, with or without additional leucine, was compared to whey protein isolate on MPS in aged mice. METHODS 25-Months aged C57BL/6J-mice received an oral gavage with 70 mg of whey protein isolate (W), PS protein isolate (PS; ratio 51:49), PS fortified with 19% leucine (PS + L), or 0.5mL water (F). Mice were subcutaneously injected with puromycin (0.04 µmol/g body weight, t = 30 min) and sacrificed 60 min thereafter. Left m. tibialis anterior (TA) was used to analyse MPS by the SUnSET method and mTOR signal transduction proteins. Amino acid concentrations were determined in plasma and right TA. Dried blood spots (DBS) were analysed for postprandial dynamics of amino acids at 10-20-45-60-min. RESULTS MPS was significantly increased by W and PS + L (p < 0.003), however not by PS. Pathway protein 4EBP1 showed significant increases with W, PS and PS + L to F (p < 0.0002). W and PS + L increased plasma and muscle free leucine equally, which was confirmed by DBS. CONCLUSION A PS blend fortified with leucine stimulates MPS comparable to whey protein in this acute murine ageing model. Leucine appears to be the main driver for the anabolic responses observed.
Collapse
Affiliation(s)
- Francina J Dijk
- Danone Global Research & Innovation Center B.V., Utrecht, The Netherlands.
| | - Miriam van Dijk
- Danone Global Research & Innovation Center B.V., Utrecht, The Netherlands
| | - Justin Roberts
- Danone Global Research & Innovation Center B.V., Utrecht, The Netherlands
- Cambridge Centre for Sport and Exercise Sciences, Anglia Ruskin University, Cambridge, UK
| | - Ardy van Helvoort
- Danone Global Research & Innovation Center B.V., Utrecht, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - Matthew J W Furber
- Danone Global Research & Innovation Center B.V., Utrecht, The Netherlands
| |
Collapse
|
19
|
Pittala S, Haspula D, Cui Y, Yang WM, Kim YB, Davis RJ, Wing A, Rotman Y, McGuinness OP, Inoue A, Wess J. G 12/13-mediated signaling stimulates hepatic glucose production and has a major impact on whole body glucose homeostasis. Nat Commun 2024; 15:9996. [PMID: 39557854 PMCID: PMC11574106 DOI: 10.1038/s41467-024-54299-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Altered hepatic glucose fluxes are critical during the pathogenesis of type 2 diabetes. G protein-coupled receptors represent important regulators of hepatic glucose production. Recent studies have shown that hepatocytes express GPCRs that can couple to G12/13, a subfamily of heterotrimeric G proteins that has attracted relatively little attention in the past. Here we show, by analyzing several mutant mouse strains, that selective activation of hepatocyte G12/13 signaling leads to pronounced hyperglycemia and that this effect involves the stimulation of the ROCK1-JNK signaling cascade. Using both mouse and human hepatocytes, we also show that activation of endogenous sphingosine-1-phosphate type 1 receptors strongly promotes glucose release in a G12/13-dependent fashion. Studies with human liver samples indicate that hepatic GNA12 (encoding Gα12) expression levels positively correlate with indices of insulin resistance and impaired glucose homeostasis, consistent with a potential pathophysiological role of enhanced hepatic G12/13 signaling.
Collapse
Affiliation(s)
- Srinivas Pittala
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA.
| | - Dhanush Haspula
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - Won-Mo Yang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Allison Wing
- Liver & Energy Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Yaron Rotman
- Liver & Energy Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Owen P McGuinness
- Departments of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA.
| |
Collapse
|
20
|
Parpex G, Chassaing B, Bourdon M, Santulli P, Doridot L, Thomas M, Batteux F, Chouzenoux S, Chapron C, Nicco C, Marcellin L. Western diet promotes endometriotic lesion growth in mice and induces depletion of Akkermansia muciniphila in intestinal microbiota. BMC Med 2024; 22:513. [PMID: 39501247 PMCID: PMC11539706 DOI: 10.1186/s12916-024-03738-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Endometriosis, affecting 10% of women in their reproductive years, remains poorly understood. Both individual and environmental unexplained factors are implicated in this heterogenous condition. This study aims to examine the influence of a Western diet on endometriosis lesion development in mice and to uncover the mechanisms involved. METHODS Mice were fed either a control diet or a Western diet (high in fatty acids and low in fiber) for 4 weeks. Endometriosis was then surgically induced, and lesion development was monitored by ultrasound. After 7 weeks, the mice were sacrificed for analysis of lesion characteristics through RT-qPCR, immunohistochemistry, and flow cytometry. Additionally, the intestinal microbiota was assessed using 16S rRNA gene sequencing. RESULTS Mice on the Western diet developed lesions that were significantly twice as large compared to those on the control diet. These lesions exhibited greater fibrosis and proliferation, alongside enhanced macrophage activity and leptin pathway expression. Changes in the intestinal microbiota were significantly noted after endometriosis induction, regardless of diet. Notably, mice on the Western diet with the most substantial lesions showed a loss of Akkermansia Muciniphila in their intestinal microbiota. CONCLUSIONS A Western diet significantly exacerbates lesion size in a mouse model of endometriosis, accompanied by metabolic and immune alterations. The onset of endometriosis also leads to substantial shifts in intestinal microbiota, suggesting a potential link between diet, intestinal health, and endometriosis development.
Collapse
Affiliation(s)
- Guillaume Parpex
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France.
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France.
| | - Benoît Chassaing
- Institut Pasteur, Université Paris Cité, Microbiome-Host Interaction Group, INSERM U1306, Paris, France
| | - Mathilde Bourdon
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Pietro Santulli
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Ludivine Doridot
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Marine Thomas
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Frédéric Batteux
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | | | - Charles Chapron
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Carole Nicco
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Louis Marcellin
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| |
Collapse
|
21
|
Hagarty-Waite KA, Emmons HA, Fordahl SC, Erikson KM. The Influence of Strain and Sex on High Fat Diet-Associated Alterations of Dopamine Neurochemistry in Mice. Nutrients 2024; 16:3301. [PMID: 39408267 PMCID: PMC11479034 DOI: 10.3390/nu16193301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Objective: The objective of this study was to determine the influence of sex and strain on striatal and nucleus accumbens dopamine neurochemistry and dopamine-related behavior due to a high-saturated-fat diet (HFD). Methods: Male and female C57B6/J (B6J) and Balb/cJ (Balb/c) mice were randomly assigned to a control-fat diet (CFD) containing 10% kcal fat/g or a mineral-matched HFD containing 60% kcal fat/g for 12 weeks. Results: Intraperitoneal glucose tolerance testing (IPGTT) and elevated plus maze experiments (EPM) confirmed that an HFD produced marked blunting of glucose clearance and increased anxiety-like behavior, respectively, in male and female B6J mice. Electrically evoked dopamine release in the striatum and reuptake in the nucleus accumbens (NAc), as measured by ex vivo fast scan cyclic voltammetry, was reduced for HFD-fed B6J females. Impairment in glucose metabolism explained HFD-induced changes in dopamine neurochemistry for B6J males and, to a lesser extent, Balb/c males. The relative expressions of protein markers associated with the activation of microglia, ionized calcium binding adaptor molecule (Iba1) and cluster of differentiation molecule 11b (CD11b) in the striatum were increased due to an HFD for B6J males but were unchanged or decreased amongst HFD-fed Balb/c mice. Conclusions: Our findings demonstrate that strain and sex influence the insulin- and microglia-dependent mechanisms of alterations to dopamine neurochemistry and associated behavior due to an HFD.
Collapse
Affiliation(s)
| | | | | | - Keith M. Erikson
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (K.A.H.-W.); (H.A.E.); (S.C.F.)
| |
Collapse
|
22
|
Pereira S, Hahn MK, Humber B, Chaudhry T, Wu S, Agarwal SM, Dimitrova N, Giacca A. Protocol for the hyperinsulinemic euglycemic clamp to measure glucose kinetics in rats. STAR Protoc 2024; 5:103143. [PMID: 38900633 PMCID: PMC11245906 DOI: 10.1016/j.xpro.2024.103143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/27/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024] Open
Abstract
In rats, cannulation of the jugular vein and the carotid artery precedes the use of the hyperinsulinemic euglycemic clamp to determine insulin sensitivity in vivo. Here, we present a vascular surgery protocol to allow the infusion of substances via the vein and the collection of blood samples from the artery on the day of the hyperinsulinemic euglycemic clamp. We describe steps for preparing for and performing catheterization surgery. We then detail procedures for clamp preparation and its use. For complete details on the use and execution of this protocol, please refer to Pereira et al.1,2,3.
Collapse
Affiliation(s)
- Sandra Pereira
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| | - Margaret K Hahn
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Bailey Humber
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Talha Chaudhry
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Sally Wu
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Sri Mahavir Agarwal
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, Toronto, ON, Canada
| | - Nanka Dimitrova
- Department of Comparative Medicine, University of Toronto, Toronto, ON, Canada
| | - Adria Giacca
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, Toronto, ON, Canada.
| |
Collapse
|
23
|
Pye KR, Lantier L, Ayala JE, Beall C, Ellacott KLJ. Validation of a refined protocol for mouse oral glucose tolerance testing without gavage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612859. [PMID: 39345490 PMCID: PMC11429937 DOI: 10.1101/2024.09.13.612859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
A glucose tolerance test (GTT) is routinely used to assess glucose homeostasis in clinical settings and in preclinical research studies using rodent models. The procedure assesses the ability of the body to clear glucose from the blood in a defined time after a bolus dose. In the human clinical setting, glucose is ingested via voluntary consumption of a glucose-sweetened drink. Typically, in the rodent GTT oral gavage (gavage-oGTT) or (more commonly) intraperitoneal injection (IPGTT) are used to administer the glucose bolus. Although used less frequently, likely due to investigator technical and experience barriers, the former is the more physiologically relevant as it integrates the gastrointestinal tract (GI), including release of key incretin hormones. However, orally gavaging glucose in the GTT is also not without its limitations: gavaging glucose straight into the stomach bypasses potentially critical early glucose-sensing via the mouth (cephalic phase) and associated physiological responses. Furthermore, gavaging is stressful on mice, and this by itself can increase blood glucose levels. We have developed and validated a refined protocol for mouse oral GTT which uses a voluntary oral glucose dosing method, micropipette-guided drug administration (MDA), without the need for water deprivation. This approach is simple and non-invasive. It is less stressful for the mice, as evidenced by lower circulating corticosterone levels 10 minutes after glucose-dosing compared to oral gavage. This is significant for animal and investigator welfare, and importantly minimising the confounding effect of stress on mouse glucose homeostasis. Using a randomised cross-over design, we have validated the MDA approach in the oGTT against oral gavage in male and female C57BL/6J and C57BL/6N mice. We show the ability of this method to detect changes in glucose tolerance in diet-induced obese animals. Compared to oral gavage there was lower inter-animal variation in the MDA-oGTT. In addition to being more representative of the human procedure, the MDA-oGTT is easy and has lower barriers to adoption than the gavage oGTT as it is non-invasive and requires no specialist equipment or operator training. The MDA-oGTT a more clinically representative, accessible, and refined replacement for the gavage-oGTT for mouse metabolic phenotyping, which is simple yet overcomes significant deficiencies in the current standard experimental approaches.
Collapse
|
24
|
Pan L, He X, Xu R, Bhattarai U, Niu Z, do Carmo J, Sun Y, Zeng H, Clemmer JS, Chen JX, Chen Y. Endothelial specific prolyl hydroxylase domain-containing protein 2 deficiency attenuates aging-related obesity and exercise intolerance. GeroScience 2024; 46:3945-3956. [PMID: 38462569 PMCID: PMC11226575 DOI: 10.1007/s11357-024-01108-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 02/20/2024] [Indexed: 03/12/2024] Open
Abstract
Obesity and exercise intolerance greatly reduce the life quality of older people. Prolyl hydroxylase domain-containing protein 2 (PHD2) is an important enzyme in modulating hypoxia-inducible factor-alpha (HIF) protein. Using vascular endothelial cell-specific PHD2 gene knockout (PHD2 ECKO) mice, we investigated the role of endothelial PHD2 in aging-related obesity and exercise capacity. Briefly, PHD2 ECKO mice were obtained by crossing PHD2-floxed mice with VE-Cadherin (Cdh5)-Cre transgenic mice. The effect of PHD2 ECKO on obesity and exercise capacity in PHD2 ECKO mice and control PHD2f/f mice were determined in young mice (6 to 7 months) and aged mice (16-18 months). We found that aged PHD2 ECKO mice, but not young mice, exhibited a lean phenotype, characterized by lower fat mass, and its ratio to lean weight, body weight, or tibial length, while their food uptake was not reduced compared with controls. Moreover, as compared with aged control mice, aged PHD2 ECKO mice exhibited increased oxygen consumption at rest and during exercise, and the maximum rate of oxygen consumption (VO2 max) during exercise. Furthermore, as compared with corresponding control mice, both young and aged PHD2 ECKO mice demonstrated improved glucose tolerance and lower insulin resistance. Together, these data demonstrate that inhibition of vascular endothelial PHD2 signaling significantly attenuates aging-related obesity, exercise intolerance, and glucose intolerance.
Collapse
Affiliation(s)
- Lihong Pan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Xiaochen He
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Rui Xu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Umesh Bhattarai
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ziru Niu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jussara do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - John S Clemmer
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, School of Medicine, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
25
|
Francisca S, Gloria AF, Marco PB, Camila NC, Víctor C, Bredford K. Metformin exposure during pregnancy and lactation affects offspring's long-term body weight and adipose tissue mass independent of the maternal metabolic state. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167258. [PMID: 38788910 DOI: 10.1016/j.bbadis.2024.167258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/24/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
The increasing prevalence of obesity, type 2 diabetes mellitus (T2DM), and gestational diabetes (GDM) among pregnant women has risen dramatically worldwide. The antihyperglycemic drug metformin is the most common drug for T2DM treatment in non-pregnant individuals; nevertheless, it is increasingly being used for diabetes-complicated pregnancies. Studies on the long-term metabolic effects of this drug in offspring remain scarce. This work aimed to determine the effect of metformin exposure during pregnancy and lactation on the offspring of a model of diet-induced maternal hyperglycemia. Cohorts of pregnant mice were fed a 46% fat diet (HFD) or a control standard diet (SD). A group of dams were exposed to metformin during pregnancy and lactation. After weaning, the offspring were fed SD for 8 weeks and then challenged with a 46% HFD after puberty for 12 weeks. Irrespective of the maternal diet, offspring of metformin-exposed mothers had a lower body weight and reduced inguinal white adipose tissue (iWAT) mass after HFD challenge. This was associated with increased expression of Pparg, Fabp4, Glut4, Srebp1, and Fasn in the iWAT during adulthood in the metabolically impaired dams exposed to metformin, suggesting increased adipogenesis and de novo lipogenesis. Increased expression of Fasn associated with decreased methylation levels at its promoter and proximal coding region in the iWAT was found. These results suggest that metformin modulates gene expression levels by epigenetic mechanisms in maternal metabolic-impaired conditions.
Collapse
MESH Headings
- Animals
- Metformin/pharmacology
- Female
- Pregnancy
- Lactation/drug effects
- Mice
- Prenatal Exposure Delayed Effects/metabolism
- Prenatal Exposure Delayed Effects/pathology
- Prenatal Exposure Delayed Effects/chemically induced
- Diet, High-Fat/adverse effects
- Body Weight/drug effects
- Sterol Regulatory Element Binding Protein 1/metabolism
- Sterol Regulatory Element Binding Protein 1/genetics
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Glucose Transporter Type 4/metabolism
- Glucose Transporter Type 4/genetics
- Hypoglycemic Agents/pharmacology
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/drug effects
- Obesity/metabolism
- Obesity/pathology
- Obesity/chemically induced
- Fatty Acid Synthase, Type I/metabolism
- Fatty Acid Synthase, Type I/genetics
- Male
- Mice, Inbred C57BL
- Adipose Tissue/metabolism
- Adipose Tissue/drug effects
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/chemically induced
Collapse
Affiliation(s)
- Stolzenbach Francisca
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencias, Universidad San Sebastián, Lota 2465, Providencia, Santiago 7510157, Chile; Doctorado en Ciencias mención Biología Celular y Molecular, Facultad de Ciencias, Universidad Austral de Chile, Isla Teja s/n, 5110566 Valdivia, Chile; Centro de Estudios Científicos-CECs, Av. Arturo Prat 540, 5110466 Valdivia, Chile
| | - Alarcón-Fernández Gloria
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencias, Universidad San Sebastián, Lota 2465, Providencia, Santiago 7510157, Chile; Centro de Estudios Científicos-CECs, Av. Arturo Prat 540, 5110466 Valdivia, Chile
| | - Pérez-Bustamante Marco
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencias, Universidad San Sebastián, Lota 2465, Providencia, Santiago 7510157, Chile; Doctorado en Ciencias mención Biología Celular y Molecular, Facultad de Ciencias, Universidad Austral de Chile, Isla Teja s/n, 5110566 Valdivia, Chile; Centro de Estudios Científicos-CECs, Av. Arturo Prat 540, 5110466 Valdivia, Chile
| | | | - Cortés Víctor
- Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Kerr Bredford
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencias, Universidad San Sebastián, Lota 2465, Providencia, Santiago 7510157, Chile; Centro de Estudios Científicos-CECs, Av. Arturo Prat 540, 5110466 Valdivia, Chile.
| |
Collapse
|
26
|
Zaccaron RP, de Roch Casagrande L, Venturini LM, Bittencourt JVS, da Costa C, de Pieri E, Thirupathi A, Rezin GT, Machado-de-Ávila RA, Silveira PCL. IL-1β Antagonist Receptor Peptide Associated with Photobiomodulation Accelerates Diabetic Wound Tissue Repair. Inflammation 2024; 47:1262-1277. [PMID: 38236386 DOI: 10.1007/s10753-024-01974-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Chronic hyperglycemia caused by diabetes mellitus (DM) slows down the healing process due to prolonged inflammation which impedes the regeneration progression. Photobiomodulation (PBM) is considered a non-pharmacological intervention and has anti-inflammatory and biostimulatory effects that accelerate the healing process. Currently found IL-1β inhibitors are difficult to implement due to their cytotoxic potential, excessive amounts, and invasive administration, and therefore, the application of this peptide in diabetic wounds represents a promising intervention to help resolve the inflammatory response. This study aimed to investigate the effect of an IL-1β inhibitor molecule associated with PBM irradiation in a model of epithelial injury in diabetic mice. After the induction of the DM model with streptozotocin (STZ), the skin lesion model was implemented through surgical excision. Sixty C57BL/6 mice divided into five experimental groups (n = 12) were used: excisional wound (EW), DM + EW, DM + EW + DAP 1-2 (inhibitor peptide), DM + EW + PBM, and DM + EW + PBM + DAP 1-2. Treatment started 12 h after wound induction and was performed daily for 5 days. Twenty-four hours after the last application, the animals were euthanized and the outer edge of the wound was removed. The results obtained demonstrate that the DM + EW + PBM + DAP 1-2 group caused a reduction in the levels of pro-inflammatory cytokines, an increase in anti-inflammatory cytokines, and an increase in TGF-β and maintenance of the cellular redox state with a consequent reduction in levels of inflammatory infiltrate and concomitant stimulation of type III collagen gene expression, as well as a decrease in the size of the wound in square centimeter 6 days after the injury. Only the combination of therapies was able to favor the process of tissue regeneration due to the development of an approach capable of acting at different stages of the regenerative process, through the mechanisms of action of interventions on the inflammatory process by avoiding its stagnation and stimulating progression of regeneration.
Collapse
Affiliation(s)
- Rubya Pereira Zaccaron
- Laboratory of Experimental Physiopathology, Program of Postgraduate in Science of Health, Universidade Do Extremo Sul Catarinense, Santa Catarina State, Av. Universitária, 1105 Universitário-Block S, Room 17, Criciúma, 88806-000, Brazil
| | - Laura de Roch Casagrande
- Laboratory of Experimental Physiopathology, Program of Postgraduate in Science of Health, Universidade Do Extremo Sul Catarinense, Santa Catarina State, Av. Universitária, 1105 Universitário-Block S, Room 17, Criciúma, 88806-000, Brazil
| | - Ligia Milanez Venturini
- Laboratory of Experimental Physiopathology, Program of Postgraduate in Science of Health, Universidade Do Extremo Sul Catarinense, Santa Catarina State, Av. Universitária, 1105 Universitário-Block S, Room 17, Criciúma, 88806-000, Brazil
| | - João Vitor Silvano Bittencourt
- Laboratory of Experimental Physiopathology, Program of Postgraduate in Science of Health, Universidade Do Extremo Sul Catarinense, Santa Catarina State, Av. Universitária, 1105 Universitário-Block S, Room 17, Criciúma, 88806-000, Brazil
| | - Camila da Costa
- Laboratory of Experimental Physiopathology, Program of Postgraduate in Science of Health, Universidade Do Extremo Sul Catarinense, Santa Catarina State, Av. Universitária, 1105 Universitário-Block S, Room 17, Criciúma, 88806-000, Brazil
| | - Ellen de Pieri
- Laboratory of Experimental Physiopathology, Program of Postgraduate in Science of Health, Universidade Do Extremo Sul Catarinense, Santa Catarina State, Av. Universitária, 1105 Universitário-Block S, Room 17, Criciúma, 88806-000, Brazil
| | - Anand Thirupathi
- Research Academy of Medicine Combining Sports, Ningbo No. 2 Hospital, Ningbo, 315099, China
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Ricardo Andrez Machado-de-Ávila
- Laboratory of Experimental Physiopathology, Program of Postgraduate in Science of Health, Universidade Do Extremo Sul Catarinense, Santa Catarina State, Av. Universitária, 1105 Universitário-Block S, Room 17, Criciúma, 88806-000, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Physiopathology, Program of Postgraduate in Science of Health, Universidade Do Extremo Sul Catarinense, Santa Catarina State, Av. Universitária, 1105 Universitário-Block S, Room 17, Criciúma, 88806-000, Brazil.
| |
Collapse
|
27
|
Ghoshal K, Luther JM, Pakala SB, Chetyrkin S, Falck JR, Zent R, Wasserman DH, Pozzi A. Epoxygenase Cyp2c44 Regulates Hepatic Lipid Metabolism and Insulin Signaling by Controlling FATP2 Localization and Activation of the DAG/PKCδ Axis. Diabetes 2024; 73:1229-1243. [PMID: 38743615 PMCID: PMC11262046 DOI: 10.2337/db23-0493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Abstract
Cytochrome P450 epoxygenase Cyp2c44, a murine epoxyeicosatrienoic acid (EET)-producing enzyme, promotes insulin sensitivity, and Cyp2c44-/- mice show hepatic insulin resistance. Because insulin resistance leads to hepatic lipid accumulation and hyperlipidemia, we hypothesized that Cyp2c44 regulates hepatic lipid metabolism. Standard chow diet (SCD)-fed male Cyp2c44-/- mice had significantly decreased EET levels and increased hepatic and plasma lipid levels compared with wild-type mice. We showed increased hepatic plasma membrane localization of the FA transporter 2 (FATP2) and total unsaturated fatty acids and diacylglycerol (DAG) levels. Cyp2c44-/- mice had impaired glucose tolerance and increased hepatic plasma membrane-associated PKCδ and phosphorylated IRS-1, two negative regulators of insulin signaling. Surprisingly, SCD and high-fat diet (HFD)-fed Cyp2c44-/- mice had similar glucose tolerance and hepatic plasma membrane PKCδ levels, suggesting that SCD-fed Cyp2c44-/- mice have reached their maximal glucose intolerance. Inhibition of PKCδ resulted in decreased IRS-1 serine phosphorylation and improved insulin-mediated signaling in Cyp2c44-/- hepatocytes. Finally, Cyp2c44-/- HFD-fed mice treated with the analog EET-A showed decreased hepatic plasma membrane FATP2 and PCKδ levels with improved glucose tolerance and insulin signaling. In conclusion, loss of Cyp2c44 with concomitant decreased EET levels leads to increased hepatic FATP2 plasma membrane localization, DAG accumulation, and PKCδ-mediated attenuation of insulin signaling. Thus, Cyp2c44 acts as a regulator of lipid metabolism by linking it to insulin signaling. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Kakali Ghoshal
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - James M Luther
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Suman B Pakala
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Sergei Chetyrkin
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Nashville, Nashville, TN
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Nashville, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
28
|
Shetty S, Duesman SJ, Patel S, Huynh P, Toh P, Shroff S, Das A, Chowhan D, Keller B, Alvarez J, Fisher-Foye R, Sebra R, Beaumont K, McAlpine CS, Rajbhandari P, Rajbhandari AK. Sex-specific role of high-fat diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. Biol Sex Differ 2024; 15:55. [PMID: 39010139 PMCID: PMC11247790 DOI: 10.1186/s13293-024-00628-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/11/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Scientific evidence highlights the influence of biological sex on the relationship between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress concurrently contribute to metabolic dysregulation in both males and females. Our study aimed to investigate the combined effects of high-fat diet (HFD) induced obesity and repeated stress on fear-related behaviors, metabolic, immune, and hypothalamic outcomes in male and female mice. METHODS To investigate this, we used a highly reliable rodent behavioral model that faithfully recapitulates key aspects of post-traumatic stress disorder (PTSD)-like fear. We subjected mice to footshock stressor followed by a weekly singular footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. At weeks 10 and 14 we conducted glucose tolerance and insulin sensitivity measurements. Additionally, we placed the mice in metabolic chambers to perform indirect calorimetric measurements. Finally, we collected brain and peripheral tissues for cellular analysis. RESULTS We observed that HFD-induced obesity disrupted fear memory extinction, increased glucose intolerance, and affected energy expenditure specifically in male mice. Conversely, female mice on HFD exhibited reduced respiratory exchange ratio (RER), and a significant defect in glucose tolerance only when subjected to repeated stress. Furthermore, the combination of repeated stress and HFD led to sex-specific alterations in proinflammatory markers and hematopoietic stem cells across various peripheral metabolic tissues. Single-nuclei RNA sequencing (snRNAseq) analysis of the ventromedial hypothalamus (VMH) revealed microglial activation in female mice on HFD, while male mice on HFD exhibited astrocytic activation under repeated stress. CONCLUSIONS Overall, our findings provide insights into complex interplay between repeated stress, high-fat diet regimen, and their cumulative effects on health, including their potential contribution to the development of PTSD-like stress and metabolic dysfunctions, emphasizing the need for further research to fully understand these interconnected pathways and their implications for health.
Collapse
Affiliation(s)
- Sanutha Shetty
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Samuel J Duesman
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sanil Patel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pacific Huynh
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pamela Toh
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sanjana Shroff
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anika Das
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Excellence in Youth Education, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Disha Chowhan
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Keller
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Johana Alvarez
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rachel Fisher-Foye
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Sebra
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin Beaumont
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Abha K Rajbhandari
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
29
|
Jang HJ, Min HY, Kang YP, Boo HJ, Kim J, Ahn JH, Oh SH, Jung JH, Park CS, Park JS, Kim SY, Lee HY. Tobacco-induced hyperglycemia promotes lung cancer progression via cancer cell-macrophage interaction through paracrine IGF2/IR/NPM1-driven PD-L1 expression. Nat Commun 2024; 15:4909. [PMID: 38851766 PMCID: PMC11162468 DOI: 10.1038/s41467-024-49199-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 05/23/2024] [Indexed: 06/10/2024] Open
Abstract
Tobacco smoking (TS) is implicated in lung cancer (LC) progression through the development of metabolic syndrome. However, direct evidence linking metabolic syndrome to TS-mediated LC progression remains to be established. Our findings demonstrate that 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and benzo[a]pyrene (NNK and BaP; NB), components of tobacco smoke, induce metabolic syndrome characteristics, particularly hyperglycemia, promoting lung cancer progression in male C57BL/6 J mice. NB enhances glucose uptake in tumor-associated macrophages by increasing the expression and surface localization of glucose transporter (GLUT) 1 and 3, thereby leading to transcriptional upregulation of insulin-like growth factor 2 (IGF2), which subsequently activates insulin receptor (IR) in LC cells in a paracrine manner, promoting its nuclear import. Nuclear IR binds to nucleophosmin (NPM1), resulting in IR/NPM1-mediated activation of the CD274 promoter and expression of programmed death ligand-1 (PD-L1). Restricting glycolysis, depleting macrophages, or blocking PD-L1 inhibits NB-mediated LC progression. Analysis of patient tissues and public databases reveals elevated levels of IGF2 and GLUT1 in tumor-associated macrophages, as well as tumoral PD-L1 and phosphorylated insulin-like growth factor 1 receptor/insulin receptor (pIGF-1R/IR) expression, suggesting potential poor prognostic biomarkers for LC patients. Our data indicate that paracrine IGF2/IR/NPM1/PD-L1 signaling, facilitated by NB-induced dysregulation of glucose levels and metabolic reprogramming of macrophages, contributes to TS-mediated LC progression.
Collapse
Affiliation(s)
- Hyun-Ji Jang
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Young Min
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Jin Boo
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Histology, College of Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jisung Kim
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jee Hwan Ahn
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Ho Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jin Hwa Jung
- PET core, Convergence Medicine Research Center, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Choon-Sik Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do, 14584, Republic of Korea
| | - Jong-Sook Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do, 14584, Republic of Korea
| | - Seog-Young Kim
- PET core, Convergence Medicine Research Center, Asan Medical Center, Seoul, 05505, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Ho-Young Lee
- Creative Research Initiative Center for concurrent control of emphysema and lung cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
30
|
Bednarski TK, Rahim M, Hasenour CM, Banerjee DR, Trenary IA, Wasserman DH, Young JD. Pharmacological SERCA activation limits diet-induced steatohepatitis and restores liver metabolic function in mice. J Lipid Res 2024; 65:100558. [PMID: 38729350 PMCID: PMC11179628 DOI: 10.1016/j.jlr.2024.100558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/12/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease is the most common form of liver disease and poses significant health risks to patients who progress to metabolic dysfunction-associated steatohepatitis. Fatty acid overload alters endoplasmic reticulum (ER) calcium stores and induces mitochondrial oxidative stress in hepatocytes, leading to hepatocellular inflammation and apoptosis. Obese mice have impaired liver sarco/ER Ca2+-ATPase (SERCA) function, which normally maintains intracellular calcium homeostasis by transporting Ca2+ ions from the cytoplasm to the ER. We hypothesized that restoration of SERCA activity would improve diet-induced steatohepatitis in mice by limiting ER stress and mitochondrial dysfunction. WT and melanocortin-4 receptor KO (Mc4r-/-) mice were placed on either chow or Western diet (WD) for 8 weeks. Half of the WD-fed mice were administered CDN1163 to activate SERCA, which reduced liver fibrosis and inflammation. SERCA activation also restored glucose tolerance and insulin sensitivity, improved histological markers of metabolic dysfunction-associated steatohepatitis, increased expression of antioxidant enzymes, and decreased expression of oxidative stress and ER stress genes. CDN1163 decreased hepatic citric acid cycle flux and liver pyruvate cycling, enhanced expression of mitochondrial respiratory genes, and shifted hepatocellular [NADH]/[NAD+] and [NADPH]/[NADP+] ratios to a less oxidized state, which was associated with elevated PUFA content of liver lipids. In sum, the data demonstrate that pharmacological SERCA activation limits metabolic dysfunction-associated steatotic liver disease progression and prevents metabolic dysfunction induced by WD feeding in mice.
Collapse
Affiliation(s)
- Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina A Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
31
|
Srisutha J, Watari I, Akakura M, Watanabe M, Changsiripun C, Ono T. P2X7R and P2X4R expression of mice submandibular gland in high-fat diet/streptozotocin-induced type 2 diabetes. Sci Rep 2024; 14:10855. [PMID: 38740782 PMCID: PMC11091137 DOI: 10.1038/s41598-024-60519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic inflammatory disease that can compromise the functioning of various organs, including the salivary glands (SG). The purinergic system is one of the most important inflammatory pathways in T2DM condition, and P2X7R and P2X4R are the primary purinergic receptors in SG that regulate inflammatory homeostasis. This study aimed to evaluate P2X7R and P2X4R expression, and morphological changes in the submandibular gland (SMG) in T2DM. Twenty-four 5-week-old mice were randomly assigned to control (CON) and diabetes mellitus (DM) groups (n = 12 each). Body weight, diet, and blood glucose levels were monitored weekly. The histomorphology of the SMG and the expression of the P2X7R, and P2X7R was evaluated by immunohistochemistry (IHC) staining and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) at 11 and 13 weeks of age. Our findings indicate a significant increase in food consumption, body weight, and blood glucose levels in the DM group. Although a significant increase in P2X7R and P2X4R expression was observed in the DM groups, the receptor location remained unchanged. We also observed a significant increase in the acinar area in the DM13w group, and a significant decrease in the ductal area in the DM11w and DM13w groups. Targeting purinergic receptors may offer novel therapeutic methods for diabetic complications.
Collapse
MESH Headings
- Animals
- Mice
- Blood Glucose/metabolism
- Body Weight
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat/adverse effects
- Mice, Inbred C57BL
- Receptors, Purinergic P2X4/metabolism
- Receptors, Purinergic P2X4/genetics
- Receptors, Purinergic P2X7/metabolism
- Receptors, Purinergic P2X7/genetics
- Streptozocin
- Submandibular Gland/metabolism
- Submandibular Gland/pathology
Collapse
Affiliation(s)
- Jiratchaya Srisutha
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Ippei Watari
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan.
| | - Masato Akakura
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan
| | - Minami Watanabe
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan
| | - Chidsanu Changsiripun
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo city, Tokyo, 113-8510, Japan
| |
Collapse
|
32
|
Aboelnazar S, Ghoneim H, Shalaby T, Sorour S, Osman EM. Modulatory effect of interleukin-2 loaded chitosan nano sphere on regulatory T cell activity in streptozotocin-induced diabetic mice. Int Immunopharmacol 2024; 132:112019. [PMID: 38599099 DOI: 10.1016/j.intimp.2024.112019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024]
Abstract
OBJECTIVE The current study aimed to assess the modulating effect of IL-2 encapsulated chitosan-nanoparticles (CSNPs) on the function of Treg cells through induction of type 1 diabetes (T1D). Treg cell function was monitored by the forkhead box P3 (FoxP3) and transforming growth factor beta (TGFβ) levels, correlating them with blood glucose and serum insulin levels. MATERIALS AND METHODS In this case-control study, a low dose of IL-2 (free and chitosan-loaded) was injected into a diabetic mice group. The levels of FoxP3 and TGF-β 1 were assessed using Enzyme-Linked Immunosorbent Assay. In addition, blood glucose and serum insulin levels were determined. RESULTS The mean glucose level decreased significantly after free rIL-2 or rIL-2 / CSNPs treatment. Meanwhile, the mean serum insulin level was significantly increased after treatment with free rIL-2 or rIL-2/CSNPs. The mean levels of FoxP3 and TGFβ 1 were significantly increased with either free rIL-2 or rIL-2/CSNPs compared to the T1D untreated group (P < 0.001). In the treated mice group receiving free CSNPs, there was a significant negative correlation between glucose and insulin levels. Moreover, FoxP3 & TGFβ 1 levels had a significant positive correlation. In treated mice groups with free rIL-2 and IL-2 CSNPs, there was a significant positive correlation between FoxP3 and glucose levels. A significant negative correlation was found after conducting a correlation between insulin level and FoxP3 in the T1D/ rIL-2 / CSNPs group. CONCLUSIONS Low-dose IL-2 selectively modulates FoxP3 + Tregs, and TGFβ 1 increases their levels. These results demonstrated that IL-2-free and chitosan-loaded nanoparticles can be therapeutic agents in T1D.
Collapse
Affiliation(s)
- Salma Aboelnazar
- Department of Immunology and Allergy, Medical Research Institute, Alexandria University, Egypt
| | - Hossam Ghoneim
- Department of Immunology and Allergy, Medical Research Institute, Alexandria University, Egypt
| | - Thanaa Shalaby
- Department of Biophysics, Medical Research Institute, Alexandria University, Egypt
| | - Sally Sorour
- Department of Biophysics, Medical Research Institute, Alexandria University, Egypt
| | - Eman M Osman
- Department of Immunology and Allergy, Medical Research Institute, Alexandria University, Egypt.
| |
Collapse
|
33
|
Zagalo L, Pereira G, Casal D, Gonçalves LL, Zagalo C, Oliveira MJ, Oliveira P, Brito JAA. Impact of infrasound exposure and streptozotocin-induced glucose intolerance on bone composition in Wistar rats. BMC Res Notes 2024; 17:128. [PMID: 38711110 DOI: 10.1186/s13104-024-06784-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
The elemental composition of chemical elements can vary between healthy and diseased tissues, providing essential insights into metabolic processes in physiological and diseased states. This study aimed to evaluate the calcium (Ca) and phosphorus (P) levels in the bones of rats with/without streptozotocin-induced diabetes and/or exposure to infrasound. X-ray fluorescence spectroscopy was used to determine the concentrations of Ca and P in Wistar rat tibiae samples.The results showed a significant decrease in bone P concentration in streptozotocin-induced diabetic rats compared to untreated animals. Similarly, the Ca/P ratio was higher in the streptozotocin-induced diabetic group. No significant differences were observed in bone Ca concentration between the studied groups or between animals exposed and not exposed to infrasound.Moreover, streptozotocin-induced diabetic rats had lower bone P concentration but unaltered bone Ca concentration compared to untreated rats. Infrasound exposure did not impact bone Ca or P levels. The reduced bone P concentration may be associated with an increased risk of bone fractures in diabetes.
Collapse
Affiliation(s)
- Luísa Zagalo
- Center for Interdisciplinary Research Egas Moniz (CiiEM), Monte da Caparica, Portugal
| | - Gonçalo Pereira
- Center for Interdisciplinary Research Egas Moniz (CiiEM), Monte da Caparica, Portugal
| | - Diogo Casal
- Plastic Surgery Consultant - Central, Lisbon University Hospital Centre, Lisbon, Portugal
| | - Luísa L Gonçalves
- Center for Interdisciplinary Research Egas Moniz (CiiEM), Monte da Caparica, Portugal
| | - Carlos Zagalo
- Center for Interdisciplinary Research Egas Moniz (CiiEM), Monte da Caparica, Portugal
| | - Maria João Oliveira
- Department of Anatomy and UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
- CINTESIS@RISE, University of Porto, Porto, Portugal
| | - Pedro Oliveira
- Center for Interdisciplinary Research Egas Moniz (CiiEM), Monte da Caparica, Portugal
- Anatomy Institute, School of Medicine, University of Lisbon, Lisbon, Portugal
| | - José A A Brito
- Center for Interdisciplinary Research Egas Moniz (CiiEM), Monte da Caparica, Portugal.
| |
Collapse
|
34
|
Ghooray DT, Xu M, Shi H, McClain CJ, Song M. Hepatocyte-Specific Fads1 Overexpression Attenuates Western Diet-Induced Metabolic Phenotypes in a Rat Model. Int J Mol Sci 2024; 25:4836. [PMID: 38732052 PMCID: PMC11084797 DOI: 10.3390/ijms25094836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/01/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Fatty acid desaturase 1 (FADS1) is a rate-limiting enzyme in long-chain polyunsaturated fatty acid (LCPUFA) synthesis. Reduced activity of FADS1 was observed in metabolic dysfunction-associated steatotic liver disease (MASLD). The aim of this study was to determine whether adeno-associated virus serotype 8 (AAV8) mediated hepatocyte-specific overexpression of Fads1 (AAV8-Fads1) attenuates western diet-induced metabolic phenotypes in a rat model. Male weanling Sprague-Dawley rats were fed with a chow diet, or low-fat high-fructose (LFHFr) or high-fat high-fructose diet (HFHFr) ad libitum for 8 weeks. Metabolic phenotypes were evaluated at the endpoint. AAV8-Fads1 injection restored hepatic FADS1 protein levels in both LFHFr and HFHFr-fed rats. While AAV8-Fads1 injection led to improved glucose tolerance and insulin signaling in LFHFr-fed rats, it significantly reduced plasma triglyceride (by ~50%) and hepatic cholesterol levels (by ~25%) in HFHFr-fed rats. Hepatic lipidomics analysis showed that FADS1 activity was rescued by AAV8-FADS1 in HFHFr-fed rats, as shown by the restored arachidonic acid (AA)/dihomo-γ-linolenic acid (DGLA) ratio, and that was associated with reduced monounsaturated fatty acid (MUFA). Our data suggest that the beneficial role of AAV8-Fads1 is likely mediated by the inhibition of fatty acid re-esterification. FADS1 is a promising therapeutic target for MASLD in a diet-dependent manner.
Collapse
Affiliation(s)
- Dushan T. Ghooray
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (D.T.G.); (M.X.); (C.J.M.)
| | - Manman Xu
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (D.T.G.); (M.X.); (C.J.M.)
| | - Hongxue Shi
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Craig J. McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (D.T.G.); (M.X.); (C.J.M.)
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA
| | - Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (D.T.G.); (M.X.); (C.J.M.)
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
35
|
Vatashchuk MV, Hurza VV, Stefanyshyn N, Bayliak MM, Gospodaryov DV, Garaschuk O, Lushchak VI. Impact of caloric restriction on oxidative stress and key glycolytic enzymes in the cerebral cortex, liver and kidney of old and middle-aged mice. Neuropharmacology 2024; 247:109859. [PMID: 38340956 DOI: 10.1016/j.neuropharm.2024.109859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/28/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024]
Abstract
Caloric restriction (CR) is proposed as a strategy to prevent age-related alterations like impaired glucose metabolism and intensification of oxidative stress. In this study, we examined effects of aging and CR on the activities of glycolytic enzymes and parameters of oxidative stress in the cerebral cortex, liver, and kidney of middle-aged (9 months old) and old (18 months old) C57BL6/N mice. Control middle-aged and old mice were fed ad libitum (AL groups), whereas age-matched CR groups were subjected to CR (70% of individual ad libitum food intake) for 6 and 12 months, respectively. There were no significant differences in the activities of key glycolytic and antioxidant enzymes and oxidative stress indices between the cortices of middle-aged and old AL mice. The livers and kidneys of old AL mice showed higher activity of glucose-6-phosphate dehydrogenase, an enzyme that produces NADPH in the pentose phosphate pathway, compared to those of middle-aged mice. CR regimen modulated some biochemical parameters in middle-aged but not in old mice. In particular, CR decreased oxidative stress intensity in the liver and kidney but had no effects on those parameters in the cerebral cortex. In the liver, CR led to lower activities of glycolytic enzymes, whereas its effect was the opposite in the kidney. The results suggest that during physiological aging there is no significant intensification of oxidative stress and glycolysis decline in mouse tissues during the transition from middle to old age. The CR regimen has tissue-specific effects and improves the metabolic state of middle-aged mice. This article is part of the Special Issue on "Ukrainian Neuroscience".
Collapse
Affiliation(s)
- Myroslava V Vatashchuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk, 76018, Ukraine
| | - Viktoriia V Hurza
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk, 76018, Ukraine
| | - Nadiia Stefanyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk, 76018, Ukraine
| | - Maria M Bayliak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk, 76018, Ukraine
| | - Dmytro V Gospodaryov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk, 76018, Ukraine
| | - Olga Garaschuk
- Department of Neurophysiology, University of Tübingen, Tübingen, 72074, Germany.
| | - Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk, 76018, Ukraine; Research and Development University, 13a Shota Rustaveli Str., Ivano-Frankivsk, 76018, Ukraine.
| |
Collapse
|
36
|
Rojas-Criollo M, Novau-Ferré N, Gutierrez-Tordera L, Ettcheto M, Folch J, Papandreou C, Panisello L, Cano A, Mostafa H, Mateu-Fabregat J, Carrasco M, Camins A, Bulló M. Effects of a High-Fat Diet on Insulin-Related miRNAs in Plasma and Brain Tissue in APP Swe/PS1dE9 and Wild-Type C57BL/6J Mice. Nutrients 2024; 16:955. [PMID: 38612989 PMCID: PMC11013640 DOI: 10.3390/nu16070955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Insulin resistance (IR)-related miRNAs have been associated with the development and progression of Alzheimer's disease (AD). The dietary modulation of these miRNAs could become a potential strategy to manage AD. The aim of this study was to evaluate the effect of a high-fat diet (HFD), which aggravates AD-related pathogenic processes, on serum, cortex and hippocampus IR-related miRNA expression. C57BL/6J WT and APPSwe/PS1dE9 mice were fed either an HFD or a conventional diet till 6 months of age. The mice fed with the HFD showed a significant increase in body weight and worsening glucose and insulin metabolism. miR-19a-3p was found to be up-regulated in the cortex, hippocampus and serum of APP/PS1 mice and in the serum and hippocampus of WT mice fed with the HFD. miR-34a-5p and miR-146a-5p were up-regulated in the serum of both groups of mice after consuming the HFD. Serum miR-29c-3p was overexpressed after consuming the HFD, along with hippocampal miR-338-3p and miR-125b-5p, only in WT mice. The HFD modulated the expression of peripheral and brain miRNAs related to glucose and insulin metabolism, suggesting the potential role of these miRNAs not only as therapeutic targets of AD but also as peripheral biomarkers for monitoring AD.
Collapse
Affiliation(s)
- Melina Rojas-Criollo
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Nil Novau-Ferré
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Laia Gutierrez-Tordera
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain; (M.E.); (M.C.); (A.C.)
- Institute of Neuroscience, Universitat de Barcelona, 08034 Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, 28029 Madrid, Spain;
| | - Jaume Folch
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, 28029 Madrid, Spain;
| | - Christopher Papandreou
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Laura Panisello
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, 28029 Madrid, Spain;
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08028 Barcelona, Spain
| | - Hamza Mostafa
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Javier Mateu-Fabregat
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Marina Carrasco
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain; (M.E.); (M.C.); (A.C.)
- Institute of Neuroscience, Universitat de Barcelona, 08034 Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, 28029 Madrid, Spain;
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain; (M.E.); (M.C.); (A.C.)
- Institute of Neuroscience, Universitat de Barcelona, 08034 Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, 28029 Madrid, Spain;
| | - Mònica Bulló
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain; (M.R.-C.); (N.N.-F.); (L.G.-T.); (J.F.); (C.P.); (L.P.); (H.M.); (J.M.-F.)
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology—TecnATox, Rovira i Virgili University, 43201 Reus, Spain
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
37
|
Kusamoto A, Harada M, Minemura A, Matsumoto A, Oka K, Takahashi M, Sakaguchi N, Azhary JMK, Koike H, Xu Z, Tanaka T, Urata Y, Kunitomi C, Takahashi N, Wada-Hiraike O, Hirota Y, Osuga Y. Effects of the prenatal and postnatal nurturing environment on the phenotype and gut microbiota of mice with polycystic ovary syndrome induced by prenatal androgen exposure: a cross-fostering study. Front Cell Dev Biol 2024; 12:1365624. [PMID: 38590777 PMCID: PMC10999616 DOI: 10.3389/fcell.2024.1365624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/05/2024] [Indexed: 04/10/2024] Open
Abstract
The gut microbiome is implicated in the pathogenesis of polycystic ovary syndrome (PCOS), and prenatal androgen exposure is involved in the development of PCOS in later life. Our previous study of a mouse model of PCOS induced by prenatal dihydrotestosterone (DHT) exposure showed that the reproductive phenotype of PCOS appears from puberty, followed by the appearance of the metabolic phenotype after young adulthood, while changes in the gut microbiota was already apparent before puberty. To determine whether the prenatal or postnatal nurturing environment primarily contributes to these changes that characterize prenatally androgenized (PNA) offspring, we used a cross-fostering model to evaluate the effects of changes in the postnatal early-life environment of PNA offspring on the development of PCOS-like phenotypes and alterations in the gut microbiota in later life. Female PNA offspring fostered by normal dams (exposed to an abnormal prenatal environment only, fostered PNA) exhibited less marked PCOS-like phenotypes than PNA offspring, especially with respect to the metabolic phenotype. The gut microbiota of the fostered PNA offspring was similar to that of controls before adolescence, but differences between the fostered PNA and control groups became apparent after young adulthood. In conclusion, both prenatal androgen exposure and the postnatal early-life environment created by the DHT injection of mothers contribute to the development of PCOS-like phenotypes and the alterations in the gut microbiota that characterize PNA offspring. Thus, both the pre- and postnatal environments represent targets for the prevention of PCOS and the associated alteration in the gut microbiota in later life.
Collapse
Affiliation(s)
- Akari Kusamoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Ayaka Minemura
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Asami Matsumoto
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Kentaro Oka
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | | | - Nanoka Sakaguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Jerilee M. K. Azhary
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hiroshi Koike
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Zixin Xu
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Tsurugi Tanaka
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Yoko Urata
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Chisato Kunitomi
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Nozomi Takahashi
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
38
|
Juttla PK, Chege BM, Mwangi PW, Bukachi F. Dapagliflozin Pretreatment Prevents Cardiac Electrophysiological Changes in a Diet and Streptozotocin Induction of Type 2 Diabetes in Rats: A Potential New First-Line? J Exp Pharmacol 2024; 16:123-133. [PMID: 38525051 PMCID: PMC10961018 DOI: 10.2147/jep.s443169] [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: 10/15/2023] [Accepted: 02/22/2024] [Indexed: 03/26/2024] Open
Abstract
Purpose Dapagliflozin exerts cardioprotective effects in Type 2 Diabetes Mellitus (T2DM). However, whether these effects prevent electrocardiographic changes associated with T2DM altogether remain unknown. Our aim was to investigate the prophylactic effect of dapagliflozin pretreatment on the rat ECG using a high-fat, high-fructose (HFHf) diet and a low dose streptozotocin (STZ) model of T2DM. Methods Twenty-five (25) rats were randomized into five (5) groups: normal control receiving a normal diet while the other groups received an 8-week HFHf and 40mg/kg STZ on day 42, and either: saline for the diabetic control (1 mg/kg/d), low dose (1.0 mg/kg/d) and high dose dapagliflozin (1.6 mg/kg/d), or metformin (250 mg/kg/d). Oral glucose tolerance (OGT), electrocardiograms (ECGs), paracardial adipose mass, and left ventricular fibrosis were determined. Data were analyzed using GraphPad version 9.0.0.121, with the level of significance at p < 0.05. Results Compared to the diabetic control group, a high dose of dapagliflozin preserved the OGT (p = 0.0001), QRS-duration (p = 0.0263), QT-interval (p = 0.0399), and QTc intervals (p = 0.0463). Furthermore, the high dose dapagliflozin group had the lowest paracardial adipose mass (p = 0.0104) and fibrotic area (p = 0.0001). In contrast, while metformin showed favorable effects on OGT (p = 0.0025), paracardial adiposity (p = 0.0153) and ventricular fibrosis (p = 0.0291), it did not demonstrate significant antiarrhythmic effects. Conclusion Pretreatment with higher doses of Dapagliflozin exhibits prophylactic cardioprotective characteristics against diabetic cardiomyopathy that include antifibrotic and antiarrhythmic qualities. This suggests that higher doses of dapagliflozin could be a more effective initial therapeutic option in T2DM.
Collapse
Affiliation(s)
| | | | | | - Frederick Bukachi
- Department of Medical Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
39
|
Hahn MK, Giacca A, Pereira S. In vivo techniques for assessment of insulin sensitivity and glucose metabolism. J Endocrinol 2024; 260:e230308. [PMID: 38198372 PMCID: PMC10895285 DOI: 10.1530/joe-23-0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
Metabolic tests are vital to determine in vivo insulin sensitivity and glucose metabolism in preclinical models, usually rodents. Such tests include glucose tolerance tests, insulin tolerance tests, and glucose clamps. Although these tests are not standardized, there are general guidelines for their completion and analysis that are constantly being refined. In this review, we describe metabolic tests in rodents as well as factors to consider when designing and performing these tests.
Collapse
Affiliation(s)
- Margaret K Hahn
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Toronto, Ontario, Canada
| | - Adria Giacca
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Osei-Ntansah A, Oliver T, Lofton T, Falzarano C, Carr K, Huang R, Wilson A, Damaser E, Harvey G, Rahman MA, Andrisse S. Liver Androgen Receptor Knockout Improved High-fat Diet Induced Glucose Dysregulation in Female Mice But Not Male Mice. J Endocr Soc 2024; 8:bvae021. [PMID: 38425436 PMCID: PMC10904101 DOI: 10.1210/jendso/bvae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Indexed: 03/02/2024] Open
Abstract
Previous research has indicated that liver androgen receptors may play a role in modulating disease. This study aims to investigate the pathophysiology of high-fat diet (HFD) induced dysglycemia in male and female liver androgen receptor knockout (LivARKO) mice. We performed metabolic tests on LivARKO female and male mice fed a HFD or a control diet (from Research Diets Inc.) during months 1 or 2 after starting the diet. Additionally, we performed Western blot and quantitative real-time PCR analysis on the livers of the mice to examine intermediates in the insulin signaling pathway. LivARKO-HFD female mice displayed no difference in glucose tolerance compared to female LivARKO-Control (Con) mice, whereas in wild-type female mice, HFD impaired glucose tolerance (IGT). Our data suggests that starting at 1 month, LivARKO may be protecting female mice from HFD-induced metabolic dysfunction. LivARKO-HFD female mice displayed significantly worse insulin sensitivity at 15 minutes compared to LivARKO-Con female mice, but, strangely, LivARKO-HFD female mice had significantly better insulin sensitivity at 60 and 90 minutes compared to LivARKO-Con female mice. Despite protecting against IGT, LivARKO did not protect against HFD-induced hyperinsulinemia in female mice. In contrast to females, male LivARKO-HFD mice displayed impaired glucose tolerance compared to male LivARKO-Con mice. Thus, LivARKO is not protective against HFD-induced glucose metabolic dysfunction in male mice. Lastly, LivARKO-HFD female mice maintained hepatic insulin sensitivity whereas LivARKO-HFD male mice displayed hepatic insulin resistance. These findings suggest that LivARKO delayed the onset of HFD-induced dysglycemia in female mice.
Collapse
Affiliation(s)
- Adjoa Osei-Ntansah
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Trinitee Oliver
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Taylor Lofton
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Claire Falzarano
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Kiana Carr
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Ruthe Huang
- From Prison Cells To PhD, Baltimore, MD 21224, USA
| | - Andre Wilson
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Ella Damaser
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Guyton Harvey
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Md Ahasanur Rahman
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Stanley Andrisse
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| |
Collapse
|
41
|
Gilley SP, Zarate MA, Zheng L, Jambal P, Yazza DN, Chintapalli SV, MacLean PS, Wright CJ, Rozance PJ, Shankar K. Metabolic and fecal microbial changes in adult fetal growth restricted mice. Pediatr Res 2024; 95:647-659. [PMID: 37935884 PMCID: PMC10899111 DOI: 10.1038/s41390-023-02869-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Fetal growth restriction (FGR) increases risk for development of obesity and type 2 diabetes. Using a mouse model of FGR, we tested whether metabolic outcomes were exacerbated by high-fat diet challenge or associated with fecal microbial taxa. METHODS FGR was induced by maternal calorie restriction from gestation day 9 to 19. Control and FGR offspring were weaned to control (CON) or 45% fat diet (HFD). At age 16 weeks, offspring underwent intraperitoneal glucose tolerance testing, quantitative MRI body composition assessment, and energy balance studies. Total microbial DNA was used for amplification of the V4 variable region of the 16 S rRNA gene. Multivariable associations between groups and genera abundance were assessed using MaAsLin2. RESULTS Adult male FGR mice fed HFD gained weight faster and had impaired glucose tolerance compared to control HFD males, without differences among females. Irrespective of weaning diet, adult FGR males had depletion of Akkermansia, a mucin-residing genus known to be associated with weight gain and glucose handling. FGR females had diminished Bifidobacterium. Metabolic changes in FGR offspring were associated with persistent gut microbial changes. CONCLUSION FGR results in persistent gut microbial dysbiosis that may be a therapeutic target to improve metabolic outcomes. IMPACT Fetal growth restriction increases risk for metabolic syndrome later in life, especially if followed by rapid postnatal weight gain. We report that a high fat diet impacts weight and glucose handling in a mouse model of fetal growth restriction in a sexually dimorphic manner. Adult growth-restricted offspring had persistent changes in fecal microbial taxa known to be associated with weight, glucose homeostasis, and bile acid metabolism, particularly Akkermansia, Bilophilia and Bifidobacteria. The gut microbiome may represent a therapeutic target to improve long-term metabolic outcomes related to fetal growth restriction.
Collapse
Affiliation(s)
- Stephanie P Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Miguel A Zarate
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lijun Zheng
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Purevsuren Jambal
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Deaunabah N Yazza
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul S MacLean
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Clyde J Wright
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paul J Rozance
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
42
|
Shin MK, Tang WY, Amorim MR, Sham JSK, Polotsky VY. Carotid body denervation improves hyperglycemia in obese mice. J Appl Physiol (1985) 2024; 136:233-243. [PMID: 38126089 PMCID: PMC11219014 DOI: 10.1152/japplphysiol.00215.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
The carotid bodies (CBs) have been implicated in glucose abnormalities in obesity via elevation of activity of the sympathetic nervous system. Obesity-induced hypertension is mediated by insulin receptor (INSR) signaling and by leptin, which binds to the leptin receptor (LEPRb) in CB and activates transient receptor potential channel subfamily M member 7 (TRPM7). We hypothesize that in mice with diet-induced obesity, hyperglycemia, glucose intolerance, and insulin resistance will be attenuated by the CB denervation (carotid sinus nerve dissection, CSND) and by knockdown of Leprb, Trpm7, and Insr gene expression in CB. In series of experiments in 75 male diet-induced obese (DIO) mice, we performed either CSND (vs. sham) surgeries or shRNA-induced suppression of Leprb, Trpm7, or Insr gene expression in CB, followed by blood pressure telemetry, intraperitoneal glucose tolerance and insulin tolerance tests, and measurements of fasting plasma insulin, leptin, corticosterone, glucagon and free fatty acids (FFAs) levels, hepatic expression of gluconeogenesis enzymes phosphoenolpyruvate carboxykinase (PEPCK) and glucose 6-phosphatase (G-6-Pase) mRNA and liver glycogen levels. CSND decreased blood pressure, fasting blood glucose levels and improved glucose tolerance without any effect on insulin resistance. CSND did not affect any hormone levels and gluconeogenesis enzymes, but increased liver glycogen level. Genetic knockdown of CB Leprb, Trpm7, and Insr had no effect on glucose metabolism. We conclude that CB contributes to hyperglycemia of obesity, probably by modulation of the glycogen-glucose equilibrium. Diabetogenic effects of obesity on CB in mice do not occur via activation of CB Leprb, Trpm7, and Insr.NEW & NOTEWORTHY This paper provides first evidence that carotid body denervation abolishes hypertension and improves fasting blood glucose levels and glucose tolerance in mice with diet-induced obesity. Furthermore, we have shown that this phenomenon is associated with increased liver glycogen content, whereas insulin sensitivity and enzymes of gluconeogenesis were not affected.
Collapse
Affiliation(s)
- Mi-Kyung Shin
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States
| | - Mateus R Amorim
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - James S-K Sham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Vsevolod Y Polotsky
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| |
Collapse
|
43
|
Negm A, Stobbe K, Ben Fradj S, Sanchez C, Landra-Willm A, Richter M, Fleuriot L, Debayle D, Deval E, Lingueglia E, Rovere C, Noel J. Acid-sensing ion channel 3 mediates pain hypersensitivity associated with high-fat diet consumption in mice. Pain 2024; 165:470-486. [PMID: 37733484 DOI: 10.1097/j.pain.0000000000003030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 07/07/2023] [Indexed: 09/23/2023]
Abstract
ABSTRACT Lipid-rich diet is the major cause of obesity, affecting 13% of the worldwide adult population. Obesity is a major risk factor for metabolic syndrome that includes hyperlipidemia and diabetes mellitus. The early phases of metabolic syndrome are often associated with hyperexcitability of peripheral small diameter sensory fibers and painful diabetic neuropathy. Here, we investigated the effect of high-fat diet-induced obesity on the activity of dorsal root ganglion (DRG) sensory neurons and pain perception. We deciphered the underlying cellular mechanisms involving the acid-sensing ion channel 3 (ASIC3). We show that mice made obese through consuming high-fat diet developed the metabolic syndrome and prediabetes that was associated with heat pain hypersensitivity, whereas mechanical sensitivity was not affected. Concurrently, the slow conducting C fibers in the skin of obese mice showed increased activity on heating, whereas their mechanosensitivity was not altered. Although ASIC3 knockout mice fed with high-fat diet became obese, and showed signs of metabolic syndrome and prediabetes, genetic deletion, and in vivo pharmacological inhibition of ASIC3, protected mice from obesity-induced thermal hypersensitivity. We then deciphered the mechanisms involved in the heat hypersensitivity of mice and found that serum from high-fat diet-fed mice was enriched in lysophosphatidylcholine (LPC16:0, LPC18:0, and LPC18:1). These enriched lipid species directly increased the activity of DRG neurons through activating the lipid sensitive ASIC3 channel. Our results identify ASIC3 channel in DRG neurons and circulating lipid species as a mechanism contributing to the hyperexcitability of nociceptive neurons that can cause pain associated with lipid-rich diet consumption and obesity.
Collapse
Affiliation(s)
- Ahmed Negm
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Katharina Stobbe
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Selma Ben Fradj
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Clara Sanchez
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Arnaud Landra-Willm
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Margaux Richter
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | | | | | - Emmanuel Deval
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Eric Lingueglia
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| | - Carole Rovere
- Université Côte d'Azur, CNRS, IPMC, LabEx SIGNALIFE, Valbonne, France
| | - Jacques Noel
- Université Côte d'Azur, CNRS, IPMC, LabEx ICST, FHU InovPain, Valbonne, France. Negm is now with the Université Clermont-Auvergne, Laboratoire Neurodol, UMR 1107 Inserm, Clermont-Ferrand, France
| |
Collapse
|
44
|
Chhabra KH, Bathina S, Faniyan TS, Samuel DJ, Raza MU, de Souza Cordeiro LM, Viana Di Prisco G, Atwood BK, Robles J, Bainbridge L, Davis A. ADGRL1 is a glucose receptor involved in mediating energy and glucose homeostasis. Diabetologia 2024; 67:170-189. [PMID: 37712955 PMCID: PMC10709246 DOI: 10.1007/s00125-023-06010-6] [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: 05/25/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023]
Abstract
AIMS/HYPOTHESIS The brain is a major consumer of glucose as an energy source and regulates systemic glucose as well as energy balance. Although glucose transporters such as GLUT2 and sodium-glucose cotransporter 2 (SGLT2) are known to regulate glucose homeostasis and metabolism, the identity of a receptor that binds glucose to activate glucose signalling pathways in the brain is unknown. In this study, we aimed to discover a glucose receptor in the mouse hypothalamus. METHODS Here we used a high molecular mass glucose-biotin polymer to enrich glucose-bound mouse hypothalamic neurons through cell-based affinity chromatography. We then subjected the enriched neurons to proteomic analyses and identified adhesion G-protein coupled receptor 1 (ADGRL1) as a top candidate for a glucose receptor. We validated glucose-ADGRL1 interactions using CHO cells stably expressing human ADGRL1 and ligand-receptor binding assays. We generated and determined the phenotype of global Adgrl1-knockout mice and hypothalamus-specific Adgrl1-deficient mice. We measured the variables related to glucose and energy homeostasis in these mice. We also generated an Adgrl1Cre mouse model to investigate the role of ADGRL1 in sensing glucose using electrophysiology. RESULTS Adgrl1 is highly expressed in the ventromedial nucleus of the hypothalamus (VMH) in mice. Lack of Adgrl1 in the VMH in mice caused fasting hyperinsulinaemia, enhanced glucose-stimulated insulin secretion and insulin resistance. In addition, the Adgrl1-deficient mice had impaired feeding responses to glucose and fasting coupled with abnormal glucose sensing and decreased physical activity before development of obesity and hyperglycaemia. In female mice, ovariectomy was necessary to reveal the contribution of ADGRL1 to energy and glucose homeostasis. CONCLUSIONS/INTERPRETATION Altogether, our findings demonstrate that ADGRL1 binds glucose and is involved in energy as well as glucose homeostasis in a sex-dependent manner. Targeting ADGRL1 may introduce a new class of drugs for the treatment of type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Kavaljit H Chhabra
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.
| | - Siresha Bathina
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Tumininu S Faniyan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Dennis J Samuel
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Muhammad Ummear Raza
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Leticia Maria de Souza Cordeiro
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Gonzalo Viana Di Prisco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brady K Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jorge Robles
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Lauren Bainbridge
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Autumn Davis
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
45
|
Ho PY, Koh YC, Lu TJ, Liao PL, Pan MH. Purple Napiergrass ( Pennisetum purpureum Schumach) Hot Water Extracts Ameliorate High-Fat Diet-Induced Obesity and Metabolic Disorders in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20701-20712. [PMID: 38088361 DOI: 10.1021/acs.jafc.3c05678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Purple Pennisetum (Pennisetum purpureum Schumach), a hybrid between Taihucao No. 2 and the local wild species of purple Pennisetum, has dark red stems and leaves due to its anthocyanin content. This study explores the potential of purple napiergrass extracts (PNE) in alleviating obesity and metabolic disorders induced by a high-fat diet in mice, where 50% of the caloric content is derived from fat. Mice were orally administered low-dose or high-dose PNE alongside a high-fat diet. Experimental findings indicate that PNE attenuated weight gain, reduced liver, and adipose tissue weight, and lowered blood cholesterol, triglyceride, low-density lipoprotein, and blood sugar levels. Stained sections showed that PNE inhibited lipid accumulation and fat hypertrophy in the liver. Immunoblotting analysis suggested that PNE improved the inflammatory response associated with obesity, dyslipidemia, and hyperglycemia induced by a high-fat diet. Furthermore, PNE potentially functions as a PPAR-γ agonist, increasing the adiponectin (ADIPOQ) concentration and suppressing inflammatory factors, while elevating the anti-inflammatory factor interleukin-10 (IL-10) in the liver. PNE-treated mice showed enhanced activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and AMP-activated protein kinase (AMPK) pathways and increased fatty acid oxidation and liver lipolysis. In conclusion, this study elucidated the mechanisms underlying the anti-inflammatory, PI3K/Akt, and AMPK pathways in a high-fat diet-induced obesity model. These findings highlight the potential of PNE in reducing weight, inhibiting inflammation, and improving blood sugar and lipid levels, showing the potential for addressing obesity-related metabolic disorders in humans.
Collapse
Affiliation(s)
- Pin-Yu Ho
- Institute of Food Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan ROC
| | - Yen-Chun Koh
- Institute of Food Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan ROC
| | - Ting-Jang Lu
- Institute of Food Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan ROC
| | - Po-Lin Liao
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University-Yangming Campus, 155, Sec.2, Linong Street, Taipei 11221 Taiwan ROC
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan ROC
- Department of Public Health, China Medical University, 91, Hsueh-Shih Road, Taichung 40402, Taiwan ROC
- Department of Food Nutrition and Health Biotechnology, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan, ROC
| |
Collapse
|
46
|
Hao L, Chen CY, Nie YH, Kaliannan K, Kang JX. Differential Interventional Effects of Omega-6 and Omega-3 Polyunsaturated Fatty Acids on High Fat Diet-Induced Obesity and Hepatic Pathology. Int J Mol Sci 2023; 24:17261. [PMID: 38139090 PMCID: PMC10743920 DOI: 10.3390/ijms242417261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Current Dietary Guidelines for Americans recommend replacing saturated fat (SFA) intake with polyunsaturated fatty acids (PUFAs) and monosaturated fatty acids (MUFAs) but do not specify the type of PUFAs, which consist of two functionally distinct classes: omega-6 (n-6) and omega-3 (n-3) PUFAs. Given that modern Western diets are already rich in n-6 PUFAs and the risk of chronic disease remains high today, we hypothesized that increased intake of n-3 PUFAs, rather than n-6 PUFAs, would be a beneficial intervention against obesity and related liver diseases caused by high-fat diets. To test this hypothesis, we fed C57BL/6J mice with a high-fat diet (HF) for 10 weeks to induce obesity, then divided the obese mice into three groups and continued feeding for another 10 weeks with one of the following three diets: HF, HF+n-6 (substituted half of SFA with n-6 PUFAs), and HF+n-3 (substituted half of SFA with n-3 PUFAs), followed by assessment of body weight, fat mass, insulin sensitivity, hepatic pathology, and lipogenesis. Interestingly, we found that the HF+n-6 group, like the HF group, had a continuous increase in body weight and fat mass, while the HF+n-3 group had a significant decrease in body weight and fat mass, although all groups had the same calorie intake. Accordingly, insulin resistance and fatty liver pathology (steatosis and fat levels) were evident in the HF+n-6 and HF groups but barely seen in the HF+n-3 group. Furthermore, the expression of lipogenesis-related genes in the liver was upregulated in the HF+n-6 group but downregulated in the HF+n-3 group. Our findings demonstrate that n-6 PUFAs and n-3 PUFAs have differential effects on obesity and fatty liver disease and highlight the importance of increasing n-3 PUFAs and reducing n-6 PUFAs (balancing the n-6/n-3 ratio) in clinical interventions and dietary guidelines for the management of obesity and related diseases.
Collapse
Affiliation(s)
- Lei Hao
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (L.H.); (C.-Y.C.)
- Department of Nursing and Allied Health Professions, Indiana University of Pennsylvania, Indiana, PA 15705, USA
| | - Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (L.H.); (C.-Y.C.)
- Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Yong-Hui Nie
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (L.H.); (C.-Y.C.)
| | - Kanakaraju Kaliannan
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (L.H.); (C.-Y.C.)
| | - Jing X. Kang
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (L.H.); (C.-Y.C.)
- Omega-3 and Global Health Institute, Boston, MA 02129, USA
| |
Collapse
|
47
|
Roy RV, Means N, Rao G, Asfa S, Madka V, Dey A, Zhang Y, Choudhury M, Fung KM, Dhanasekaran DN, Friedman JE, Crawford HC, Rao CV, Bhattacharya R, Mukherjee P. Pancreatic Ubap2 deletion regulates glucose tolerance, inflammation, and protection from cerulein-induced pancreatitis. Cancer Lett 2023; 578:216455. [PMID: 37865160 PMCID: PMC10897936 DOI: 10.1016/j.canlet.2023.216455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
Ubiquitin-binding associated protein 2 (UBAP2) is reported to promote macropinocytosis and pancreatic adenocarcinoma (PDAC) growth, however, its role in normal pancreatic function remains unknown. We addressed this knowledge gap by generating UBAP2 knockout (U2KO) mice under a pancreas-specific Cre recombinase (Pdx1-Cre). Pancreatic architecture remained intact in U2KO animals, but they demonstrated slight glucose intolerance compared to controls. Upon cerulein challenge to induce pancreatitis, U2KO animals had reduced levels of several pancreatitis-relevant cytokines, amylase and lipase in the serum, reduced tissue damage, and lessened neutrophil infiltration into the pancreatic tissue. Mechanistically, cerulein-challenged U2KO animals revealed reduced NF-κB activation compared to controls. In vitro promoter binding studies confirmed the reduction of NF-κB binding to its target molecules supporting UBAP2 as a new regulator of inflammation in pancreatitis and may be exploited as a therapeutic target in future to inhibit pancreatitis.
Collapse
Affiliation(s)
- Ram Vinod Roy
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Nicolas Means
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sima Asfa
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anindya Dey
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yushan Zhang
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Monalisa Choudhury
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Danny N Dhanasekaran
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Howard C Crawford
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health System, Detroit, MI, USA
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Resham Bhattacharya
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
48
|
Ozcaliskan Ilkay H, Karabulut D, Kamaci Ozocak G, Mehmetbeyoglu E, Kaymak E, Kisioglu B, Cicek B, Akyol A. Quinoa ( Chenopodium quinoa Willd.) supplemented cafeteria diet ameliorates glucose intolerance in rats. Food Sci Nutr 2023; 11:6920-6930. [PMID: 37970433 PMCID: PMC10630841 DOI: 10.1002/fsn3.3603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 11/17/2023] Open
Abstract
Quinoa (Chenopodium quinoa Willd.) is a pseudocereal with rich nutritional composition, gluten free, and organoleptic. The primary aim of this study was to elucidate the possible protective roles of quinoa in glucose homeostasis in a model of cafeteria diet-induced obesity. Male Wistar rats (3 weeks of age) were randomly allocated to be fed by; control chow (CON; n = 6), quinoa (QUI; n = 6), cafeteria (CAF; n = 6), or quinoa and cafeteria (CAFQ; n = 6) for 15 weeks. CAFQ resulted in decreased saturated fat, sugar, and sodium intake in comparison with CAF. Compared to CON, CAF increased body weight gain, plasma insulin, plasma glucose, decreased liver IRS-1, AMPK mRNA expressions, and pancreatic β-cell insulin immunoreactivity, and developed hepatocyte degeneration and microvesicular steatosis. Compared to CAF, QUI lowered body weight, plasma glucose, and plasma insulin, increased liver IRS-1 and AMPK mRNA expressions, and pancreatic β-cell insulin immunoreactivity. Compared to CAF, CAFQ lowered plasma glucose, increased liver IRS-1 mRNA expressions, increased pancreatic β-cell insulin immunoreactivity, and lowered hepatocyte degeneration and microvesicular steatosis. Dietary treatments did not influence IRS-2, AKT2, and INSR mRNA expressions. HOMA-IR, HOMA-β, and QUICKI were also similar between groups. Restoration of insulin in CAFQ islets was as well as that of CON and QUI groups. In conclusion, as a functional food, quinoa may be useful in the prevention of obesity and associated metabolic outcomes such as glucose intolerance, disrupted pancreatic β-cell function, hepatic insulin resistance, and lipid accumulation.
Collapse
Affiliation(s)
- Hatice Ozcaliskan Ilkay
- Faculty of Health Sciences, Department of Nutrition and DieteticsHacettepe UniversityAnkaraTurkey
- Faculty of Health Sciences, Department of Nutrition and DieteticsErciyes UniversityKayseriTurkey
| | - Derya Karabulut
- Faculty of Medicine, Department of Histology and EmbryologyErciyes UniversityKayseriTurkey
| | - Gonca Kamaci Ozocak
- Faculty of Veterinary Medicine, Department of Laboratory Animals ScienceErciyes UniversityKayseriTurkey
| | | | - Emin Kaymak
- Faculty of Medicine, Department of Histology and EmbryologyYozgat Bozok UniversityYozgatTurkey
| | - Betul Kisioglu
- Faculty of Health Sciences, Department of Nutrition and DieteticsHacettepe UniversityAnkaraTurkey
| | - Betul Cicek
- Faculty of Health Sciences, Department of Nutrition and DieteticsErciyes UniversityKayseriTurkey
| | - Asli Akyol
- Faculty of Health Sciences, Department of Nutrition and DieteticsHacettepe UniversityAnkaraTurkey
| |
Collapse
|
49
|
Rubio WB, Cortopassi MD, Ramachandran D, Walker SJ, Balough EM, Wang J, Banks AS. Not so fast: Paradoxically increased variability in the glucose tolerance test due to food withdrawal in continuous glucose-monitored mice. Mol Metab 2023; 77:101795. [PMID: 37640144 PMCID: PMC10493264 DOI: 10.1016/j.molmet.2023.101795] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVE This study was performed to determine the effect of fasting on reproducibility of the glucose tolerance test. Due to individual variation in animal feeding behaviors, fasting animals prior to metabolic and behavioral experiments is widely held to reduce inter-subject variation in glucose and metabolic parameters of preclinical rodent models. Reducing variability is especially important for studies where initial metabolite levels can influence the magnitude of experimental interventions, but fasting also imposes stress that may distort the variables of interest. One such intervention is the glucose tolerance test (GTT) which measures the maximum response and recovery following a bolus of exogenous glucose. We sought to investigate how fasting affects the response of individual mice to a GTT. METHODS Using simultaneous continuous glucose monitoring (CGM) and indirect calorimetry, we quantified blood glucose, physical activity, body temperature, metabolic rates, and food consumption levels on a minute-to-minute basis in adult male mice for 4 weeks. We tested the effects of a 4-h or 18-h fast on the GTT to examine the effect of food withdrawal in light or dark photoperiods. Studies were also performed with 4-h fasting in additional mice without implanted CGM probes. RESULTS Contrary to our expectations, a 4-h fast during the light photoperiod promotes a paradoxical increase in inter-animal variation in metabolic rate, physical activity, body temperature, glycemia, and glucose tolerance. This hyperglycemic and hyper-metabolic phenotype promotes increased corticosterone levels and is consistent with a behavioral stress response to food deprivation, even in well-fed mice. We find that mice undergoing an 18-h fast entered torpor, a hibernation-like state. In addition to low body temperature and metabolic rate, torpor is also associated with glucose levels 56 mg/dl lower than those seen in mice with ad libitum access to food. Moreover, the time spent in torpor affects the response to a GTT. CONCLUSION Our results suggest fasting mice before glucose tolerance testing, and perhaps other experiments, can have the opposite of the intended effect where fasting can increase, rather than decrease, experimental variability.
Collapse
Affiliation(s)
- William B Rubio
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Marissa D Cortopassi
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Deepti Ramachandran
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Samuel J Walker
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Elizabeth M Balough
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jiefu Wang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Alexander S Banks
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
50
|
Kozlova EV, Bishay AE, Denys ME, Chinthirla BD, Valdez MC, Spurgin KA, Krum JM, Basappa KR, Currás-Collazo MC. Gene deletion of the PACAP/VIP receptor, VPAC2R, alters glycemic responses during metabolic and psychogenic stress in adult female mice. J Neuroendocrinol 2023; 35:e13354. [PMID: 37946684 DOI: 10.1111/jne.13354] [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: 05/03/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 11/12/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and the homologous peptide, vasoactive intestinal peptide (VIP), participate in glucose homeostasis using insulinotropic and counterregulatory processes. The role of VIP receptor 2 (VPAC2R) in these opposing actions needs further characterization. In this study, we examined the participation of VPAC2R on basal glycemia, fasted levels of glucoregulatory hormones and on glycemia responses during metabolic and psychogenic stress using gene-deleted (Vipr2-/- ) female mice. The mean basal glycemia was significantly greater in Vipr2-/- in the fed state and after an 8-h overnight fast as compared to wild-type (WT) mice. Insulin tolerance testing following a 5-h fast (morning fast, 0.38 U/kg insulin) indicated no effect of genotype. However, during a more intense metabolic challenge (8 h, ON fast, 0.25 U/kg insulin), Vipr2-/- females displayed significantly impaired insulin hypoglycemia. During immobilization stress, the hyperglycemic response and plasma epinephrine levels were significantly elevated above basal in Vipr2-/- , but not WT mice, in spite of similar stress levels of plasma corticosterone. Together, these results implicate participation of VPAC2R in upregulated counterregulatory processes influenced by enhanced sympathoexcitation. Moreover, the suppression of plasma GLP-1 levels in Vipr2-/- mice may have removed the inhibition on hepatic glucose production and the promotion of glucose disposal by GLP-1. qPCR analysis indicated deregulation of central gene markers of PACAP/VIP signaling in Vipr2-/- , upregulated medulla tyrosine hydroxylase (Th) and downregulated hypothalamic Vip transcripts. These results demonstrate a physiological role for VPAC2R in glucose metabolism, especially during insulin challenge and psychogenic stress, likely involving the participation of sympathoadrenal activity and/or metabolic hormones.
Collapse
Affiliation(s)
- Elena V Kozlova
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
- Neuroscience Graduate Program, University of California, Riverside, California, USA
| | - Anthony E Bishay
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Maximilian E Denys
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Bhuvaneswari D Chinthirla
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Matthew C Valdez
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
- Neuroscience Graduate Program, University of California, Riverside, California, USA
| | - Kurt A Spurgin
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
- Neuroscience Graduate Program, University of California, Riverside, California, USA
| | - Julia M Krum
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Karthik R Basappa
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | | |
Collapse
|